Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Mol Sci ; 22(7)2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33804860

ABSTRACT

The transcriptional regulator peroxisome proliferator activated receptor gamma coactivator 1A (PGC-1α), encoded by PPARGC1A, has been linked to neurodegenerative diseases. Recently discovered CNS-specific PPARGC1A transcripts are initiated far upstream of the reference promoter, spliced to exon 2 of the reference gene, and are more abundant than reference gene transcripts in post-mortem human brain samples. The proteins translated from the CNS and reference transcripts differ only at their N-terminal regions. To dissect functional differences between CNS-specific isoforms and reference proteins, we used clustered regularly interspaced short palindromic repeats transcriptional activation (CRISPRa) for selective endogenous activation of the CNS or the reference promoters in SH-SY5Y cells. Expression and/or exon usage of the targets was ascertained by RNA sequencing. Compared to controls, more differentially expressed genes were observed after activation of the CNS than the reference gene promoter, while the magnitude of alternative exon usage was comparable between activation of the two promoters. Promoter-selective associations were observed with canonical signaling pathways, mitochondrial and nervous system functions and neurological diseases. The distinct N-terminal as well as the shared downstream regions of PGC-1α isoforms affect the exon usage of numerous genes. Furthermore, associations of risk genes of amyotrophic lateral sclerosis and Parkinson's disease were noted with differentially expressed genes resulting from the activation of the CNS and reference gene promoter, respectively. Thus, CNS-specific isoforms markedly amplify the biological functions of PPARGC1A and CNS-specific isoforms and reference proteins have common, complementary and selective functions relevant for neurodegenerative diseases.


Subject(s)
Gene Regulatory Networks , Neurodegenerative Diseases/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Promoter Regions, Genetic , Transcriptional Activation , Cell Line, Tumor , Exons , HEK293 Cells , Humans , Neurons/metabolism , Nucleotide Motifs , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcriptome
2.
Mol Genet Genomic Med ; 8(8): e1317, 2020 08.
Article in English | MEDLINE | ID: mdl-32472747

ABSTRACT

BACKGROUND: The APOE-ε4 allele is an established risk factor for Alzheimer's disease (AD). TOMM40 located adjacent to APOE has also been implicated in AD but reports of TOMM40 associations with AD that are independent of APOE-ε4 are at variance. METHODS: We investigated associations of AD with haplotypes defined by three TOMM40 and two APOE single nucleotide polymorphisms in 73 and 71 autopsy cases with intermediate and high likelihood of AD (defined by BRAAK stages 0.02. The two haplotypes encoding APOE-E4 showed strong associations with AD that did not differ between intermediate and high likelihood AD. In contrast, a TOMM40 haplotype encoding APOE-E3 was identified as risk haplotype of high- (p = .0186), but not intermediate likelihood AD (p = .7530). Furthermore, the variant allele of rs2075650 located in intron 2 of TOMM40, increased the risk of high-, but not intermediate likelihood AD on the APOE-ε3/ε3 background (p = .0230). CONCLUSION: The striking association of TOMM40 only with high likelihood AD may explain some contrasting results for TOMM40 in clinical studies and may reflect an association with more advanced disease and/or suggest a role of TOMM40 in the pathogenesis of neurofibrillary tangles.


Subject(s)
Alzheimer Disease/genetics , Apolipoproteins E/genetics , Membrane Transport Proteins/genetics , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Female , Gene Frequency , Haplotypes , Humans , Male , Mitochondrial Precursor Protein Import Complex Proteins
3.
Mol Neurobiol ; 57(2): 752-764, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31471878

ABSTRACT

PPARGC1A encodes a transcriptional co-activator also termed peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1-alpha (PGC-1α) which orchestrates multiple transcriptional programs. We have recently identified CNS-specific transcripts that are initiated far upstream of the reference gene (RG) promoter. The regulation of these isoforms may be relevant, as experimental and genetic studies implicated the PPARGC1A locus in neurodegenerative diseases. We therefore studied cis- and trans-regulatory elements activating the CNS promoter in comparison to the RG promoter in human neuronal cell lines. A naturally occurring variable guanidine thymidine (GT) repeat polymorphism within a microsatellite region in the proximal CNS promoter increases promoter activity in neuronal cell lines. Both the RG and the CNS promoters are activated by ESRRA, and the PGC-1α isoforms co-activate ESRRA on their own promoters suggesting an autoregulatory feedback loop. The proximal CNS, but not the RG, promoter is induced by FOXA2 and co-activated by PGC-1α resulting in robust activation. Furthermore, the CNS, but not the RG, promoter is targeted by the canonical hypoxia response involving HIF1A. Importantly, the transactivation by HIF1A is modulated by the size of the GT polymorphism. Increased expression of CNS-specific transcripts in response to hypoxia was observed in an established rat model, while RG transcripts encoding the full-length reference protein were not increased. These results suggest a role of the CNS region of the PPARGC1A locus in ischemia and warrant further studies in humans as the activity of the CNS promoter as well as its induction by hypoxia is subject to inter-individual variability due to the GT polymorphism.


Subject(s)
Central Nervous System/metabolism , Gene Expression Regulation , Hypoxia/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Polymorphism, Genetic , Repetitive Sequences, Nucleic Acid/genetics , Animals , Base Sequence , Brain Ischemia/genetics , Brain Ischemia/pathology , Cell Line , Ciclopirox/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Organ Specificity/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar
4.
Neurobiol Dis ; 121: 34-46, 2019 01.
Article in English | MEDLINE | ID: mdl-30236862

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. PGC-1α, encoded by PPARGC1A, is a transcriptional co-activator that has been implicated in the pathogenesis of neurodegenerative disorders. We recently discovered multiple new PPARGC1A transcripts that initiate from a novel promoter located far upstream of the reference gene promoter, are CNS-specific and are more abundant than reference gene transcripts in whole brain. These CNS-specific transcripts encode two main full-length and several truncated isoforms via alternative splicing. Truncated CNS-isoforms include 17 kDa proteins that lack the second LXXLL motif serving as an interaction site for several nuclear receptors. We now determined expression levels of CNS- and reference gene transcripts in 5 brain regions of 21, 8, and 13 deceased subjects with idiopathic PD, Lewy body dementia and controls without neurodegenerative disorders, respectively. We observed reductions of CNS-specific transcripts (encoding full-length isoforms) only in the substantia nigra pars compacta of PD and Lewy body dementia. However, in the substantia nigra and globus pallidus of PD cases we found an up-regulation of transcripts encoding the 17 kDa proteins that inhibited the co-activation of several transcription factors by full-length PGC-1α proteins in transfection assays. In two established animal models of PD, the PPARGC1A expression profiles differed from the profile in human PD in that the levels of CNS- and reference gene transcripts were decreased in several brain regions. Furthermore, we identified haplotypes in the CNS-specific region of PPARGC1A that appeared protective for PD in a clinical cohort and a post-mortem sample (P = .0002). Thus, functional and genetic studies support a role of the CNS-specific PPARGC1A locus in PD.


Subject(s)
Brain/metabolism , Parkinson Disease/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Aged , Aged, 80 and over , Animals , Female , Genetic Loci , Humans , Male , Mice, Inbred C57BL , Protein Isoforms/genetics
5.
Cell Physiol Biochem ; 43(6): 2297-2309, 2017.
Article in English | MEDLINE | ID: mdl-29073588

ABSTRACT

BACKGROUND/AIM: Accurate genotyping of CYP2D6 is challenging due to its inherent genetic variation, copy number variation (duplications and deletions) and hybrid formation with highly homologous pseudogenes. Because a relatively high percentage (∼25%) of clinically prescribed drugs are substrates for this enzyme, accurate determination of its genotype for phenotype prediction is essential. METHODS: A cohort of 365 patient samples was genotyped for CYP2D6 using Sanger sequencing (as the gold standard), hydrolysis probe assays or pyrosequencing. RESULTS: A discrepant result between the three genotyping methods for the loss of function CYP2D6*3 (g.2549delA, rs35742686) genetic variant was found in one of the samples. This sample also contained the CYP2D6 g.2470T>C (rs17002852) variation, which had an allele frequency of 2.47% in our cohort. Redesign of the CYP2D6*3 pyrosequencing and hydrolysis probe assays to avoid CYP2D6 g.2470 corrected the anomaly. CONCLUSION: To evidence allele drop out and increase the accuracy of genotyping, intra-patient validation of the same genetic variation with at least two separate methods should be considered.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , DNA Copy Number Variations , Genotyping Techniques/methods , Alleles , Cohort Studies , DNA/chemistry , DNA/isolation & purification , DNA/metabolism , Discriminant Analysis , Gene Frequency , Genotype , Haplotypes , Humans , Phenotype , Sequence Analysis, DNA
6.
Cell Physiol Biochem ; 41(4): 1491-1502, 2017.
Article in English | MEDLINE | ID: mdl-28365704

ABSTRACT

Pendrin is upregulated in bronchial epithelial cells following IL-4 stimulation via binding of STAT6 to an N4 GAS motif. Basal CpG methylation of the pendrin promoter is cell-specific. We studied if a correlation exists between IL-4 sensitivity and the CpG methylation status of the pendrin promoter in human bronchial epithelial cell models. METHODS: Real-time PCR and pyrosequencing were used to respectively quantify pendrin mRNA levels and methylation of pendrin promoter, with and without IL-4 stimulation, in healthy and diseased primary HBE cells, as well as NCI-H292 cells. RESULTS: Increases in pendrin mRNA after IL-4 stimulation was more robust in NCI-H292 cells than in primary cells. The amount of gDNA methylated varied greatly between the cell types. In particular, CpG site 90 located near the N4 GAS motif was highly methylated in the primary cells. An additional CpG site (90bis), created by a SNP, was found only in the primary cells. IL-4 stimulation resulted in dramatic demethylation of CpG sites 90 and 90bis in the primary cells. CONCLUSIONS: IL-4 induces demethylation of specific CpG sites within the pendrin promoter. These epigenetic alterations are cell type specific, and may in part dictate pendrin mRNA transcription.


Subject(s)
Bronchi/metabolism , CpG Islands , DNA Methylation , Epithelial Cells/metabolism , Interleukin-4/metabolism , Membrane Transport Proteins/biosynthesis , Response Elements , Bronchi/cytology , Cell Line , Epigenesis, Genetic , Epithelial Cells/cytology , Female , Humans , Middle Aged , RNA, Messenger/biosynthesis , Sulfate Transporters
7.
J Sci Med Sport ; 20(7): 700-705, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28185807

ABSTRACT

OBJECTIVES: Regular aerobic exercise provides beneficial effects on human health and reduces all-cause mortality. Aerobic exercise has profound metabolic effects, and specific metabolites may reflect physiological changes. We aimed to identify endogenous metabolites that distinguish the trained from the untrained state to increase the spectrum of analytes amenable for hypothesis testing and to expand understanding of putative beneficial pathways. DESIGN: Cross sectional laboratory repeated measures study. METHODS: Exercise testing was performed in 37 healthy male participants and serum samples were obtained before and after completion of a ten-week standardized exercise program. Samples were analyzed for routine clinical parameters and for 188 endogenous metabolites by LC-MS/MS. RESULTS: Indicating the effectiveness of the intervention program, parameters of sport physiology were different after training. After correcting for multiple testing, serum concentrations of several metabolites differed between the trained and untrained state. Serine and glutamate decreased in response to exercise, whereas sarcosine and kynurenine increased. Phosphatidylcholines showed a mixed response in that four species increased and three decreased. However, all seven lysophosphatidylcholines and all four plasmalogens that differed between the trained and untrained state, increased. One short-chain acylcarnitine also decreased. In receiver operator characteristics analyses, sarcosine displayed the highest AUC value (0.839; 95% CI: 0.734-0.926) in discriminating the pre- from the post-trained state. CONCLUSIONS: Our study detected metabolites that clearly differentiate the trained from the untrained state. These metabolites may be targeted in mechanistic studies to understand underlying biochemical pathways and could serve to improve the design, monitoring and individualization of training programs.


Subject(s)
Amino Acids/blood , Biogenic Amines/blood , Carnitine/analogs & derivatives , Exercise/physiology , Phospholipids/blood , Biomarkers/blood , Carnitine/blood , Chromatography, Liquid , Cross-Sectional Studies , Humans , Male , Middle Aged , Tandem Mass Spectrometry
8.
J Diabetes Res ; 2015: 405371, 2015.
Article in English | MEDLINE | ID: mdl-26090471

ABSTRACT

The genomic region ~500 kb upstream of IRS1 has been implicated in insulin resistance, type 2 diabetes, adverse lipid profile, and cardiovascular risk. To gain further insight into this chromosomal region, we typed four SNPs in a cross-sectional cohort and subjects with type 2 diabetes recruited from the same geographic region. From 16 possible haplotypes, 6 haplotypes with frequencies >0.01 were observed. We identified one haplotype that was protective against insulin resistance (determined by HOMA-IR and fasting plasma insulin levels), type 2 diabetes, an adverse lipid profile, increased C-reactive protein, and asymptomatic atherosclerotic disease (assessed by intima media thickness of the common carotid arteries). BMI and total adipose tissue mass as well as visceral and subcutaneous adipose tissue mass did not differ between the reference and protective haplotypes. In 92 subjects, we observed an association of the protective haplotype with higher skeletal muscle mRNA levels of LOC646736, which is located in the same haplotype block as the informative SNPs and is mainly expressed in skeletal muscle, but only at very low levels in liver or adipose tissues. These data suggest a role for LOC646736 in human insulin resistance and warrant further studies on the functional effects of this locus.


Subject(s)
Atherosclerosis/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Insulin Receptor Substrate Proteins/genetics , Insulin Resistance , Muscle, Skeletal/metabolism , 5' Untranslated Regions , Asymptomatic Diseases , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Austria , Case-Control Studies , Cohort Studies , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Disease Resistance , Dyslipidemias/complications , Dyslipidemias/genetics , Dyslipidemias/metabolism , Dyslipidemias/pathology , Female , Genetic Association Studies , Haplotypes , Humans , Insulin Receptor Substrate Proteins/metabolism , Male , Middle Aged , Muscle, Skeletal/pathology , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism
9.
Trends Pharmacol Sci ; 36(6): 406-16, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26005080

ABSTRACT

Over the past few decades, mortality resulting from cardiovascular disease (CVD) steadily decreased in western countries; however, in recent years, the decline has become offset by the increase in obesity. Obesity is strongly associated with the metabolic syndrome and its atherogenic dyslipidemia resulting from insulin resistance. While lifestyle treatment would be effective, drugs targeting individual risk factors are often required. Such treatment may result in polypharmacy. Novel approaches are directed towards the treatment of several risk factors with one drug. Studies in animal models and humans suggest a central role for sterol regulatory-element binding proteins (SREBPs) in the pathophysiology of the metabolic syndrome. Four recent studies targeting the maturation or transcriptional activities of SREBPs provide proof of concept for the efficacy of SREBP inhibition in this syndrome.


Subject(s)
Metabolic Syndrome/metabolism , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors , Animals , Humans , Metabolic Syndrome/drug therapy , Metabolic Syndrome/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use , Triterpenes/pharmacology , Triterpenes/therapeutic use
10.
Cell ; 158(1): 25-40, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995976

ABSTRACT

Obesity and diabetes affect more than half a billion individuals worldwide. Interestingly, the two conditions do not always coincide and the molecular determinants of "healthy" versus "unhealthy" obesity remain ill-defined. Chronic metabolic inflammation (metaflammation) is believed to be pivotal. Here, we tested a hypothesized anti-inflammatory role for heme oxygenase-1 (HO-1) in the development of metabolic disease. Surprisingly, in matched biopsies from "healthy" versus insulin-resistant obese subjects we find HO-1 to be among the strongest positive predictors of metabolic disease in humans. We find that hepatocyte and macrophage conditional HO-1 deletion in mice evokes resistance to diet-induced insulin resistance and inflammation, dramatically reducing secondary disease such as steatosis and liver toxicity. Intriguingly, cellular assays show that HO-1 defines prestimulation thresholds for inflammatory skewing and NF-κB amplification in macrophages and for insulin signaling in hepatocytes. These findings identify HO-1 inhibition as a potential therapeutic strategy for metabolic disease.


Subject(s)
Heme Oxygenase-1/metabolism , Insulin Resistance , Membrane Proteins/metabolism , Obesity/complications , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Hepatocytes/metabolism , Humans , Inflammation/metabolism , Liver/metabolism , Macrophages/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Mice , Mice, Knockout , Obesity/physiopathology , Reactive Oxygen Species/metabolism
11.
BMC Neurol ; 14: 1, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24383721

ABSTRACT

BACKGROUND: Genetic modifiers are important clues for the identification of therapeutic targets in neurodegenerative diseases. Huntington disease (HD) is one of the most common autosomal dominant inherited neurodegenerative diseases. The clinical symptoms include motor abnormalities, cognitive decline and behavioral disturbances. Symptom onset is typically between 40 and 50 years of age, but can vary by several decades in extreme cases and this is in part determined by modifying genetic factors. The metabolic master regulator PGC-1α, coded by the PPARGC1A gene, coordinates cellular respiration and was shown to play a role in neurodegenerative diseases, including HD. METHODS: Using a candidate gene approach we analyzed a large European cohort (n = 1706) from the REGISTRY study for associations between PPARGC1A genotype and age at onset (AO) in HD. RESULTS: We report that a coding variant (rs3736265) in PPARGC1A is associated with an earlier motor AO in men but not women carrying the HD mutation. CONCLUSIONS: These results further strengthen the evidence for a role of PGC-1α in HD and unexpectedly suggest a gender effect.


Subject(s)
Huntington Disease/epidemiology , Huntington Disease/genetics , Open Reading Frames/genetics , Polymorphism, Single Nucleotide/genetics , Transcription Factors/genetics , Age of Onset , Cohort Studies , Europe/epidemiology , Female , Humans , Huntington Disease/diagnosis , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Registries
12.
Hum Mol Genet ; 22(17): 3477-84, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23669350

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating, adult-onset neurodegenerative disorder of the upper and lower motor systems. It leads to paresis, muscle wasting and inevitably to death, typically within 3-5 years. However, disease onset and survival vary considerably ranging in extreme cases from a few months to several decades. The genetic and environmental factors underlying this variability are of great interest as potential therapeutic targets. In ALS, men are affected more often and have an earlier age of onset than women. This gender difference is recapitulated in transgenic rodent models, but no underlying mechanism has been elucidated. Here we report that SNPs in the brain-specific promoter region of the transcriptional co-activator PGC-1α, a master regulator of metabolism, modulate age of onset and survival in two large and independent ALS populations and this occurs in a strictly male-specific manner. In complementary animal studies, we show that deficiency of full-length (FL) Pgc-1α leads to a significantly earlier age of onset and a borderline shortened survival in male, but not in female ALS-transgenic mice. In the animal model, FL Pgc-1α-loss is associated with reduced mRNA levels of the trophic factor Vegf-A in males, but not in females. In summary, we indentify PGC-1α as a novel and clinically relevant disease modifier of human and experimental ALS and report a sex-dependent effect of PGC-1α in this neurodegenerative disorder.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Adult , Age of Onset , Aged , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Polymorphism, Single Nucleotide , Sex Characteristics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Cell Physiol Biochem ; 32(7): 238-48, 2013.
Article in English | MEDLINE | ID: mdl-24429829

ABSTRACT

BACKGROUND: Pendrin, an anion exchanger associated with the inner ear, thyroid and kidney, plays a significant role in respiratory tissues and diseases, where its expression is increased following IL-4 and IL-13 exposure. The mechanism leading to increased pendrin expression is in part due to binding of STAT6 to a consensus sequence (N4 GAS motif) located in the pendrin promoter. As retrospective analyses of the 5' upstream sequence of the human pendrin promoter revealed an additional N4 GAS motif (1660 base pairs upstream of the one previously identified), we set out to define its contribution to IL-4 stimulated changes in pendrin promoter activity. METHODS AND RESULTS: Electrophoretic mobility shift assays showed that STAT6 bound to oligonucleotides corresponding to both N4 GAS motifs in vitro, while dual luciferase promoter assays revealed that only one of the N4 GAS motifs was necessary for IL-4 -stimulated increases in pendrin promoter activity in living cells. We then examined the ability of STAT6 to bind each of the N4 GAS motifs in vivo with a site-specific ChIP assay, the results of which showed that STAT6 interacted with only the N4 GAS motif that was functionally implicated in increasing the activity of the pendrin promoter following IL-4 treatment. CONCLUSIONS: Of the two N4 GAS motifs located in the human pendrin promoter region analyzed in this study (nucleotides -3906 to +7), only the one located nearest to the first coding ATG participates in IL-4 stimulated increases in promoter activity.


Subject(s)
Membrane Transport Proteins/genetics , Nucleotide Motifs/genetics , Promoter Regions, Genetic , STAT6 Transcription Factor/genetics , Binding Sites , Humans , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/chemistry , Protein Binding , STAT6 Transcription Factor/chemistry , Sulfate Transporters
14.
Hum Mol Genet ; 21(15): 3461-73, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22589246

ABSTRACT

PGC-1α has been implicated in the pathogenesis of neurodegenerative disorders. Several single-nucleotide polymorphisms (SNPs) located in two separate haplotype blocks of PPARGC1A have shown associations with Huntington's disease (HD) and Parkinson's disease, but causative SNPs have not been identified. One SNP (rs7665116) was located in a highly conserved 233 bp region of intron 2. To determine whether rs7665116 is located in an alternative exon, we performed 5'-RLM-RACE from exon 3 and discovered multiple new transcripts that initiated from a common novel promoter located 587 kb upstream of exon 2, but did not contain the conserved region harboring rs7665116. Using real-time polymerase chain reaction, RNase protection assays and northern blotting, we show that the majority of these transcripts are brain specific and are at least equally or perhaps more abundant than the reference sequence PPARGC1A transcripts in whole brain. Two main transcripts containing independent methionine start codons encode full-length brain-specific PGC-1α proteins that differ only at their N-termini (NTs) from PGC-1α, encoded by the reference sequence. Additional truncated isoforms containing these NTs that are similar to NT-PGC-1α exist. Other transcripts may encode potential dominant negative forms, as they are predicted to lack the second LXXLL motif that serves as an interaction site for several nuclear receptors. Furthermore, we show that the new promoter is active in neuronal cell lines and describe haplotypes encompassing this region that are associated with HD age of onset. The discovery of such a large PPARGC1A genomic locus and multiple isoforms in brain warrants further functional studies and may provide new tissue-specific targets for treating neurodegenerative diseases.


Subject(s)
Brain/metabolism , Genome, Human , Heat-Shock Proteins/genetics , Huntington Disease/epidemiology , Huntington Disease/genetics , Transcription Factors/genetics , Age of Onset , Exons , Genomics , Heat-Shock Proteins/metabolism , Humans , Introns , Molecular Sequence Data , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/metabolism
15.
Arterioscler Thromb Vasc Biol ; 32(6): 1535-44, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22460558

ABSTRACT

OBJECTIVE: Genetic studies implicated upstream stimulatory factor 1 (USF1) in familial combined hyperlipidemia because the rs2073658 minor allele was associated with reduced risk of familial combined hyperlipidemia and related disorders. The molecular mechanisms whereby rs2073658 influences trait expression have remained elusive. METHODS AND RESULTS: Plasma lipids, rs2073658 genotypes (N=372), and hepatic transcript levels (N=96) of USF1 and genes involved in hepatic lipoprotein production were determined in obese subjects. The rs2073658 minor allele was associated with reduced plasma triglycerides (TGs) (P<0.001), hepatic USF1 (P<0.01), and microsomal TG transfer protein transcript levels (P<0.05). Functional studies in human hepatocellular carcinoma cells showed that rs2073658 is located in a forkhead box A2 (FOXA2) binding site and that major allele constructs displayed higher transcriptional activity than minor allele constructs. Knockdown of FOXA2 reduced the activity of major, but not minor allele constructs. Furthermore, an interaction between hepatic FOXA2 transcript levels and rs2073658 minor allele carrier status on hepatic USF1 transcript levels was observed in vivo (P<0.05). USF1 activated the transcription of FOXA2 and FOXA2 strongly activated the transcription of microsomal TG transfer protein. CONCLUSIONS: A feed-forward loop comprising activation of USF1 transcription by FOXA2 and activation of FOXA2 transcription by USF1, driving microsomal TG transfer protein expression, is modulated by rs2073658. Hence, rs2073658 likely influences hepatic TG secretion.


Subject(s)
Hyperlipidemia, Familial Combined/genetics , Liver/metabolism , Obesity/genetics , Polymorphism, Single Nucleotide , Upstream Stimulatory Factors/genetics , Adult , Aged , Analysis of Variance , Austria , Binding Sites , Carrier Proteins/metabolism , Chi-Square Distribution , Feedback, Physiological , Female , Gene Frequency , Genetic Predisposition to Disease , Hep G2 Cells , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Hyperlipidemia, Familial Combined/blood , Linear Models , Linkage Disequilibrium , Male , Middle Aged , Obesity/blood , Phenotype , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Transcriptional Activation , Transfection , Triglycerides/blood
16.
J Biol Chem ; 286(50): 42923-36, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22009745

ABSTRACT

Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a transcriptional coactivator that contributes to the regulation of numerous transcriptional programs including the hepatic response to fasting. Mechanisms at transcriptional and post-transcriptional levels allow PGC-1α to support distinct biological pathways. Here we describe a novel human liver-specific PGC-1α transcript that results from alternative promoter usage and is induced by FOXO1 as well as glucocorticoids and cAMP-response element-binding protein signaling but is not present in other mammals. Hepatic tissue levels of novel and wild-type transcripts were similar but were only moderately associated (p < 0.003). Novel mRNA levels were associated with a polymorphism located in its promoter region, whereas wild-type transcript levels were not. Furthermore, hepatic PCK1 mRNA levels exhibited stronger associations with the novel than with the wild-type transcript levels. Except for a deletion of 127 amino acids at the N terminus, the protein, termed L-PGC-1α, is identical to PGC-1α. L-PGC-1α was localized in the nucleus and showed coactivation properties that overlap with those of PGC-1α. Collectively, our data support a role of L-PGC-1α in gluconeogenesis, but functional differences predicted from the altered structure suggest that L-PGC-1α may have arisen to adapt PGC-1α to more complex metabolic pathways in humans.


Subject(s)
Heat-Shock Proteins/metabolism , Liver/metabolism , Protein Isoforms/metabolism , Transcription Factors/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Blotting, Northern , Chromatin Immunoprecipitation , Dexamethasone/pharmacology , Female , Gene Expression/drug effects , Gene Expression/genetics , Genotype , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Immunoblotting , In Vitro Techniques , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Isoforms/genetics , RNA, Messenger/genetics , Rats , Transcription Factors/genetics
17.
Atherosclerosis ; 216(2): 374-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21371711

ABSTRACT

OBJECTIVE: HDL modifying effects of cholesteryl ester transfer protein (CETP) and hepatic lipase (LIPC) depend in part on each other. We studied associations of CETP-Taq1B and -514C>T-LIPC polymorphisms with hepatic mRNA levels, and their combined effects on plasma lipids and carotid atherosclerosis. METHODS: We genotyped the CETP-Taq1B and the -514C>T-LIPC polymorphisms in 67 obese women in whom hepatic CETP and LIPC transcript levels were determined as well as in 1549 participants of the Salzburg Atherosclerosis Prevention Program in Subjects at High Individual Risk (SAPHIR). Carotid atherosclerosis was assessed by intima-media thickness and extent of plaques (B-score) of the carotid arteries. RESULTS: In obese women, CETP-Taq1B and -514C>T-LIPC variant alleles were associated with reduced hepatic levels of CETP and LIPC mRNA, respectively. The CETP and LIPC polymorphisms accounted for 12.9 and 14.4% of the variability in respective transcripts. In the SAPHIR population, CETP-Taq1B showed independent effects on LDL diameter, HDL and LDL cholesterol, apolipoproteins AI and B and cholesterol/HDL cholesterol, while -514C>T-LIPC revealed independent effects on HDL cholesterol and apolipoprotein AI. The two polymorphisms displayed interactions at the level of HDL cholesterol. Compared to subjects carrying wild-type alleles at both loci, subjects homozygous for the CETP wild-type allele, but heterozygous for the LIPC polymorphism and subjects heterozygous for the CETP polymorphism, but homozygous for the LIPC wild-type allele showed an increased risk of carotid atherosclerosis (both P<0.05). CONCLUSIONS: CETP and LIPC polymorphisms influence the respective hepatic transcript levels, demonstrate interactions on HDL cholesterol and suggest that imbalances between CETP and LIPC activities may modulate the risk of carotid atherosclerosis.


Subject(s)
Cholesterol Ester Transfer Proteins/genetics , Lipase/genetics , Liver/metabolism , Polymorphism, Genetic , Adult , Aged , Carotid Artery Diseases/genetics , Female , Genotype , Humans , Lipids/chemistry , Lipoproteins, HDL/metabolism , Male , Middle Aged , Obesity/complications , Risk
18.
Transgenic Res ; 20(4): 827-40, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21120693

ABSTRACT

Although constitutive murine transgenic models have provided important insights into ß-catenin signaling in tissue morphogenesis and tumorigenesis, these models are unable to express activated ß-catenin in a temporally controlled manner. Therefore, to enable the induction (and subsequent de-induction) of ß-catenin signaling during a predetermined time-period or developmental stage, we have generated and characterized a TETO-ΔN89ß-catenin responder transgenic mouse. Crossed with the MTB transgenic effector mouse, which targets the expression of the reverse tetracycline transactivator (rtTA) to the mammary epithelium, we demonstrate that the stabilized (and activated) form of ß-catenin (ΔN89ß-catenin) is expressed only in the presence doxycycline-activated rtTA in the mammary epithelial compartment. Furthermore, we show that transgene-derived ΔN89ß-catenin elicits significant mammary epithelial proliferation and precocious alveologenesis in the virgin doxycycline-treated MTB/TETO-ΔN89ß-catenin bitransgenic. Remarkably, deinduction of TETO-ΔN89ß-catenin transgene expression (through doxycycline withdrawal) results in the reversal of these morphological changes. Importantly, continued activation of the TETO-ΔN89ß-catenin transgene results in palpable mammary tumors (within 7-9 months) in the doxycycline-treated virgin MTB/TETO-ΔN89ß-catenin bigenic but not in the same bitransgenic without doxycycline administration. Collectively, these mammary epithelial responses to ΔN89ß-catenin expression agree with previous reports using conventional transgenesis and therefore confirm that ΔN89ß-catenin functions as expected in this doxycycline-responsive bigenic system. In sum, our mammary gland studies demonstrate "proof-of-principle" for using the TETO-ΔN89ß-catenin transgenic responder to activate (and then de-activate) ß-catenin signaling in any tissue of interest in a spatiotemporal specific fashion.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/embryology , Mammary Neoplasms, Animal/genetics , beta Catenin/biosynthesis , Animals , Doxycycline/pharmacology , Epithelial Cells/metabolism , Female , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Morphogenesis , Signal Transduction , beta Catenin/genetics
19.
FASEB J ; 24(11): 4408-19, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20605949

ABSTRACT

Despite support for receptor of activated NF-κB ligand (RANKL) as a mediator of mammary progesterone action, the extent to which this cytokine can functionally contribute to established progesterone-induced mammary morphogenetic responses in the absence of other presumptive effectors is still unclear. To address this uncertainty, we developed an innovative bigenic system for the doxycycline-inducible expression of RANKL in the mammary epithelium of the progesterone receptor knockout (PRKO) mouse. In response to acute doxycycline exposure, RANKL is specifically expressed in the estrogen receptor α (ER) positive/progesterone receptor negative (ER(+)/PR(-)) cell type in the PRKO mammary epithelium, a cell type that is equivalent to the ER(+)/PR(+) cell type in the wild-type (WT) mammary epithelium. Notably, the ER(+)/PR(+) mammary cell normally expresses RANKL in the WT mammary epithelium during pregnancy. In this PRKO bigenic system, acute doxycycline-induced expression of RANKL results in ordered mammary ductal side branching and alveologenesis, morphological changes that normally occur in the parous WT mouse. This mammary epithelial expansion is accompanied by significant RANKL-induced luminal epithelial proliferation, which is driven, in part, by indirect induction of cyclin D1. Collectively, our findings support the conclusion that RANKL represents a critical mediator of mammary PR action and that restricted expression of this effector to the ER(+)/PR(+) mammary cell-type is necessary for a spatially ordered morphogenetic response to progesterone.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , RANK Ligand/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Cell Proliferation , Doxycycline/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Knockout , Mice, Transgenic , Morphogenesis/genetics , Pregnancy , RANK Ligand/genetics
20.
Genesis ; 48(2): 106-13, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20029965

ABSTRACT

Considering the regulatory complexities of progesterone receptor (PR) action throughout the female reproductive axis and mammary gland, we generated a mouse model that enables conditional ablation of PR function in a spatiotemporal specific manner. Exon 2 of the murine PR gene was floxed to generate a conditional PR allele (PR(flox)) in mice. Crossing the PR(flox/flox) mouse with the ZP3-cre transgenic demonstrated that the PR(flox) allele recombines to a PR null allele (PR(d)). Mice homozygous for the recombined null PR allele (PR(d/d)) exhibit uterine, ovarian, and mammary gland defects that phenocopy those of our previously described PR knockout (PRKO) model. Therefore, this conditional mouse model for PR ablation represents an invaluable resource with which to further define in a developmental and/or reproductive stage-specific manner the individual and integrative roles of distinct PR populations resident in multiple progesterone-responsive target sites.


Subject(s)
Mammary Glands, Animal/physiology , Models, Genetic , Ovary/physiology , Progesterone/metabolism , Uterus/physiology , Animals , Crosses, Genetic , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Female , Gene Targeting , Genetic Engineering , Homozygote , Immunohistochemistry , Integrases/genetics , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Progesterone/metabolism , Reproduction/genetics , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...