1
|
He W, Loganathan N, Belsham DD. IGF1 Signaling Regulates Neuropeptide Expression in Hypothalamic Neurons Under Physiological and Pathological Conditions. Endocrinology 2025; 166:bqaf051. [PMID: 40105689 PMCID: PMC11949690 DOI: 10.1210/endocr/bqaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/28/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
Insulin-like growth factor 1 (IGF1) plays a critical role in metabolism and aging, but its role in the brain remains unclear. This study examined whether hypothalamic neurons respond to IGF1 and how its actions are modulated. RT-qPCR and single-cell RNA sequencing indicated that Igf1r mRNA is expressed in neuropeptide Y/Agouti-related peptide (NPY/AgRP) neurons but has higher expression in pro-opiomelanocortin (POMC) neurons. IGF1 binding proteins Igfbp3 and Igfbp5 were significantly expressed, whereby Igfbp5 levels were modulated by fasting, nutrient availability, and circadian rhythms, implying that IGF1 signaling can be controlled by multiple mechanisms. In mouse and human models, IGF1 regulated Agrp, Npy, Pomc, Cartpt, Spx, Gal, and Fam237b expression, producing an overall anorexigenic profile. Hyperinsulinemia induced IGF1 resistance, accompanied by reduced IGF1R protein, as well as Igf1r and Irs2 mRNA expression via over-activation of phosphoinositide 3-kinase/forkhead box O1 (PI3K-FOXO1) signaling. Thus, hypothalamic neurons respond to IGF1 under physiological conditions, and hyperinsulinemia is a novel mechanism that drives cellular IGF1 resistance.
Collapse
Affiliation(s)
- Wenyuan He
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Neruja Loganathan
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
- Department of Medicine, University of Toronto, Toronto, ON, Canada M5S 1A8
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
2
|
Meda C, Benedusi V, Cherubini A, Valenti L, Maggi A, Della Torre S. Hepatic estrogen receptor alpha drives masculinization in post-menopausal women with metabolic dysfunction-associated steatotic liver disease. JHEP Rep 2024; 6:101143. [PMID: 39308985 PMCID: PMC11414671 DOI: 10.1016/j.jhepr.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 09/25/2024] Open
Abstract
Background & Aims The loss of ovarian functions defining menopause leads to profound metabolic changes and heightens the risk of developing metabolic dysfunction-associated steatotic liver disease (MASLD). Although estrogens primarily act on the female liver through estrogen receptor alpha (ERα), the specific contribution of impaired ERα signaling in triggering MASLD after menopause remains unclear. Methods To address this gap in knowledge, we compared the liver transcriptomes of sham-operated (SHAM) and ovariectomized (OVX) control and liver ERα knockout (LERKO) female mice by performing RNA-Seq analysis. Results OVX led to 1426 differentially expressed genes (DEGs) in the liver of control mice compared to 245 DEGs in LERKO mice. Gene ontology analysis revealed a distinct ovariectomy-induced modulation of the liver transcriptome in LERKO compared with controls, indicating that hepatic ERα is functional and necessary for the complete reprogramming of liver metabolism in response to estrogen depletion. Additionally, we observed an ovariectomy-dependent induction of male-biased genes, especially in the liver of control females, pointing to hepatic ERα involvement in the masculinization of the liver after estrogen loss. To investigate the translational relevance of such findings, we assessed liver samples from a cohort of 60 severely obese individuals (51 women; 9 men). Notably, a shift of the liver transcriptome toward a male-like profile was also observed only in obese women with MASLD (n = 43), especially in women ≥51 years old (15/15), suggesting that masculinization of the female liver contributes to MASLD development in obese women. Conclusions These results highlight the role of hepatic ERα in driving masculinization of the liver transcriptome following menopause, pointing to this receptor as a potential pharmacological target for preventing MASLD in post-menopausal women. Impact and implications Despite the increased risk of developing MASLD after menopause, the specific contribution of impaired hepatic estrogen signaling in driving MASLD in females has not been a major research focus, and, thus, has limited the development of tailored strategies that address the specific mechanisms underlying MASLD in post-menopausal women. This study reveals the functional role of hepatic ERα in mediating liver metabolic changes in response to estrogens loss, leading to a shift in the liver transcriptome towards a male-like profile. In women with obesity, this shift is associated with the development of MASLD. These findings underscore the potential of targeting hepatic ERα as a promising approach for developing effective, sex-specific treatments to preserve liver health and prevent or limit the development and progression of MASLD in post-menopausal women.
Collapse
Affiliation(s)
- Clara Meda
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valeria Benedusi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alessandro Cherubini
- Precision Medicine–Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Precision Medicine–Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Adriana Maggi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
3
|
Vishnevsky OV, Bocharnikov AV, Ignatieva EV. Peak Scores Significantly Depend on the Relationships between Contextual Signals in ChIP-Seq Peaks. Int J Mol Sci 2024; 25:1011. [PMID: 38256085 PMCID: PMC10816497 DOI: 10.3390/ijms25021011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Chromatin immunoprecipitation followed by massively parallel DNA sequencing (ChIP-seq) is a central genome-wide method for in vivo analyses of DNA-protein interactions in various cellular conditions. Numerous studies have demonstrated the complex contextual organization of ChIP-seq peak sequences and the presence of binding sites for transcription factors in them. We assessed the dependence of the ChIP-seq peak score on the presence of different contextual signals in the peak sequences by analyzing these sequences from several ChIP-seq experiments using our fully enumerative GPU-based de novo motif discovery method, Argo_CUDA. Analysis revealed sets of significant IUPAC motifs corresponding to the binding sites of the target and partner transcription factors. For these ChIP-seq experiments, multiple regression models were constructed, demonstrating a significant dependence of the peak scores on the presence in the peak sequences of not only highly significant target motifs but also less significant motifs corresponding to the binding sites of the partner transcription factors. A significant correlation was shown between the presence of the target motifs FOXA2 and the partner motifs HNF4G, which found experimental confirmation in the scientific literature, demonstrating the important contribution of the partner transcription factors to the binding of the target transcription factor to DNA and, consequently, their important contribution to the peak score.
Collapse
Affiliation(s)
- Oleg V. Vishnevsky
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia;
- Department of Natural Science, Novosibirsk State University, 630090 Novosibirsk, Russia;
| | - Andrey V. Bocharnikov
- Department of Natural Science, Novosibirsk State University, 630090 Novosibirsk, Russia;
| | - Elena V. Ignatieva
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia;
- Department of Natural Science, Novosibirsk State University, 630090 Novosibirsk, Russia;
| |
Collapse
|
4
|
Fadel L, Dacic M, Fonda V, Sokolsky BA, Quagliarini F, Rogatsky I, Uhlenhaut NH. Modulating glucocorticoid receptor actions in physiology and pathology: Insights from coregulators. Pharmacol Ther 2023; 251:108531. [PMID: 37717739 PMCID: PMC10841922 DOI: 10.1016/j.pharmthera.2023.108531] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Glucocorticoids (GCs) are a class of steroid hormones that regulate key physiological processes such as metabolism, immune function, and stress responses. The effects of GCs are mediated by the glucocorticoid receptor (GR), a ligand-dependent transcription factor that activates or represses the expression of hundreds to thousands of genes in a tissue- and physiological state-specific manner. The activity of GR is modulated by numerous coregulator proteins that interact with GR in response to different stimuli assembling into a multitude of DNA-protein complexes and facilitate the integration of these signals, helping GR to communicate with basal transcriptional machinery and chromatin. Here, we provide a brief overview of the physiological and molecular functions of GR, and discuss the roles of GR coregulators in the immune system, key metabolic tissues and the central nervous system. We also present an analysis of the GR interactome in different cells and tissues, which suggests tissue-specific utilization of GR coregulators, despite widespread functions shared by some of them.
Collapse
Affiliation(s)
- Lina Fadel
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Marija Dacic
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Vlera Fonda
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Baila A Sokolsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Fabiana Quagliarini
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Inez Rogatsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| | - N Henriette Uhlenhaut
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany; Metabolic Programming, TUM School of Life Sciences & ZIEL Institute for Food and Health, Gregor11 Mendel-Str. 2, 85354 Freising, Germany.
| |
Collapse
|
5
|
Kitamoto T, Accili D. Unraveling the mysteries of hepatic insulin signaling: deconvoluting the nuclear targets of insulin. Endocr J 2023; 70:851-866. [PMID: 37245960 DOI: 10.1507/endocrj.ej23-0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/30/2023] Open
Abstract
Over 100 years have passed since insulin was first administered to a diabetic patient. Since then great strides have been made in diabetes research. It has determined where insulin is secreted from, which organs it acts on, how it is transferred into the cell and is delivered to the nucleus, how it orchestrates the expression pattern of the genes, and how it works with each organ to maintain systemic metabolism. Any breakdown in this system leads to diabetes. Thanks to the numerous researchers who have dedicated their lives to cure diabetes, we now know that there are three major organs where insulin acts to maintain glucose/lipid metabolism: the liver, muscles, and fat. The failure of insulin action on these organs, such as insulin resistance, result in hyperglycemia and/or dyslipidemia. The primary trigger of this condition and its association among these tissues still remain to be uncovered. Among the major organs, the liver finely tunes the glucose/lipid metabolism to maintain metabolic flexibility, and plays a crucial role in glucose/lipid abnormality due to insulin resistance. Insulin resistance disrupts this tuning, and selective insulin resistance arises. The glucose metabolism loses its sensitivity to insulin, while the lipid metabolism maintains it. The clarification of its mechanism is warranted to reverse the metabolic abnormalities due to insulin resistance. This review will provide a brief historical review for the progress of the pathophysiology of diabetes since the discovery of insulin, followed by a review of the current research clarifying our understanding of selective insulin resistance.
Collapse
Affiliation(s)
- Takumi Kitamoto
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba 260-8670, Japan
| | - Domenico Accili
- Department of Medicine and Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032 USA
| |
Collapse
|
6
|
Zhang Y, Fang X, Shuang F, Chen G. Dexamethasone potentiates the insulin-induced Srebp-1c expression in primary rat hepatocytes. FOOD SCIENCE AND HUMAN WELLNESS 2023; 12:1519-1525. [DOI: 10.1016/j.fshw.2023.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
7
|
Correia CM, Præstholm SM, Havelund JF, Pedersen FB, Siersbæk MS, Ebbesen MF, Gerhart-Hines Z, Heeren J, Brewer J, Larsen S, Blagoev B, Færgeman NJ, Grøntved L. Acute Deletion of the Glucocorticoid Receptor in Hepatocytes Disrupts Postprandial Lipid Metabolism in Male Mice. Endocrinology 2023; 164:bqad128. [PMID: 37610219 DOI: 10.1210/endocr/bqad128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/09/2023] [Accepted: 08/21/2023] [Indexed: 08/24/2023]
Abstract
Hepatic lipid metabolism is highly dynamic, and disruption of several circadian transcriptional regulators results in hepatic steatosis. This includes genetic disruption of the glucocorticoid receptor (GR) as the liver develops. To address the functional role of GR in the adult liver, we used an acute hepatocyte-specific GR knockout model to study temporal hepatic lipid metabolism governed by GR at several preprandial and postprandial circadian timepoints. Lipidomics analysis revealed significant temporal lipid metabolism, where GR disruption results in impaired regulation of specific triglycerides, nonesterified fatty acids, and sphingolipids. This correlates with increased number and size of lipid droplets and mildly reduced mitochondrial respiration, most noticeably in the postprandial phase. Proteomics and transcriptomics analyses suggest that dysregulated lipid metabolism originates from pronounced induced expression of enzymes involved in fatty acid synthesis, β-oxidation, and sphingolipid metabolism. Integration of GR cistromic data suggests that induced gene expression is a result of regulatory actions secondary to direct GR effects on gene transcription.
Collapse
Affiliation(s)
- Catarina Mendes Correia
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Stine Marie Præstholm
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Jesper Foged Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Felix Boel Pedersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Majken Storm Siersbæk
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Morten Frendø Ebbesen
- DaMBIC, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Zach Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research (CBMR), Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonathan Brewer
- DaMBIC, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Steen Larsen
- Xlab, Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Nils Joakim Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
8
|
Inoue Y, Suzuki Y, Kunishima Y, Washio T, Morishita S, Takeda H. High-fat diet in early life triggers both reversible and persistent epigenetic changes in the medaka fish (Oryzias latipes). BMC Genomics 2023; 24:472. [PMID: 37605229 PMCID: PMC10441761 DOI: 10.1186/s12864-023-09557-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The nutritional status during early life can have enduring effects on an animal's metabolism, although the mechanisms underlying these long-term effects are still unclear. Epigenetic modifications are considered a prime candidate mechanism for encoding early-life nutritional memories during this critical developmental period. However, the extent to which these epigenetic changes occur and persist over time remains uncertain, in part due to challenges associated with directly stimulating the fetus with specific nutrients in viviparous mammalian systems. RESULTS In this study, we used medaka as an oviparous vertebrate model to establish an early-life high-fat diet (HFD) model. Larvae were fed with HFD from the hatching stages (one week after fertilization) for six weeks, followed by normal chow (NC) for eight weeks until the adult stage. We examined the changes in the transcriptomic and epigenetic state of the liver over this period. We found that HFD induces simple liver steatosis, accompanied by drastic changes in the hepatic transcriptome, chromatin accessibility, and histone modifications, especially in metabolic genes. These changes were largely reversed after the long-term NC, demonstrating the high plasticity of the epigenetic state in hepatocytes. However, we found a certain number of genomic loci showing non-reversible epigenetic changes, especially around genes related to cell signaling, liver fibrosis, and hepatocellular carcinoma, implying persistent changes in the cellular state of the liver triggered by early-life HFD feeding. CONCLUSION In summary, our data show that early-life HFD feeding triggers both reversible and persistent epigenetic changes in medaka hepatocytes. Our data provide novel insights into the epigenetic mechanism of nutritional programming and a comprehensive atlas of the long-term epigenetic state in an early-life HFD model of non-mammalian vertebrates.
Collapse
Affiliation(s)
- Yusuke Inoue
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
| | - Yuta Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8561, Japan
| | - Yoshimi Kunishima
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Terumi Washio
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Shinichi Morishita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8561, Japan.
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto, 603-8555, Japan.
| |
Collapse
|
9
|
Sen D, Maniyadath B, Chowdhury S, Kaur A, Khatri S, Chakraborty A, Mehendale N, Nadagouda S, Sandra U, Kamat SS, Kolthur-Seetharam U. Metabolic regulation of CTCF expression and chromatin association dictates starvation response in mice and flies. iScience 2023; 26:107128. [PMID: 37416476 PMCID: PMC10320512 DOI: 10.1016/j.isci.2023.107128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 05/16/2023] [Accepted: 06/10/2023] [Indexed: 07/08/2023] Open
Abstract
Coordinated temporal control of gene expression is essential for physiological homeostasis, especially during metabolic transitions. However, the interplay between chromatin architectural proteins and metabolism in regulating transcription is less understood. Here, we demonstrate a conserved bidirectional interplay between CTCF (CCCTC-binding factor) expression/function and metabolic inputs during feed-fast cycles. Our results indicate that its loci-specific functional diversity is associated with physiological plasticity in mouse hepatocytes. CTCF differential expression and long non-coding RNA-Jpx mediated changes in chromatin occupancy, unraveled its paradoxical yet tuneable functions, which are governed by metabolic inputs. We illustrate the key role of CTCF in controlling temporal cascade of transcriptional response, with effects on hepatic mitochondrial energetics and lipidome. Underscoring the evolutionary conservation of CTCF-dependent metabolic homeostasis, CTCF knockdown in flies abrogated starvation resistance. In summary, we demonstrate the interplay between CTCF and metabolic inputs that highlights the coupled plasticity of physiological responses and chromatin function.
Collapse
Affiliation(s)
- Devashish Sen
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Babukrishna Maniyadath
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Shreyam Chowdhury
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Arshdeep Kaur
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Subhash Khatri
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Arnab Chakraborty
- Department of Biology, Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| | - Neelay Mehendale
- Department of Biology, Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| | - Snigdha Nadagouda
- Tata Institute of Fundamental Research- Hyderabad (TIFR-H), Hyderabad, Telangana 500046, India
| | - U.S. Sandra
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Siddhesh S. Kamat
- Department of Biology, Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
- Tata Institute of Fundamental Research- Hyderabad (TIFR-H), Hyderabad, Telangana 500046, India
| |
Collapse
|
10
|
Caratti G, Desgeorges T, Juban G, Stifel U, Fessard A, Koenen M, Caratti B, Théret M, Skurk C, Chazaud B, Tuckermann JP, Mounier R. Macrophagic AMPKα1 orchestrates regenerative inflammation induced by glucocorticoids. EMBO Rep 2023; 24:e55363. [PMID: 36520372 PMCID: PMC9900347 DOI: 10.15252/embr.202255363] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophages are key cells after tissue damage since they mediate both acute inflammatory phase and regenerative inflammation by shifting from pro-inflammatory to restorative cells. Glucocorticoids (GCs) are the most potent anti-inflammatory hormone in clinical use, still their actions on macrophages are not fully understood. We show that the metabolic sensor AMP-activated protein kinase (AMPK) is required for GCs to induce restorative macrophages. GC Dexamethasone activates AMPK in macrophages and GC receptor (GR) phosphorylation is decreased in AMPK-deficient macrophages. Loss of AMPK in macrophages abrogates the GC-induced acquisition of their repair phenotype and impairs GC-induced resolution of inflammation in vivo during post-injury muscle regeneration and acute lung injury. Mechanistically, two categories of genes are impacted by GC treatment in macrophages. Firstly, canonical cytokine regulation by GCs is not affected by AMPK loss. Secondly, AMPK-dependent GC-induced genes required for the phenotypic transition of macrophages are co-regulated by the transcription factor FOXO3, an AMPK substrate. Thus, beyond cytokine regulation, GR requires AMPK-FOXO3 for immunomodulatory actions in macrophages, linking their metabolic status to transcriptional control in regenerative inflammation.
Collapse
Affiliation(s)
- Giorgio Caratti
- Institute of Comparative Molecular EndocrinologyUniversität UlmUlmGermany
| | - Thibaut Desgeorges
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217Université de LyonLyonFrance
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217Université de LyonLyonFrance
| | - Ulrich Stifel
- Institute of Comparative Molecular EndocrinologyUniversität UlmUlmGermany
| | - Aurélie Fessard
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217Université de LyonLyonFrance
| | - Mascha Koenen
- Institute of Comparative Molecular EndocrinologyUniversität UlmUlmGermany
- Present address:
Laboratory of Molecular MetabolismThe Rockefeller UniversityNew YorkNYUSA
| | - Bozhena Caratti
- Institute of Comparative Molecular EndocrinologyUniversität UlmUlmGermany
| | - Marine Théret
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217Université de LyonLyonFrance
- Present address:
Department of Medical GeneticsSchool of Biomedical Engineering and the Biomedical Research CentreVancouverBCCanada
| | - Carsten Skurk
- Department of CardiologyCharité Universitätsmedizin BerlinBerlinGermany
- Franklin/German Centre for Cardiovascular Research (DZHK), Partner Site Berlin/Institute of Health (BIH)BerlinGermany
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217Université de LyonLyonFrance
| | - Jan P Tuckermann
- Institute of Comparative Molecular EndocrinologyUniversität UlmUlmGermany
| | - Rémi Mounier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217Université de LyonLyonFrance
| |
Collapse
|
11
|
Akalestou E, Lopez-Noriega L, Christakis I, Hu M, Miras AD, Leclerc I, Rutter GA. Vertical sleeve gastrectomy normalizes circulating glucocorticoid levels and lowers glucocorticoid action tissue-selectively in mice. Front Endocrinol (Lausanne) 2022; 13:1020576. [PMID: 36246869 PMCID: PMC9556837 DOI: 10.3389/fendo.2022.1020576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Glucocorticoids produced by the adrenal cortex are essential for the maintenance of metabolic homeostasis. Glucocorticoid activation is catalysed by 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1). Excess glucocorticoids are associated with insulin resistance and hyperglycaemia. A small number of studies have demonstrated effects on glucocorticoid metabolism of bariatric surgery, a group of gastrointestinal procedures known to improve insulin sensitivity and secretion, which were assumed to result from weight loss. In this study, we hypothesize that a reduction in glucocorticoid action following bariatric surgery contributes to the widely observed euglycemic effects of the treatment. Methods Glucose and insulin tolerance tests were performed at ten weeks post operatively and circulating corticosterone was measured. Liver and adipose tissues were harvested from fed mice and 11β-HSD1 levels were measured by quantitative RT-PCR or Western (immuno-) blotting, respectively. 11β-HSD1 null mice (Hsd11b1 -/-) were generated using CRISPR/Cas9 genome editing. Wild type and littermate Hsd11b1 -/- mice underwent Vertical Sleeve Gastrectomy (VSG) or sham surgery. Results Under the conditions used, no differences in weight loss were observed between VSG treated and sham operated mice. However, both lean and obese WT VSG mice displayed significantly improved glucose clearance and insulin sensitivity. Remarkably, VSG restored physiological corticosterone production in HFD mice and reduced 11β-HSD1 expression in liver and adipose tissue post-surgery. Elimination of the 11β-HSD1/Hsd11b1 gene by CRISPR/Cas9 mimicked the effects of VSG on body weight and tolerance to 1g/kg glucose challenge. However, at higher glucose loads, the euglycemic effect of VSG was superior to Hsd11b1 elimination. Conclusions Bariatric surgery improves insulin sensitivity and reduces glucocorticoid activation at the tissular level, under physiological and pathophysiological (obesity) conditions, irrespective of weight loss. These findings point towards a physiologically relevant gut-glucocorticoid axis, and suggest that lowered glucocorticoid exposure may represent an additional contribution to the health benefits of bariatric surgery.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Livia Lopez-Noriega
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Ioannis Christakis
- Endocrine and General Surgery, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alexander D. Miras
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
12
|
Santoleri D, Lim HW, Emmett MJ, Stoute J, Gavin MJ, Sostre-Colón J, Uehara K, Welles JE, Liu KF, Lazar MA, Titchenell PM. Global-run on sequencing identifies Gm11967 as an Akt-dependent long noncoding RNA involved in insulin sensitivity. iScience 2022; 25:104410. [PMID: 35663017 PMCID: PMC9156944 DOI: 10.1016/j.isci.2022.104410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/18/2022] [Accepted: 05/11/2022] [Indexed: 01/07/2023] Open
Abstract
The insulin responsive Akt and FoxO1 signaling axis is a key regulator of the hepatic transcriptional response to nutrient intake. Here, we used global run-on sequencing (GRO-seq) to measure the nascent transcriptional response to fasting and refeeding as well as define the specific role of hepatic Akt and FoxO1 signaling in mediating this response. We identified 599 feeding-regulated transcripts, as well as over 6,000 eRNAs, and mapped their dependency on Akt and FoxO1 signaling. Further, we identified several feeding-regulated lncRNAs, including the lncRNA Gm11967, whose expression was dependent upon the liver Akt-FoxO1 axis. Restoring Gm11967 expression in mice lacking liver Akt improved insulin sensitivity and induced glucokinase protein expression, indicating that Akt-dependent control of Gm11967 contributes to the translational control of glucokinase. More broadly, we have generated a unique genome-wide dataset that defines the feeding and Akt/FoxO1-dependent transcriptional changes in response to nutrient availability.
Collapse
Affiliation(s)
- Dominic Santoleri
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Matthew J. Emmett
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Julian Stoute
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Matthew J. Gavin
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Jaimarie Sostre-Colón
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Kahealani Uehara
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Jaclyn E. Welles
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Kathy Fange Liu
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mitchell A. Lazar
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul M. Titchenell
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Hussein HK, Aubead NM, Kzar HH, Karim YS, Amin AH, Al-Gazally ME, Ahmed TI, Jawad MA, Hammid AT, Jalil AT, Mustafa YF, Saleh MM, Heydari H. Association of cord blood asprosin concentration with atherogenic lipid profile and anthropometric indices. Diabetol Metab Syndr 2022; 14:74. [PMID: 35585615 PMCID: PMC9118590 DOI: 10.1186/s13098-022-00844-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Elevated lipids in umbilical cord blood affect fetal programming, leading to a higher risk of developing cardiovascular disease in later life. However, the causes of changes in the lipid profile of umbilical cord blood are not clear yet. This study aimed for the first time to determine the association of asprosin concentration with TAG, TC, HDL-C, LDL-C concentrations and TAG/HDL-C, TC/HDL-C, LDL-C/HDL-C and non-HDL-C/HDL-C ratio in umbilical cord blood as well as newborn anthropometric indices. This cross-sectional study was based on 450 mother- newborn pairs of a birth cohort study in Sabzevar, Iran. Multiple linear regression was used to estimate the association of lipid concentration and lipid ratios as well as birth weight (BW), birth length (BL), head circumference (HC) and chest circumference (CC) with asprosin in cord blood samples controlled for the relevant covariates. RESULT In fully adjusted models, each 1 ng/mL increase in asprosin was associated with 0.19 (95% CI 0.06, 0.31, P < 0.01), 0.19 (95% CI 0.10, 0.29, P < 0.01), 0.17 (95% CI 0.09, 0.25, P < 0.01), 0.17 (95% CI 0.09, 0.25, P < 0.01), 0.01 (95% CI 0.00, 0.013, P < 0.01), 0.01 (95% CI 0.01, 0.01, P < 0.01), 0.01 (95% CI 0.01, 0.01, P < 0.01) and 0.01 (95% CI 0.01, 0.01, P < 0.01) increase in TAG, TC, LDL-C, TAG/HDL-C, TC/HDL-C, LDL-C/HDL-C and non-HDL-C/HDL-C ratio respectively. Moreover, higher asprosin levels was positively associated with newborn BW, BL, HC and CC; however, these associations were not statistically significant. CONCLUSION Overall, our findings support the positive association between cord asprosin concentration and the development of atherogenic lipid profile in newborns. Further studies are needed to confirm the findings of this study in other populations.
Collapse
Affiliation(s)
| | - Nassrin Malik Aubead
- Department of Obstetrics and Gynecology, Hammurabi College of Medicine, University of Babylon, Babil, Iraq
| | - Hamzah H Kzar
- Veterinary Medicine College, Al-Qasim Green University, Al-Qasim, Iraq
| | | | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | | | | | | | - Ali Thaeer Hammid
- Computer Engineering Techniques, Faculty of Information Technology, Imam Ja'afar Al-Sadiq University, Baghdad, Iraq
| | - Abduladheem Turki Jalil
- Faculty of Biology and Ecology, Yanka Kupala State University of Grodno, 230023, Grodno, Belarus
- College of Technical Engineering, The Islamic University, Najaf, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Marwan Mahmood Saleh
- Department of Biophysics, College of Applied Sciences, University Of Anbar, Ramadi, Iraq
| | - Hafez Heydari
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| |
Collapse
|
14
|
McKimpson WM, Kuo T, Kitamoto T, Higuchi S, Mills JC, Haeusler RA, Accili D. FOXO1 Is Present in Stomach Epithelium and Determines Gastric Cell Distribution. GASTRO HEP ADVANCES 2022; 1:733-745. [PMID: 36117550 PMCID: PMC9481069 DOI: 10.1016/j.gastha.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND AIMS Stomach cells can be converted to insulin-producing cells by Neurog3, MafA, and Pdxl over-expression. Enteroendocrine cells can be similarly made to produce insulin by the deletion of FOXO1. Characteristics and functional properties of FOXO1-expressing stomach cells are not known. METHODS Using mice bearing a FOXO1-GFP knock-in allele and primary cell cultures, we examined the identity of FOXO1-expressing stomach cells and analyzed their features through loss-of-function studies with red-to-green fluorescent reporters. RESULTS FOXO1 localizes to a subset of Neurog3 and parietal cells. FOXO1 deletion ex vivo or in vivo using Neurog3-cre or Atp4b-cre increased numbers of parietal cells, generated insulin- and C-peptide-immunoreactive cells, and raised Neurog3 messenger RNA. Gene expression and ChIP- seq experiments identified the cell cycle regulator cyclin E1 (CCNE1) as a FOXO1 target. CONCLUSION FOXO1 is expressed in a subset of stomach cells. Its ablation increases parietal cells and yields insulin-immunoreactive cells, consistent with a role in lineage determination.
Collapse
Affiliation(s)
- Wendy M. McKimpson
- Division of Endocrinology, Department of Medicine, Columbia University, New York, New York
- Naomi Berrie Diabetes Center, Columbia University, New York, New York
| | - Taiyi Kuo
- Division of Endocrinology, Department of Medicine, Columbia University, New York, New York
- Naomi Berrie Diabetes Center, Columbia University, New York, New York
| | - Takumi Kitamoto
- Division of Endocrinology, Department of Medicine, Columbia University, New York, New York
- Naomi Berrie Diabetes Center, Columbia University, New York, New York
| | - Sei Higuchi
- Naomi Berrie Diabetes Center, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Jason C. Mills
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Rebecca A. Haeusler
- Naomi Berrie Diabetes Center, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Domenico Accili
- Division of Endocrinology, Department of Medicine, Columbia University, New York, New York
- Naomi Berrie Diabetes Center, Columbia University, New York, New York
| |
Collapse
|
15
|
Takeuchi Y, Yahagi N, Aita Y, Mehrazad-Saber Z, Ho MH, Huyan Y, Murayama Y, Shikama A, Masuda Y, Izumida Y, Miyamoto T, Matsuzaka T, Kawakami Y, Shimano H. FoxO-KLF15 pathway switches the flow of macronutrients under the control of insulin. iScience 2021; 24:103446. [PMID: 34988390 PMCID: PMC8710527 DOI: 10.1016/j.isci.2021.103446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/18/2021] [Accepted: 11/11/2021] [Indexed: 11/15/2022] Open
Abstract
KLF15 is a transcription factor that plays an important role in the activation of gluconeogenesis from amino acids as well as the suppression of lipogenesis from glucose. Here we identified the transcription start site of liver-specific KLF15 transcript and showed that FoxO1/3 transcriptionally regulates Klf15 gene expression by directly binding to the liver-specific Klf15 promoter. To achieve this, we performed a precise in vivo promoter analysis combined with the genome-wide transcription-factor-screening method "TFEL scan", using our original Transcription Factor Expression Library (TFEL), which covers nearly all the transcription factors in the mouse genome. Hepatic Klf15 expression is significantly increased via FoxOs by attenuating insulin signaling. Furthermore, FoxOs elevate the expression levels of amino acid catabolic enzymes and suppress SREBP-1c via KLF15, resulting in accelerated amino acid breakdown and suppressed lipogenesis during fasting. Thus, the FoxO-KLF15 pathway contributes to switching the macronutrient flow in the liver under the control of insulin.
Collapse
Affiliation(s)
- Yoshinori Takeuchi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Naoya Yahagi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuichi Aita
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Zahra Mehrazad-Saber
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Man Hei Ho
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yiren Huyan
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Murayama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Akito Shikama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yukari Masuda
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshihiko Izumida
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takafumi Miyamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasushi Kawakami
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
16
|
Kitamoto T, Kuo T, Okabe A, Kaneda A, Accili D. An integrative transcriptional logic model of hepatic insulin resistance. Proc Natl Acad Sci U S A 2021; 118:e2102222118. [PMID: 34732569 PMCID: PMC8609333 DOI: 10.1073/pnas.2102222118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 12/27/2022] Open
Abstract
Abnormalities of lipid/lipoprotein and glucose metabolism are hallmarks of hepatic insulin resistance in type 2 diabetes. The former antedate the latter, but the latter become progressively refractory to treatment and contribute to therapeutic failures. It's unclear whether the two processes share a common pathogenesis and what underlies their progressive nature. In this study, we investigated the hypothesis that genes in the lipid/lipoprotein pathway and those in the glucose metabolic pathway are governed by different transcriptional regulatory logics that affect their response to physiologic (fasting/refeeding) as well as pathophysiologic cues (insulin resistance and hyperglycemia). To this end, we obtained genomic and transcriptomic maps of the key insulin-regulated transcription factor, FoxO1, and integrated them with those of CREB, PPAR-α, and glucocorticoid receptor. We found that glucose metabolic genes are primarily regulated by promoter and intergenic enhancers in a fasting-dependent manner, while lipid genes are regulated through fasting-dependent intron enhancers and fasting-independent enhancerless introns. Glucose genes also showed a remarkable transcriptional resiliency (i.e., the ability to compensate following constitutive FoxO1 ablation through an enrichment of active marks at shared PPAR-α/FoxO1 regulatory elements). Unexpectedly, insulin resistance and hyperglycemia were associated with a "spreading" of FoxO1 binding to enhancers and the emergence of unique target sites. We surmise that this unusual pattern correlates with the progressively intractable nature of hepatic insulin resistance. This transcriptional logic provides an integrated model to interpret the combined lipid and glucose abnormalities of type 2 diabetes.
Collapse
Affiliation(s)
- Takumi Kitamoto
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032;
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Taiyi Kuo
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Atsushi Okabe
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan
| | - Domenico Accili
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Naomi Berrie Diabetes Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
17
|
Præstholm SM, Correia CM, Goitea VE, Siersbæk MS, Jørgensen M, Havelund JF, Pedersen TÅ, Færgeman NJ, Grøntved L. Impaired glucocorticoid receptor expression in liver disrupts feeding-induced gene expression, glucose uptake, and glycogen storage. Cell Rep 2021; 37:109938. [PMID: 34731602 DOI: 10.1016/j.celrep.2021.109938] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/08/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022] Open
Abstract
The transition from a fasted to a fed state is associated with extensive transcriptional remodeling in hepatocytes facilitated by hormonal- and nutritional-regulated transcription factors. Here, we use a liver-specific glucocorticoid receptor (GR) knockout (L-GRKO) model to investigate the temporal hepatic expression of GR target genes in response to feeding. Interestingly, in addition to the well-described fasting-regulated genes, we identify a subset of hepatic feeding-induced genes that requires GR for full expression. This includes Gck, which is important for hepatic glucose uptake, utilization, and storage. We show that insulin and glucocorticoids cooperatively regulate hepatic Gck expression in a direct GR-dependent manner by a 4.6 kb upstream GR binding site operating as a Gck enhancer. L-GRKO blunts preprandial and early postprandial Gck expression, which ultimately affects early postprandial hepatic glucose uptake, phosphorylation, and glycogen storage. Thus, GR is positively involved in feeding-induced gene expression and important for postprandial glucose metabolism in the liver.
Collapse
Affiliation(s)
- Stine M Præstholm
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Catarina M Correia
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Victor E Goitea
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Majken S Siersbæk
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Mathilde Jørgensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Jesper F Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | | | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark.
| |
Collapse
|
18
|
Xu S, Liu Y, Hu R, Wang M, Stöhr O, Xiong Y, Chen L, Kang H, Zheng L, Cai S, He L, Wang C, Copps KD, White MF, Miao J. TAZ inhibits glucocorticoid receptor and coordinates hepatic glucose homeostasis in normal physiological states. eLife 2021; 10:e57462. [PMID: 34622775 PMCID: PMC8555985 DOI: 10.7554/elife.57462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/13/2021] [Indexed: 12/11/2022] Open
Abstract
The elucidation of the mechanisms whereby the liver maintains glucose homeostasis is crucial for the understanding of physiological and pathological states. Here, we show a novel role of hepatic transcriptional co-activator with PDZ-binding motif (TAZ) in the inhibition of glucocorticoid receptor (GR). TAZ is abundantly expressed in pericentral hepatocytes and its expression is markedly reduced by fasting. TAZ interacts via its WW domain with the ligand-binding domain of GR to limit the binding of GR to the GR response element in gluconeogenic gene promoters. Therefore, liver-specific TAZ knockout mice show increases in glucose production and blood glucose concentration. Conversely, the overexpression of TAZ in mouse liver reduces the binding of GR to gluconeogenic gene promoters and glucose production. Thus, our findings demonstrate that hepatic TAZ inhibits GR transactivation of gluconeogenic genes and coordinates gluconeogenesis in response to physiological fasting and feeding.
Collapse
Affiliation(s)
- Simiao Xu
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Branch of the National Clinical Research Center for Metabolic DiseaseWuhanChina
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Yangyang Liu
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Ruixiang Hu
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Min Wang
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of Pathology, Beth Israel Deaconess Medical CenterBostonUnited States
| | - Oliver Stöhr
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Yibo Xiong
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Liang Chen
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- College of Science, Northeastern UniversityBostonUnited States
| | - Hong Kang
- Department of Systemic Biology, Harvard Medical SchoolBostonUnited States
| | - Lingyun Zheng
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Songjie Cai
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Li He
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Cunchuan Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Kyle D Copps
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Morris F White
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Ji Miao
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Pediatrics, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
19
|
Schilperoort M, Kroon J, Kooijman S, Smit AE, Gentenaar M, Mletzko K, Schmidt FN, van Ruijven L, Busse B, Pereira AM, Appelman‐Dijkstra NM, Bravenboer N, Rensen PC, Meijer OC, Winter EM. Loss of glucocorticoid rhythm induces an osteoporotic phenotype in female mice. Aging Cell 2021; 20:e13474. [PMID: 34592793 PMCID: PMC8520718 DOI: 10.1111/acel.13474] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/07/2021] [Indexed: 12/21/2022] Open
Abstract
Glucocorticoid (GC)-induced osteoporosis is a widespread health problem that is accompanied with increased fracture risk. Detrimental effects of anti-inflammatory GC therapy on bone have been ascribed to the excess in GC exposure, but it is unknown whether there is also a role for disruption of the endogenous GC rhythm that is inherent to GC therapy. To investigate this, we implanted female C57Bl/6J mice with slow-release corticosterone (CORT) pellets to blunt the rhythm in CORT levels without inducing hypercortisolism. Flattening of CORT rhythm reduced cortical and trabecular bone volume and thickness, whilst bone structure was maintained in mice injected with supraphysiologic CORT at the time of their endogenous GC peak. Mechanistically, mice with a flattened CORT rhythm showed disrupted circadian gene expression patterns in bone, along with changes in circulating bone turnover markers indicative of a negative balance in bone remodelling. Indeed, double calcein labelling of bone in vivo revealed a reduced bone formation in mice with a flattened CORT rhythm. Collectively, these perturbations in bone turnover and structure decreased bone strength and stiffness, as determined by mechanical testing. In conclusion, we demonstrate for the first time that flattening of the GC rhythm disrupts the circadian clock in bone and results in an osteoporotic phenotype in mice. Our findings indicate that at least part of the fracture risk associated with GC therapy may be the consequence of a disturbed GC rhythm, rather than excess GC exposure alone, and that a dampened GC rhythm may contribute to the age-related risk of osteoporosis.
Collapse
Affiliation(s)
- Maaike Schilperoort
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
| | - Jan Kroon
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
| | - Sander Kooijman
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
| | - Annelies E. Smit
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
| | - Max Gentenaar
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
| | - Kathrin Mletzko
- Department of Osteology and Biomechanics (IOBM)University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Felix N. Schmidt
- Department of Osteology and Biomechanics (IOBM)University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Leo van Ruijven
- Department of Functional AnatomyAcademic Center for Dentistry Amsterdam (ACTA)AmsterdamThe Netherlands
| | - Björn Busse
- Department of Osteology and Biomechanics (IOBM)University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Alberto M. Pereira
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
| | - Natasha M. Appelman‐Dijkstra
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Department of MedicineCenter for Bone QualityLeiden University Medical CenterLeidenThe Netherlands
| | - Nathalie Bravenboer
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Department of MedicineCenter for Bone QualityLeiden University Medical CenterLeidenThe Netherlands
- Department of Clinical ChemistryVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Patrick C.N. Rensen
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
| | - Onno C. Meijer
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
| | - Elizabeth M. Winter
- Department of MedicineDivision of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeidenThe Netherlands
- Department of MedicineCenter for Bone QualityLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
20
|
Spies LML, Verhoog NJD, Louw A. Acquired Glucocorticoid Resistance Due to Homologous Glucocorticoid Receptor Downregulation: A Modern Look at an Age-Old Problem. Cells 2021; 10:2529. [PMID: 34685511 PMCID: PMC8533966 DOI: 10.3390/cells10102529] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
For over 70 years, the unique anti-inflammatory properties of glucocorticoids (GCs), which mediate their effects via the ligand-activated transcription factor, the glucocorticoid receptor alpha (GRα), have allowed for the use of these steroid hormones in the treatment of various autoimmune and inflammatory-linked diseases. However, aside from the onset of severe side-effects, chronic GC therapy often leads to the ligand-mediated downregulation of the GRα which, in turn, leads to a decrease in GC sensitivity, and effectively, the development of acquired GC resistance. Although the ligand-mediated downregulation of GRα is well documented, the precise factors which influence this process are not well understood and, thus, the development of an acquired GC resistance presents an ever-increasing challenge to the pharmaceutical industry. Recently, however, studies have correlated the dimerization status of the GRα with its ligand-mediated downregulation. Therefore, the current review will be discussing the major role-players in the homologous downregulation of the GRα pool, with a specific focus on previously reported GC-mediated reductions in GRα mRNA and protein levels, the molecular mechanisms through which the GRα functional pool is maintained and the possible impact of receptor conformation on GC-mediated GRα downregulation.
Collapse
Affiliation(s)
| | | | - Ann Louw
- Department of Biochemistry, Stellenbosch University, Van de Byl Street, Stellenbosch 7200, South Africa; (L.-M.L.S.); (N.J.D.V.)
| |
Collapse
|
21
|
An inhibitor-mediated beta-cell dedifferentiation model reveals distinct roles for FoxO1 in glucagon repression and insulin maturation. Mol Metab 2021; 54:101329. [PMID: 34454092 PMCID: PMC8476777 DOI: 10.1016/j.molmet.2021.101329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE The loss of forkhead box protein O1 (FoxO1) signaling in response to metabolic stress contributes to the etiology of type II diabetes, causing the dedifferentiation of pancreatic beta cells to a cell type reminiscent of endocrine progenitors. Lack of methods to easily model this process in vitro, however, have hindered progress into the identification of key downstream targets and potential inhibitors. We therefore aimed to establish such an in vitro cellular dedifferentiation model and apply it to identify novel agents involved in the maintenance of beta-cell identity. METHODS The murine beta-cell line, Min6, was used for primary experiments and high-content screening. Screens encompassed a library of small-molecule drugs representing the chemical and target space of all FDA-approved small molecules with an automated immunofluorescence readout. Validation experiments were performed in a murine alpha-cell line as well as in primary murine and human diabetic islets. Developmental effects were studied in zebrafish and C. elegans models, while diabetic db/db mouse models were used to elucidate global glucose metabolism outcomes. RESULTS We show that short-term pharmacological FoxO1 inhibition can model beta-cell dedifferentiation by downregulating beta-cell-specific transcription factors, resulting in the aberrant expression of progenitor genes and the alpha-cell marker glucagon. From a high-content screen, we identified loperamide as a small molecule that can prevent FoxO inhibitor-induced glucagon expression and further stimulate insulin protein processing and secretion by altering calcium levels, intracellular pH, and FoxO1 localization. CONCLUSIONS Our study provides novel models, molecular targets, and drug candidates for studying and preventing beta-cell dedifferentiation.
Collapse
|
22
|
Diaz-Jimenez D, Kolb JP, Cidlowski JA. Glucocorticoids as Regulators of Macrophage-Mediated Tissue Homeostasis. Front Immunol 2021; 12:669891. [PMID: 34079551 PMCID: PMC8165320 DOI: 10.3389/fimmu.2021.669891] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
Our immune system has evolved as a complex network of cells and tissues tasked with maintaining host homeostasis. This is evident during the inflammatory responses elicited during a microbial infection or traumatic tissue damage. These responses seek to eliminate foreign material or restore tissue integrity. Even during periods without explicit disturbances, the immune system plays prominent roles in tissue homeostasis. Perhaps one of the most studied cells in this regard is the macrophage. Tissue-resident macrophages are a heterogenous group of sensory cells that respond to a variety of environmental cues and are essential for organ function. Endogenously produced glucocorticoid hormones connect external environmental stress signals with the function of many cell types, producing profound changes in immune cells, including macrophages. Here, we review the current literature which demonstrates specific effects of glucocorticoids in several organ systems. We propose that tissue-resident macrophages, through glucocorticoid signaling, may play an underappreciated role as regulators of organ homeostasis.
Collapse
Affiliation(s)
- David Diaz-Jimenez
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Joseph P Kolb
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John A Cidlowski
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
23
|
Korenfeld N, Finkel M, Buchshtab N, Bar-Shimon M, Charni-Natan M, Goldstein I. Fasting Hormones Synergistically Induce Amino Acid Catabolism Genes to Promote Gluconeogenesis. Cell Mol Gastroenterol Hepatol 2021; 12:1021-1036. [PMID: 33957303 PMCID: PMC8346669 DOI: 10.1016/j.jcmgh.2021.04.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS Gluconeogenesis from amino acids (AAs) maintains glucose homeostasis during fasting. Although glucagon is known to regulate AA catabolism, the contribution of other hormones to it and the scope of transcriptional regulation dictating AA catabolism are unknown. We explored the role of the fasting hormones glucagon and glucocorticoids in transcriptional regulation of AA catabolism genes and AA-dependent gluconeogenesis. METHODS We tested the RNA expression of AA catabolism genes and glucose production in primary mouse hepatocytes treated with fasting hormones (glucagon, corticosterone) and feeding hormones (insulin, fibroblast growth factor 19). We analyzed genomic data of chromatin accessibility and chromatin immunoprecipitation in mice and primary mouse hepatocytes. We performed chromatin immunoprecipitation in livers of fasted mice to show binding of cAMP responsive element binding protein (CREB) and the glucocorticoid receptor (GR). RESULTS Fasting induced the expression of 31 genes with various roles in AA catabolism. Of them, 15 were synergistically induced by co-treatment of glucagon and corticosterone. Synergistic gene expression relied on the activity of both CREB and GR and was abolished by treatment with either insulin or fibroblast growth factor 19. Enhancers adjacent to synergistically induced genes became more accessible and were bound by CREB and GR on fasting. Akin to the gene expression pattern, gluconeogenesis from AAs was synergistically induced by glucagon and corticosterone in a CREB- and GR-dependent manner. CONCLUSIONS Transcriptional regulation of AA catabolism genes during fasting is widespread and is driven by glucagon (via CREB) and corticosterone (via GR). Glucose production in hepatocytes is also synergistically augmented, showing that glucagon alone is insufficient in fully activating gluconeogenesis.
Collapse
Affiliation(s)
- Noga Korenfeld
- Institute of Biochemistry, Food Science and Nutrition. Robert H. Smith Faculty of Agriculture, Food and EnvironmentThe Hebrew University of Jerusalem, Rehovot, Israel
| | - Maya Finkel
- Institute of Biochemistry, Food Science and Nutrition. Robert H. Smith Faculty of Agriculture, Food and EnvironmentThe Hebrew University of Jerusalem, Rehovot, Israel
| | - Nufar Buchshtab
- Institute of Biochemistry, Food Science and Nutrition. Robert H. Smith Faculty of Agriculture, Food and EnvironmentThe Hebrew University of Jerusalem, Rehovot, Israel
| | - Meirav Bar-Shimon
- Institute of Biochemistry, Food Science and Nutrition. Robert H. Smith Faculty of Agriculture, Food and EnvironmentThe Hebrew University of Jerusalem, Rehovot, Israel
| | - Meital Charni-Natan
- Institute of Biochemistry, Food Science and Nutrition. Robert H. Smith Faculty of Agriculture, Food and EnvironmentThe Hebrew University of Jerusalem, Rehovot, Israel
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition. Robert H. Smith Faculty of Agriculture, Food and EnvironmentThe Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
24
|
Chen TC, Kuo T, Dandan M, Lee RA, Chang M, Villivalam SD, Liao SC, Costello D, Shankaran M, Mohammed H, Kang S, Hellerstein MK, Wang JC. The role of striated muscle Pik3r1 in glucose and protein metabolism following chronic glucocorticoid exposure. J Biol Chem 2021; 296:100395. [PMID: 33567340 PMCID: PMC8010618 DOI: 10.1016/j.jbc.2021.100395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 11/03/2022] Open
Abstract
Chronic glucocorticoid exposure causes insulin resistance and muscle atrophy in skeletal muscle. We previously identified phosphoinositide-3-kinase regulatory subunit 1 (Pik3r1) as a primary target gene of skeletal muscle glucocorticoid receptors involved in the glucocorticoid-mediated suppression of insulin action. However, the in vivo functions of Pik3r1 remain unclear. Here, we generated striated muscle-specific Pik3r1 knockout (MKO) mice and treated them with a dexamethasone (DEX), a synthetic glucocorticoid. Treating wildtype (WT) mice with DEX attenuated insulin activated Akt activity in liver, epididymal white adipose tissue, and gastrocnemius (GA) muscle. This DEX effect was diminished in GA muscle of MKO mice, therefore, resulting in improved glucose and insulin tolerance in DEX-treated MKO mice. Stable isotope labeling techniques revealed that in WT mice, DEX treatment decreased protein fractional synthesis rates in GA muscle. Furthermore, histology showed that in WT mice, DEX treatment reduced GA myotube diameters. In MKO mice, myotube diameters were smaller than in WT mice, and there were more fast oxidative fibers. Importantly, DEX failed to further reduce myotube diameters. Pik3r1 knockout also decreased basal protein synthesis rate (likely caused by lower 4E-BP1 phosphorylation at Thr37/Thr46) and curbed the ability of DEX to attenuate protein synthesis rate. Finally, the ability of DEX to inhibit eIF2α phosphorylation and insulin-induced 4E-BP1 phosphorylation was reduced in MKO mice. Taken together, these results demonstrate the role of Pik3r1 in glucocorticoid-mediated effects on glucose and protein metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Tzu-Chieh Chen
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Taiyi Kuo
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Mohamad Dandan
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Rebecca A Lee
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Maggie Chang
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Sneha D Villivalam
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Szu-Chi Liao
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Damian Costello
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Hussein Mohammed
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Sona Kang
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Marc K Hellerstein
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Jen-Chywan Wang
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA.
| |
Collapse
|
25
|
Præstholm SM, Correia CM, Grøntved L. Multifaceted Control of GR Signaling and Its Impact on Hepatic Transcriptional Networks and Metabolism. Front Endocrinol (Lausanne) 2020; 11:572981. [PMID: 33133019 PMCID: PMC7578419 DOI: 10.3389/fendo.2020.572981] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids (GCs) and the glucocorticoid receptor (GR) are important regulators of development, inflammation, stress response and metabolism, demonstrated in various diseases including Addison's disease, Cushing's syndrome and by the many side effects of prolonged clinical administration of GCs. These conditions include severe metabolic challenges in key metabolic organs like the liver. In the liver, GR is known to regulate the transcription of key enzymes in glucose and lipid metabolism and contribute to the regulation of circadian-expressed genes. Insights to the modes of GR regulation and the underlying functional mechanisms are key for understanding diseases and for the development of improved clinical uses of GCs. The activity and function of GR is regulated at numerous levels including ligand availability, interaction with heat shock protein (HSP) complexes, expression of GR isoforms and posttranslational modifications. Moreover, recent genomics studies show functional interaction with multiple transcription factors (TF) and coregulators in complex transcriptional networks controlling cell type-specific gene expression by GCs. In this review we describe the different regulatory steps important for GR activity and discuss how different TF interaction partners of GR selectively control hepatic gene transcription and metabolism.
Collapse
Affiliation(s)
| | | | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
26
|
C57BL/6J substrain differences in response to high-fat diet intervention. Sci Rep 2020; 10:14052. [PMID: 32820201 PMCID: PMC7441320 DOI: 10.1038/s41598-020-70765-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
C57BL/6J-related mouse strains are widely used animal models for diet-induced obesity (DIO). Multiple vendors breed C57BL/6J-related substrains which may introduce genetic drift and environmental confounders such as microbiome differences. To address potential vendor/substrain specific effects, we compared DIO of C57BL/6J-related substrains from three different vendors: C57BL/6J (Charles Rivers), C57BL/6JBomTac (Taconic Bioscience) and C57BL/6JRj (Janvier). After local acclimatization, DIO was induced by either a high-fat diet (HFD, 60% energy from fat) or western diet (WD, 42% energy from fat supplemented with fructose in the drinking water). All three groups on HFD gained a similar amount of total body weight, yet the relative amount of fat percentage and mass of inguinal- and epididymal white adipose tissue (iWAT and eWAT) was lower in C57BL/6JBomTac compared to the two other C57BL/6J-releated substrains. In contrast to HFD, the three groups on WD responded differently in terms of body weight gain, where C57BL/6J was particularly prone to WD. This was associated with a relative higher amount of eWAT, iWAT, and liver triglycerides. Although the HFD and WD had significant impact on the microbiota, we did not observe any major differences between the three groups of mice. Together, these data demonstrate significant differences in HFD- and WD-induced adiposity in C57BL/6J-related substrains, which should be considered in the design of animal DIO studies.
Collapse
|
27
|
Guan D, Xiong Y, Trinh TM, Xiao Y, Hu W, Jiang C, Dierickx P, Jang C, Rabinowitz JD, Lazar MA. The hepatocyte clock and feeding control chronophysiology of multiple liver cell types. Science 2020; 369:1388-1394. [PMID: 32732282 DOI: 10.1126/science.aba8984] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022]
Abstract
Most cells of the body contain molecular clocks, but the requirement of peripheral clocks for rhythmicity and their effects on physiology are not well understood. We show that deletion of core clock components REV-ERBα and REV-ERBβ in adult mouse hepatocytes disrupts diurnal rhythms of a subset of liver genes and alters the diurnal rhythm of de novo lipogenesis. Liver function is also influenced by nonhepatocytic cells, and the loss of hepatocyte REV-ERBs remodels the rhythmic transcriptomes and metabolomes of multiple cell types within the liver. Finally, alteration of food availability demonstrates the hierarchy of the cell-intrinsic hepatocyte clock mechanism and the feeding environment. Together, these studies reveal previously unsuspected roles of the hepatocyte clock in the physiological coordination of nutritional signals and cell-cell communication controlling rhythmic metabolism.
Collapse
Affiliation(s)
- Dongyin Guan
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Xiong
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Trang Minh Trinh
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yang Xiao
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenxiang Hu
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chunjie Jiang
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pieterjan Dierickx
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cholsoon Jang
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA. .,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Godskesen LE, Lassen TR, Jespersen NR, Siersbæk M, Yan Y, Nielsen MM, Tjønnfjord SK, Grøntved L, Madsen G, Kjems J, Bøtker HE, Schmidt MR, Krag A, Kjeldsen J. Remote ischemic conditioning in active ulcerative colitis: An explorative randomized clinical trial. Sci Rep 2020; 10:9537. [PMID: 32533085 PMCID: PMC7293253 DOI: 10.1038/s41598-020-65692-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023] Open
Abstract
Remote ischemic conditioning (RIC) by repetitive brief periods of limb ischemia and reperfusion renders organs more resistant to ischemic injury. The protection is partly through down-regulation of the inflammatory response. Our aim was to investigate the clinical and anti-inflammatory effects of RIC in patients with active ulcerative colitis (UC). We included 22 patients with active UC in this explorative, randomized, sham-controlled clinical trial. The patients were randomly assigned 1:1 to RIC (induced in the arm through four cycles of 5-min inflation and 5-min deflation of a blood-pressure cuff) or sham (incomplete inflation of the blood-pressure cuff) once daily for 10 days. Outcome variables were measured at baseline and on day 11. When compared with sham, RIC did not affect inflammation in the UC patients measured by fecal calprotectin, plasma C-reactive protein, Mayo Score, Mayo Endoscopic Subscore, Nancy Histological Index or inflammatory cytokines involved in UC and RIC. The mRNA and miRNA expression profiles in the UC patients were measured by RNA sequencing and multiplexed hybridization, respectively, but were not significantly affected by RIC. We used the Langendorff heart model to assess activation of the organ protective mechanism induced by RIC, but could not confirm activation of the organ protective mechanism in the UC patients.
Collapse
Affiliation(s)
- Line E Godskesen
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
- OPEN - Odense Patient data Explorative Network, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas R Lassen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Majken Siersbæk
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Yan Yan
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Michael M Nielsen
- Internal Medicine & Emergency Department, Odense University Hospital - Svendborg Sygehus, Svendborg, Denmark
| | - Sara K Tjønnfjord
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Gunvor Madsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael R Schmidt
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Aleksander Krag
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Jens Kjeldsen
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
29
|
JARID1a Ablation in the Liver Alters Systemic Metabolism and Adaptation to Feeding. Cell Rep 2020; 31:107668. [PMID: 32460011 DOI: 10.1016/j.celrep.2020.107668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/03/2020] [Accepted: 01/29/2020] [Indexed: 11/22/2022] Open
Abstract
The liver is a key regulator of systemic energy homeostasis whose proper function is dependent on the circadian clock. Here, we show that livers deficient in the oscillator component JARID1a exhibit a dysregulation of genes involved in energy metabolism. Importantly, we find that mice that lack hepatic JARID1a have decreased lean body mass, decreased respiratory exchange ratios, faster production of ketones, and increased glucose production in response to fasting. Finally, we find that JARID1a loss compromises the response of the hepatic transcriptome to nutrient availability. In all, ablation of hepatic JARID1a disrupts the coordination of hepatic metabolic programs with whole-body consequences.
Collapse
|
30
|
Dubois V, Gheeraert C, Vankrunkelsven W, Dubois‐Chevalier J, Dehondt H, Bobowski‐Gerard M, Vinod M, Zummo FP, Güiza F, Ploton M, Dorchies E, Pineau L, Boulinguiez A, Vallez E, Woitrain E, Baugé E, Lalloyer F, Duhem C, Rabhi N, van Kesteren RE, Chiang C, Lancel S, Duez H, Annicotte J, Paumelle R, Vanhorebeek I, Van den Berghe G, Staels B, Lefebvre P, Eeckhoute J. Endoplasmic reticulum stress actively suppresses hepatic molecular identity in damaged liver. Mol Syst Biol 2020; 16:e9156. [PMID: 32407006 PMCID: PMC7224309 DOI: 10.15252/msb.20199156] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Liver injury triggers adaptive remodeling of the hepatic transcriptome for repair/regeneration. We demonstrate that this involves particularly profound transcriptomic alterations where acute induction of genes involved in handling of endoplasmic reticulum stress (ERS) is accompanied by partial hepatic dedifferentiation. Importantly, widespread hepatic gene downregulation could not simply be ascribed to cofactor squelching secondary to ERS gene induction, but rather involves a combination of active repressive mechanisms. ERS acts through inhibition of the liver-identity (LIVER-ID) transcription factor (TF) network, initiated by rapid LIVER-ID TF protein loss. In addition, induction of the transcriptional repressor NFIL3 further contributes to LIVER-ID gene repression. Alteration to the liver TF repertoire translates into compromised activity of regulatory regions characterized by the densest co-recruitment of LIVER-ID TFs and decommissioning of BRD4 super-enhancers driving hepatic identity. While transient repression of the hepatic molecular identity is an intrinsic part of liver repair, sustained disequilibrium between the ERS and LIVER-ID transcriptional programs is linked to liver dysfunction as shown using mouse models of acute liver injury and livers from deceased human septic patients.
Collapse
Affiliation(s)
- Vanessa Dubois
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
- Present address:
Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA)KU LeuvenLeuvenBelgium
| | - Céline Gheeraert
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Wouter Vankrunkelsven
- Clinical Division and Laboratory of Intensive Care MedicineDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | | | - Hélène Dehondt
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | | | - Manjula Vinod
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | | | - Fabian Güiza
- Clinical Division and Laboratory of Intensive Care MedicineDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Maheul Ploton
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Emilie Dorchies
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Laurent Pineau
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Alexis Boulinguiez
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Emmanuelle Vallez
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Eloise Woitrain
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Eric Baugé
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Fanny Lalloyer
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Christian Duhem
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Nabil Rabhi
- UMR 8199 ‐ EGIDCNRSInstitut Pasteur de LilleUniversity of LilleLilleFrance
| | - Ronald E van Kesteren
- Center for Neurogenomics and Cognitive ResearchNeuroscience Campus AmsterdamVU UniversityAmsterdamThe Netherlands
| | - Cheng‐Ming Chiang
- Simmons Comprehensive Cancer CenterDepartments of Biochemistry and PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Steve Lancel
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Hélène Duez
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | | | - Réjane Paumelle
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Ilse Vanhorebeek
- Clinical Division and Laboratory of Intensive Care MedicineDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care MedicineDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Bart Staels
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Philippe Lefebvre
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| | - Jérôme Eeckhoute
- Inserm, CHU LilleInstitut Pasteur de LilleU1011‐EGIDUniversity of LilleLilleFrance
| |
Collapse
|
31
|
Louw A. GR Dimerization and the Impact of GR Dimerization on GR Protein Stability and Half-Life. Front Immunol 2019; 10:1693. [PMID: 31379877 PMCID: PMC6653659 DOI: 10.3389/fimmu.2019.01693] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022] Open
Abstract
Pharmacologically, glucocorticoids, which mediate their effects via the glucocorticoid receptor (GR), are a most effective therapy for inflammatory diseases despite the fact that chronic use causes side-effects and acquired GC resistance. The design of drugs with fewer side-effects and less potential for the development of resistance is therefore considered crucial for improved therapy. Dimerization of the GR is an integral step in glucocorticoid signaling and has been identified as a possible molecular site to target for drug development of anti-inflammatory drugs with an improved therapeutic index. Most of the current understanding regarding the role of GR dimerization in GC signaling derives for dimerization deficient mutants, although the role of ligands biased toward monomerization has also been described. Even though designing for loss of dimerization has mostly been applied for reduction of side-effect profile, designing for loss of dimerization may also be a fruitful strategy for the development of GC drugs with less potential to develop GC resistance. GC-induced resistance affects up to 30% of users and is due to a reduction in the GR functional pool. Several molecular mechanisms of GC-mediated reductions in GR pool have been described, one of which is the autologous down-regulation of GR density by the ubiquitin-proteasome-system (UPS). Loss of GR dimerization prevents autologous down-regulation of the receptor through modulation of interactions with components of the UPS and post-translational modifications (PTMs), such as phosphorylation, which prime the GR for degradation. Rational design of conformationally biased ligands that select for a monomeric GR conformation, which increases GC sensitivity through improving GR protein stability and increasing half-life, may be a productive avenue to explore. However, potential drawbacks to this approach should be considered as well as the advantages and disadvantages in chronic vs. acute treatment regimes.
Collapse
Affiliation(s)
- Ann Louw
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
32
|
Murani E, Trakooljul N, Hadlich F, Ponsuksili S, Wimmers K. Transcriptome Responses to Dexamethasone Depending on Dose and Glucocorticoid Receptor Sensitivity in the Liver. Front Genet 2019; 10:559. [PMID: 31249595 PMCID: PMC6582245 DOI: 10.3389/fgene.2019.00559] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/28/2019] [Indexed: 11/13/2022] Open
Abstract
Tissue sensitivity to glucocorticoids is a key factor dictating outcome of their homeostatic and therapeutic action, whereby liver represents one of the major peripheral targets. Here, we used pigs carrying a natural gain-of-function glucocorticoid receptor (GR) variant Ala610Val (GRAla610Val) as a model to identify genes and pathways related to differential glucocorticoid sensitivity. Animals with different GRAla610Val genotypes were treated either with saline or two different doses of dexamethasone. Genome-wide transcriptional responses depending on treatment, genotype, and their interaction in the liver were investigated using mRNA sequencing. Dexamethasone induced vast transcriptional responses, with more than 30% of present genes being affected. Functional annotation of genes differentially expressed due to dexamethasone treatment suggested that genes related to inflammation respond more sensitively, despite absence of an immune stimulus. In contrast, genes involved in glucose metabolism and cancer appeared to be less sensitive. Analysis of genotype and genotype × treatment interaction revealed that clustered protocadherins, particularly PCDHB7, are most prominently affected by GRAla610Val, mainly depending on dose. GRAla610Val influenced also expression of a set of glucose metabolism related genes, including PPARGC1A and CEBPB, in the absence of dexamethasone though no differences in basal plasma glucose level were observed. This might represent an adaptive response, keeping balance between receptor sensitivity, and level of circulating endogenous glucocorticoids. Administration of low dexamethasone dose changed their expression pattern and induced higher glucose response in carriers of the hypersensitive Val receptor. Our findings suggest that GRAla610Val modulates tissue responses to glucocorticoids dynamically, depending on their circulating level.
Collapse
Affiliation(s)
- Eduard Murani
- Institute for Genome Biology - Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Nares Trakooljul
- Institute for Genome Biology - Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Frieder Hadlich
- Institute for Genome Biology - Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Institute for Genome Biology - Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Klaus Wimmers
- Institute for Genome Biology - Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| |
Collapse
|
33
|
Pacheco-Bernal I, Becerril-Pérez F, Aguilar-Arnal L. Circadian rhythms in the three-dimensional genome: implications of chromatin interactions for cyclic transcription. Clin Epigenetics 2019; 11:79. [PMID: 31092281 PMCID: PMC6521413 DOI: 10.1186/s13148-019-0677-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Circadian rhythms orchestrate crucial physiological functions and behavioral aspects around a day in almost all living forms. The circadian clock is a time tracking system that permits organisms to predict and anticipate periodic environmental fluctuations. The circadian system is hierarchically organized, and a master pacemaker located in the brain synchronizes subsidiary clocks in the rest of the organism. Adequate synchrony between central and peripheral clocks ensures fitness and potentiates a healthy state. Conversely, disruption of circadian rhythmicity is associated with metabolic diseases, psychiatric disorders, or cancer, amongst other pathologies. Remarkably, the molecular machinery directing circadian rhythms consists of an intricate network of feedback loops in transcription and translation which impose 24-h cycles in gene expression across all tissues. Interestingly, the molecular clock collaborates with multitude of epigenetic remodelers to fine tune transcriptional rhythms in a tissue-specific manner. Very exciting research demonstrate that three-dimensional properties of the genome have a regulatory role on circadian transcriptional rhythmicity, from bacteria to mammals. Unexpectedly, highly dynamic long-range chromatin interactions have been revealed during the circadian cycle in mammalian cells, where thousands of regulatory elements physically interact with promoter regions every 24 h. Molecular mechanisms directing circadian dynamics on chromatin folding are emerging, and the coordinated action between the core clock and epigenetic remodelers appears to be essential for these movements. These evidences reveal a critical epigenetic regulatory layer for circadian rhythms and pave the way to uncover molecular mechanisms triggering pathological states associated to circadian misalignment.
Collapse
Affiliation(s)
- Ignacio Pacheco-Bernal
- Instituto de Investigaciones Biomédicas, Departamento de Biología Celular y Fisiología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Fernando Becerril-Pérez
- Instituto de Investigaciones Biomédicas, Departamento de Biología Celular y Fisiología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lorena Aguilar-Arnal
- Instituto de Investigaciones Biomédicas, Departamento de Biología Celular y Fisiología, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|