Cargando…

MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene

Thermogenesis by brown and beige adipocytes is a potential avenue to increased energy expenditure and thus management of obesity and metabolic syndrome. In mice, Pm20d1 has been previously identified as a candidate thermogenic gene, with its mRNA levels cold-induced and enriched in brown and beige v...

Descripción completa

Detalles Bibliográficos
Autores principales: Benson, Kiara, Hu, Wenxiang, Weller, Angela, Bennett, Alexis, Chen, Eric, Kheterpal, Sumeet, Yoshino, Satoshi, Wang, Lin, Rabinowitz, Joshua, Voight, Benjamin, Lazar, Mitchell, Soccio, Raymond
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Endocrine Society 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6550764/
http://dx.doi.org/10.1210/js.2019-MON-LB017
_version_ 1783424256625868800
author Benson, Kiara
Hu, Wenxiang
Weller, Angela
Bennett, Alexis
Chen, Eric
Kheterpal, Sumeet
Yoshino, Satoshi
Wang, Lin
Rabinowitz, Joshua
Voight, Benjamin
Lazar, Mitchell
Soccio, Raymond
author_facet Benson, Kiara
Hu, Wenxiang
Weller, Angela
Bennett, Alexis
Chen, Eric
Kheterpal, Sumeet
Yoshino, Satoshi
Wang, Lin
Rabinowitz, Joshua
Voight, Benjamin
Lazar, Mitchell
Soccio, Raymond
author_sort Benson, Kiara
collection PubMed
description Thermogenesis by brown and beige adipocytes is a potential avenue to increased energy expenditure and thus management of obesity and metabolic syndrome. In mice, Pm20d1 has been previously identified as a candidate thermogenic gene, with its mRNA levels cold-induced and enriched in brown and beige versus white adipocytes, and with the potential mechanism of generating a novel uncoupling metabolite. Thiazolidinedione (TZD) antidiabetic drugs, which activate the PPARγ nuclear receptor, are potent stimuli for adipocyte browning, yet they fail to induce Pm20d1 expression mouse adipocytes or adipose tissue. In contrast, we show here that PM20D1 is one of the most strongly TZD-induced transcripts in human adipocytes, though not in samples from all individuals. Two putative PPARγ binding sites were identified near the gene’s transcription start site (TSS) in human but not mouse adipocytes. The ~4kb upstream binding site falls in a segmental duplication of a nearly identical intronic region ~2.5kb downstream of the TSS, and this duplication event occurred in the primate lineage and is absent in other mammals like mice. We demonstrate by chromatin immunoprecipitation and reporter assays that PPARγ binding and gene activation occur via this upstream duplicated site, thus explaining the species difference. Furthermore, this functional upstream PPARγ site has genetic variation in the human population, with one single nucleotide polymorphism (SNP) allele disrupting a PPAR response element. We show that this allele gives less activation by PPARγ and TZDs in reporter assays and reduced TZD activation of PM20D1 in patient-derived cultured adipocytes. In addition to this upstream variant that determines PPARγ regulation of PM20D1 in adipocytes, genotype at a distinct and unlinked variant >40kb downstream of the TSS strongly correlates with overall levels of PM20D1 expression in human fat as well as multiple other tissues. Seven tightly linked downstream SNP alleles correlate with very low PMD201 expression and correspondingly high DNA methylation at the TSS. These low PM20D1 expression variants may have phenotypic consequences, accounting for human genetic associations in this region with metabolic and neurodegenerative diseases. Overall, human PM20D1 expression is genetically variable at two levels, with genotype at downstream SNPs correlating with overall PM20D1 expression across tissues (an “on/off switch”), while the upstream PPARγ site SNP determines PPARγ and TZD regulation in adipose tissue (a “rheostat”). This human genetic variation in PM20D1 expression may ultimately inform precision medicine approaches. Unless otherwise noted, all abstracts presented at ENDO are embargoed until the date and time of presentation. For oral presentations, the abstracts are embargoed until the session begins. Abstracts presented at a news conference are embargoed until the date and time of the news conference. The Endocrine Society reserves the right to lift the embargo on specific abstracts that are selected for promotion prior to or during ENDO.
format Online
Article
Text
id pubmed-6550764
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Endocrine Society
record_format MEDLINE/PubMed
spelling pubmed-65507642019-06-13 MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene Benson, Kiara Hu, Wenxiang Weller, Angela Bennett, Alexis Chen, Eric Kheterpal, Sumeet Yoshino, Satoshi Wang, Lin Rabinowitz, Joshua Voight, Benjamin Lazar, Mitchell Soccio, Raymond J Endocr Soc Adipose Tissue, Appetite, and Obesity Thermogenesis by brown and beige adipocytes is a potential avenue to increased energy expenditure and thus management of obesity and metabolic syndrome. In mice, Pm20d1 has been previously identified as a candidate thermogenic gene, with its mRNA levels cold-induced and enriched in brown and beige versus white adipocytes, and with the potential mechanism of generating a novel uncoupling metabolite. Thiazolidinedione (TZD) antidiabetic drugs, which activate the PPARγ nuclear receptor, are potent stimuli for adipocyte browning, yet they fail to induce Pm20d1 expression mouse adipocytes or adipose tissue. In contrast, we show here that PM20D1 is one of the most strongly TZD-induced transcripts in human adipocytes, though not in samples from all individuals. Two putative PPARγ binding sites were identified near the gene’s transcription start site (TSS) in human but not mouse adipocytes. The ~4kb upstream binding site falls in a segmental duplication of a nearly identical intronic region ~2.5kb downstream of the TSS, and this duplication event occurred in the primate lineage and is absent in other mammals like mice. We demonstrate by chromatin immunoprecipitation and reporter assays that PPARγ binding and gene activation occur via this upstream duplicated site, thus explaining the species difference. Furthermore, this functional upstream PPARγ site has genetic variation in the human population, with one single nucleotide polymorphism (SNP) allele disrupting a PPAR response element. We show that this allele gives less activation by PPARγ and TZDs in reporter assays and reduced TZD activation of PM20D1 in patient-derived cultured adipocytes. In addition to this upstream variant that determines PPARγ regulation of PM20D1 in adipocytes, genotype at a distinct and unlinked variant >40kb downstream of the TSS strongly correlates with overall levels of PM20D1 expression in human fat as well as multiple other tissues. Seven tightly linked downstream SNP alleles correlate with very low PMD201 expression and correspondingly high DNA methylation at the TSS. These low PM20D1 expression variants may have phenotypic consequences, accounting for human genetic associations in this region with metabolic and neurodegenerative diseases. Overall, human PM20D1 expression is genetically variable at two levels, with genotype at downstream SNPs correlating with overall PM20D1 expression across tissues (an “on/off switch”), while the upstream PPARγ site SNP determines PPARγ and TZD regulation in adipose tissue (a “rheostat”). This human genetic variation in PM20D1 expression may ultimately inform precision medicine approaches. Unless otherwise noted, all abstracts presented at ENDO are embargoed until the date and time of presentation. For oral presentations, the abstracts are embargoed until the session begins. Abstracts presented at a news conference are embargoed until the date and time of the news conference. The Endocrine Society reserves the right to lift the embargo on specific abstracts that are selected for promotion prior to or during ENDO. Endocrine Society 2019-04-30 /pmc/articles/PMC6550764/ http://dx.doi.org/10.1210/js.2019-MON-LB017 Text en Copyright © 2019 Endocrine Society https://creativecommons.org/licenses/by-nc-nd/4.0/ This article has been published under the terms of the Creative Commons Attribution Non-Commercial, No-Derivatives License (CC BY-NC-ND; https://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Adipose Tissue, Appetite, and Obesity
Benson, Kiara
Hu, Wenxiang
Weller, Angela
Bennett, Alexis
Chen, Eric
Kheterpal, Sumeet
Yoshino, Satoshi
Wang, Lin
Rabinowitz, Joshua
Voight, Benjamin
Lazar, Mitchell
Soccio, Raymond
MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene
title MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene
title_full MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene
title_fullStr MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene
title_full_unstemmed MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene
title_short MON-LB017 Natural Genetic Variation in Humans Determines Basal and PPAR-Inducible Expression of PM20D1, a Putative Thermogenic Gene
title_sort mon-lb017 natural genetic variation in humans determines basal and ppar-inducible expression of pm20d1, a putative thermogenic gene
topic Adipose Tissue, Appetite, and Obesity
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6550764/
http://dx.doi.org/10.1210/js.2019-MON-LB017
work_keys_str_mv AT bensonkiara monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT huwenxiang monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT wellerangela monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT bennettalexis monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT cheneric monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT kheterpalsumeet monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT yoshinosatoshi monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT wanglin monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT rabinowitzjoshua monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT voightbenjamin monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT lazarmitchell monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene
AT soccioraymond monlb017naturalgeneticvariationinhumansdeterminesbasalandpparinducibleexpressionofpm20d1aputativethermogenicgene