1
|
Goldberg D, Buchshtab N, Charni-Natan M, Goldstein I. Transcriptional cascades during fasting amplify gluconeogenesis and instigate a secondary wave of ketogenic gene transcription. Liver Int 2024; 44:2964-2982. [PMID: 39162082 DOI: 10.1111/liv.16077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND AIMS During fasting, bodily homeostasis is maintained due to hepatic production of glucose (gluconeogenesis) and ketone bodies (ketogenesis). The main hormones governing hepatic fuel production are glucagon and glucocorticoids that initiate transcriptional programs aimed at supporting gluconeogenesis and ketogenesis. METHODS Using primary mouse hepatocytes as an ex vivo model, we employed transcriptomic analysis (RNA-seq), genome-wide profiling of enhancer dynamics (ChIP-seq), perturbation experiments (inhibitors, shRNA), hepatic glucose production measurements and computational analyses. RESULTS We found that in addition to the known metabolic genes transcriptionally induced by glucagon and glucocorticoids, these hormones induce a set of genes encoding transcription factors (TFs) thereby initiating transcriptional cascades. Upon activation by glucocorticoids, the glucocorticoid receptor (GR) induced the genes encoding two TFs: CCAAT/enhancer-binding protein beta (C/EBPβ) and peroxisome proliferator-activated receptor alpha (PPARα). We found that the GR-C/EBPβ cascade mainly serves as a secondary amplifier of primary hormone-induced gene programs. C/EBPβ augmented gluconeogenic gene expression and hepatic glucose production. Conversely, the GR-PPARα cascade initiated a secondary transcriptional wave of genes supporting ketogenesis. The cascade led to synergistic induction of ketogenic genes which is dependent on protein synthesis. Genome-wide analysis of enhancer dynamics revealed numerous enhancers activated by the GR-PPARα cascade. These enhancers were proximal to ketogenic genes, enriched for the PPARα response element and showed increased PPARα binding. CONCLUSION This study reveals abundant transcriptional cascades occurring during fasting. These cascades serve two separated purposes: the amplification of the gluconeogenic transcriptional program and the induction of a gene program aimed at enhancing ketogenesis.
Collapse
Affiliation(s)
- Dana Goldberg
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Nufar Buchshtab
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Meital Charni-Natan
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
2
|
Xie H, Li G, Fu Y, Jiang N, Yi S, Kong X, Shi J, Yin S, Peng J, Jiang Y, Lu S, Deng H, Xie B. A two-step strategy to expand primary human hepatocytes in vitro with efficient metabolic and regenerative capacities. Stem Cell Res Ther 2024; 15:281. [PMID: 39227965 PMCID: PMC11373096 DOI: 10.1186/s13287-024-03911-0] [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: 05/06/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Primary human hepatocytes (PHHs) are highly valuable for drug-metabolism evaluation, liver disease modeling and hepatocyte transplantation. However, their availability is significantly restricted due to limited donor sources, alongside their constrained proliferation capabilities and reduced functionality when cultured in vitro. To address this challenge, we aimed to develop a novel method to efficiently expand PHHs in vitro without a loss of function. METHODS By mimicking the in vivo liver regeneration route, we developed a two-step strategy involving the de-differentiation/expansion and subsequent maturation of PHHs to generate abundant functional hepatocytes in vitro. Initially, we applied SiPer, a prediction algorithm, to identify candidate small molecules capable of activating liver regenerative transcription factors, thereby formulating a novel hepatic expansion medium to de-differentiate PHHs into proliferative human hepatic progenitor-like cells (ProHPLCs). These ProHPLCs were then re-differentiated into functionally mature hepatocytes using a new hepatocyte maturation condition. Additionally, we investigated the underlying mechanism of PHHs expansion under our new conditions. RESULTS The novel hepatic expansion medium containing hydrocortisone facilitated the de-differentiation of PHHs into ProHPLCs, which exhibited key hepatic progenitor characteristics and demonstrated a marked increase in proliferation capacity compared to cells cultivated in previously established expansion conditions. Remarkably, these subsequent matured hepatocytes rivaled PHHs in terms of transcriptome profiles, drug metabolizing activities and in vivo engraftment capabilities. Importantly, our findings suggest that the enhanced expansion of PHHs by hydrocortisone may be mediated through the PPARα signaling pathway and regenerative transcription factors. CONCLUSIONS This study presents a two-step strategy that initially induces PHHs into a proliferative state (ProHPLCs) to ensure sufficient cell quantity, followed by the maturation of ProHPLCs into fully functional hepatocytes to guarantee optimal cell quality. This approach offers a promising means of producing large numbers of seeding cells for hepatocyte-based applications.
Collapse
Affiliation(s)
- Huangfan Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Guangya Li
- MOE Engineering Research Center of Regenerative Medicine, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100191, China
| | - Yunxi Fu
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Nan Jiang
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Simeng Yi
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xi Kong
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jihang Shi
- Department of Gastroenterology, The Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Shigang Yin
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jianhua Peng
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yong Jiang
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Shichun Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, Key Laboratory of Digital Hepatobiliary Surgery, Institute of Hepatobiliary Surgery of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Hongkui Deng
- MOE Engineering Research Center of Regenerative Medicine, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100191, China.
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
3
|
Ruppert PMM, Kersten S. Mechanisms of hepatic fatty acid oxidation and ketogenesis during fasting. Trends Endocrinol Metab 2024; 35:107-124. [PMID: 37940485 DOI: 10.1016/j.tem.2023.10.002] [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: 08/31/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 11/10/2023]
Abstract
Fasting is part of many weight management and health-boosting regimens. Fasting causes substantial metabolic adaptations in the liver that include the stimulation of fatty acid oxidation and ketogenesis. The induction of fatty acid oxidation and ketogenesis during fasting is mainly driven by interrelated changes in plasma levels of various hormones and an increase in plasma nonesterified fatty acid (NEFA) levels and is mediated transcriptionally by the peroxisome proliferator-activated receptor (PPAR)α, supported by CREB3L3 (cyclic AMP-responsive element-binding protein 3 like 3). Compared with men, women exhibit higher ketone levels during fasting, likely due to higher NEFA availability, suggesting that the metabolic response to fasting shows sexual dimorphism. Here, we synthesize the current molecular knowledge on the impact of fasting on hepatic fatty acid oxidation and ketogenesis.
Collapse
Affiliation(s)
- Philip M M Ruppert
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5000 C Odense, Denmark
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708 WE Wageningen, The Netherlands; Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
4
|
Egami S, Watanabe T, Fukushima-Nomura A, Nomura H, Takahashi H, Yamagami J, Ohara O, Amagai M. Desmoglein-Specific B-Cell-Targeted Single-Cell Analysis Revealing Unique Gene Regulation in Patients with Pemphigus. J Invest Dermatol 2023; 143:1919-1928.e16. [PMID: 36997112 DOI: 10.1016/j.jid.2023.03.1661] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/31/2023]
Abstract
Autoreactive B cells are assumed to play a critical role in pemphigus; however, the characteristics of these cells are not yet fully understood. In this study, 23 pemphigus vulgaris or pemphigus foliaceus samples were used to isolate circulating desmoglein (DSG)-specific B cells. Transcriptome analysis of the samples was performed at the single-cell level to detect genes involved in disease activity. DSG1- or DSG3-specific B cells from three patients' differentially expressed genes related to T cell costimulation (CD137L) as well as B-cell differentiation (CD9, BATF, TIMP1) and inflammation (S100A8, S100A9, CCR3), compared with nonspecific B cells from the same patients. When the DSG1-specific B cells before and after treatment transcriptomes of the patient with pemphigus foliaceus were compared, there were changes in several B-cell activation pathways not detected in non-DSG1-specific B cells. This study clarifies the transcriptomic profile of autoreactive B cells in patients with pemphigus and documents the gene expression related to disease activity. Our approach can be applied to other autoimmune diseases and has the potential for future detection of disease-specific autoimmune cells.
Collapse
Affiliation(s)
- Shohei Egami
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| | - Takashi Watanabe
- Laboratory for integrative genomics, RIKEN Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| | | | - Hisashi Nomura
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Jun Yamagami
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Osamu Ohara
- Laboratory for integrative genomics, RIKEN Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan.
| |
Collapse
|
5
|
Zhou Z, Zhang A, Liu X, Yang Y, Zhao R, Jia Y. m 6A-Mediated PPARA Translational Suppression Contributes to Corticosterone-Induced Visceral Fat Deposition in Chickens. Int J Mol Sci 2022; 23:ijms232415761. [PMID: 36555401 PMCID: PMC9779672 DOI: 10.3390/ijms232415761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Excess fat deposition in broilers leads to great economic losses and is harmful to consumers' health. Chronic stress in the life cycle of chickens could be an important trigger. However, the underlying mechanisms are still unclear. In this study, 30-day-old chickens were subcutaneously injected with 2 mg/kg corticosterone (CORT) twice a day for 14 days to simulate long-term stress. It was shown that chronic CORT exposure significantly increased plasma triglyceride concentrations and enlarged the adipocyte sizes in chickens. Meanwhile, chronic CORT administration significantly enlarged the adipocyte sizes, increased the protein contents of FASN and decreased HSL, ATGL, Beclin1 and PPARA protein levels. Moreover, global m6A methylations were significantly reduced and accompanied by downregulated METTL3 and YTHDF2 protein expression by CORT treatment. Interestingly, the significant differences of site-specific m6A demethylation were observed in exon7 of PPARA mRNA. Additionally, a mutation of the m6A site in the PPARA gene fused GFP and revealed that demethylated RRACH in PPARA CDS impaired protein translation in vitro. In conclusion, these results indicated that m6A-mediated PPARA translational suppression contributes to CORT-induced visceral fat deposition in chickens, which may provide a new target for the treatment of Cushing's syndrome.
Collapse
Affiliation(s)
- Zixuan Zhou
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Aijia Zhang
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xinyi Liu
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yang Yang
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing 210095, China
| | - Yimin Jia
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing 210095, China
- Correspondence: ; Tel.: +86-2584396413; Fax: +86-2584398669
| |
Collapse
|
6
|
Gao H, Li Y, Chen X. Interactions between nuclear receptors glucocorticoid receptor α and peroxisome proliferator-activated receptor α form a negative feedback loop. Rev Endocr Metab Disord 2022; 23:893-903. [PMID: 35476174 DOI: 10.1007/s11154-022-09725-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2022] [Indexed: 02/05/2023]
Abstract
Both nuclear receptors glucocorticoid receptor α (GRα) and peroxisome proliferator-activated receptor α (PPARα) are involved in energy and lipid metabolism, and possess anti-inflammation effects. Previous studies indicate that a regulatory loop may exist between them. In vivo and in vitro studies showed that glucocorticoids stimulate hepatic PPARα expression via GRα at the transcriptional level. This stimulation of PPARα by GRα has physiological relevance and PPARα is involved in many glucocorticoid-induced pathophysiological processes, including gluconeogenesis and ketogenesis during fasting, insulin resistance, hypertension and anti-inflammatory effects. PPARα also synergizes with GRα to promote erythroid progenitor self-renewal. As the feedback, PPARα inhibits glucocorticoid actions at pre-receptor and receptor levels. PPARα decreases glucocorticoid production through inhibiting the expression and activity of type-1 11β-hydroxysteroid dehydrogenase, which converts inactive glucocorticoids to active glucocorticoids at local tissues, and also down-regulates hepatic GRα expression, thus forming a complete and negative feedback loop. This negative feedback loop sheds light on prospective multi-drug therapeutic treatments in inflammatory diseases through a combination of glucocorticoids and PPARα agonists. This combination may potentially enhance the anti-inflammatory effects while alleviating side effects on glucose and lipid metabolism due to GRα activation. More investigations are needed to clarify the underlying mechanism and the relevant physiological or pathological significance of this regulatory loop.
Collapse
Affiliation(s)
- Hongjiao Gao
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Zunyi Medical University (the First People's Hospital of Zunyi), 563002, Zunyi, China
| | - Yujue Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Xiang Chen
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
7
|
Seneff S, Nigh G, Kyriakopoulos AM, McCullough PA. Innate immune suppression by SARS-CoV-2 mRNA vaccinations: The role of G-quadruplexes, exosomes, and MicroRNAs. Food Chem Toxicol 2022; 164:113008. [PMID: 35436552 PMCID: PMC9012513 DOI: 10.1016/j.fct.2022.113008] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022]
Abstract
The mRNA SARS-CoV-2 vaccines were brought to market in response to the public health crises of Covid-19. The utilization of mRNA vaccines in the context of infectious disease has no precedent. The many alterations in the vaccine mRNA hide the mRNA from cellular defenses and promote a longer biological half-life and high production of spike protein. However, the immune response to the vaccine is very different from that to a SARS-CoV-2 infection. In this paper, we present evidence that vaccination induces a profound impairment in type I interferon signaling, which has diverse adverse consequences to human health. Immune cells that have taken up the vaccine nanoparticles release into circulation large numbers of exosomes containing spike protein along with critical microRNAs that induce a signaling response in recipient cells at distant sites. We also identify potential profound disturbances in regulatory control of protein synthesis and cancer surveillance. These disturbances potentially have a causal link to neurodegenerative disease, myocarditis, immune thrombocytopenia, Bell's palsy, liver disease, impaired adaptive immunity, impaired DNA damage response and tumorigenesis. We show evidence from the VAERS database supporting our hypothesis. We believe a comprehensive risk/benefit assessment of the mRNA vaccines questions them as positive contributors to public health.
Collapse
Affiliation(s)
- Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA, 02139.
| | - Greg Nigh
- Immersion Health, Portland, OR, 97214, USA.
| | - Anthony M Kyriakopoulos
- Research and Development, Nasco AD Biotechnology Laboratory, Department of Research and Development, Sachtouri 11, 18536, Piraeus, Greece.
| | | |
Collapse
|
8
|
Morikawa T, Fukami T, Gotoh-Saito S, Nakano M, Nakajima M. PPARα regulates the expression of human arylacetamide deacetylase involved in drug hydrolysis and lipid metabolism. Biochem Pharmacol 2022; 199:115010. [DOI: 10.1016/j.bcp.2022.115010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 12/01/2022]
|
9
|
Ma DD, Jiang YX, Zhang JG, Fang GZ, Huang GY, Shi WJ, Ying GG. Transgenerational effects of androstadienedione and androstenedione at environmentally relevant concentrations in zebrafish (Danio rerio). JOURNAL OF HAZARDOUS MATERIALS 2022; 423:127261. [PMID: 34844370 DOI: 10.1016/j.jhazmat.2021.127261] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 06/13/2023]
Abstract
Androgens androstadienedione (ADD) and androstenedione (AED) are predominant steroid hormones in surface water, and can disrupt the endocrine system in fish. However, little is known about the transgenerational effects of ADD and AED in fish. In the present study, F0 generation was exposed to ADD and AED from 21 to 144 days post-fertilization (dpf) at nominal concentrations of 5 (L), 50 (M) and 500 (H) ng L-1, and F1 generation was domesticated in clear water for 144 dpf. The sex ratio, histology and transcription in F0 and F1 generations were examined. In the F0 generation, ADD and AED tended to be estrogenic in zebrafish, resulting in female biased zebrafish populations. In the F1 generation, ADD at the H level caused 63.5% females, while AED at the H level resulted in 78.7% males. In brain, ADD and AED had similar effects on circadian rhythm in the F0 and F1 generations. In the F1 eleutheroembryos, transcriptomic analysis indicated that neuromast hair cell related biological processes (BPs) were overlapped in the ADD and AED groups. Taken together, ADD and AED at environmentally relevant concentrations had transgenerational effects on sex differentiation and transcription in zebrafish.
Collapse
Affiliation(s)
- Dong-Dong Ma
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Yu-Xia Jiang
- Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510535, China
| | - Jin-Ge Zhang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Gui-Zhen Fang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Guo-Yong Huang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Wen-Jun Shi
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China.
| | - Guang-Guo Ying
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China.
| |
Collapse
|
10
|
Chakraborty P, Chattarji S, Jeanneteau F. A salience hypothesis of stress in PTSD. Eur J Neurosci 2021; 54:8029-8051. [PMID: 34766390 DOI: 10.1111/ejn.15526] [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: 07/10/2021] [Revised: 09/13/2021] [Accepted: 10/30/2021] [Indexed: 11/30/2022]
Abstract
Attention to key features of contexts and things is a necessary tool for all organisms. Detecting these salient features of cues, or simply, salience, can also be affected by exposure to traumatic stress, as has been widely reported in individuals suffering from post-traumatic stress disorder (PTSD). Interestingly, similar observations have been robustly replicated across many animal models of stress as well. By using evidence from such rodent stress paradigms, in the present review, we explore PTSD through the lens of salience processing. In this context, we propose that interaction between the neurotrophin brain-derived neurotrophic factor (BDNF) and glucocorticoids determines the long lasting cellular and behavioural consequences of stress salience. We also describe the dual effect of glucocorticoid therapy in the amelioration of PTSD symptoms. Finally, by integrating in vivo observations at multiple scales of plasticity, we propose a unifying hypothesis that pivots on a crucial role of glucocorticoid signalling in dynamically orchestrating stress salience.
Collapse
Affiliation(s)
- Prabahan Chakraborty
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France.,Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India
| | - Sumantra Chattarji
- Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Freddy Jeanneteau
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France
| |
Collapse
|
11
|
The Glitazars Paradox: Cardiotoxicity of the Metabolically Beneficial Dual PPARα and PPARγ Activation. J Cardiovasc Pharmacol 2021; 76:514-526. [PMID: 33165133 DOI: 10.1097/fjc.0000000000000891] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common complications in patients with type-2 diabetes are hyperglycemia and hyperlipidemia that can lead to cardiovascular disease. Alleviation of these complications constitutes the major therapeutic approach for the treatment of diabetes mellitus. Agonists of peroxisome proliferator-activated receptor (PPAR) alpha and PPARγ are used for the treatment of hyperlipidemia and hyperglycemia, respectively. PPARs belong to the nuclear receptors superfamily and regulate fatty acid metabolism. PPARα ligands, such as fibrates, reduce circulating triglyceride levels, and PPARγ agonists, such as thiazolidinediones, improve insulin sensitivity. Dual-PPARα/γ agonists (glitazars) were developed to combine the beneficial effects of PPARα and PPARγ agonism. Although they improved metabolic parameters, they paradoxically aggravated congestive heart failure in patients with type-2 diabetes via mechanisms that remain elusive. Many of the glitazars, such as muraglitazar, tesaglitazar, and aleglitazar, were abandoned in phase-III clinical trials. The objective of this review article pertains to the understanding of how combined PPARα and PPARγ activation, which successfully targets the major complications of diabetes, causes cardiac dysfunction. Furthermore, it aims to suggest interventions that will maintain the beneficial effects of dual PPARα/γ agonism and alleviate adverse cardiac outcomes in diabetes.
Collapse
|
12
|
Rasineni K, Jordan CW, Thomes PG, Kubik JL, Staab EM, Sweeney SA, Talmon GA, Donohue TM, McNiven MA, Kharbanda KK, Casey CA. Contrasting Effects of Fasting on Liver-Adipose Axis in Alcohol-Associated and Non-alcoholic Fatty Liver. Front Physiol 2021; 12:625352. [PMID: 33746771 PMCID: PMC7966527 DOI: 10.3389/fphys.2021.625352] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/02/2021] [Indexed: 01/15/2023] Open
Abstract
Background: Fatty liver, a major health problem worldwide, is the earliest pathological change in the progression of alcohol-associated (AFL) and non-alcoholic fatty liver disease (NAFL). Though the causes of AFL and NAFL differ, both share similar histological and some common pathophysiological characteristics. In this study, we sought to examine mechanisms responsible for lipid dynamics in liver and adipose tissue in the setting of AFL and NAFL in response to 48 h of fasting. Methods: Male rats were fed Lieber-DeCarli liquid control or alcohol-containing diet (AFL model), chow or high-fat pellet diet (NAFL model). After 6-8 weeks of feeding, half of the rats from each group were fasted for 48 h while the other half remained on their respective diets. Following sacrifice, blood, adipose, and the liver were collected for analysis. Results: Though rats fed AFL and NAFL diets both showed fatty liver, the physiological mechanisms involved in the development of each was different. Here, we show that increased hepatic de novo fatty acid synthesis, increased uptake of adipose-derived free fatty acids, and impaired triglyceride breakdown contribute to the development of AFL. In the case of NAFL, however, increased dietary fatty acid uptake is the major contributor to hepatic steatosis. Likewise, the response to starvation in the two fatty liver disease models also varied. While there was a decrease in hepatic steatosis after fasting in ethanol-fed rats, the control, chow and high-fat diet-fed rats showed higher levels of hepatic steatosis than pair-fed counterparts. This diverse response was a result of increased adipose lipolysis in all experimental groups except fasted ethanol-fed rats. Conclusion: Even though AFL and NAFL are nearly histologically indistinguishable, the physiological mechanisms that cause hepatic fat accumulation are different as are their responses to starvation.
Collapse
Affiliation(s)
- Karuna Rasineni
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Clayton W. Jordan
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Paul G. Thomes
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Jacy L. Kubik
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Elizabeth M. Staab
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sarah A. Sweeney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Geoffrey A. Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Terrence M. Donohue
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Mayo Clinic, Rochester, MN, United States
| | - Kusum K. Kharbanda
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Carol A. Casey
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
13
|
Hsu CN, Jen CY, Chen YH, Peng SY, Wu SC, Yao CL. Glucocorticoid transiently upregulates mitochondrial biogenesis in the osteoblast. CHINESE J PHYSIOL 2021; 63:286-293. [PMID: 33380613 DOI: 10.4103/cjp.cjp_51_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Glucocorticoid (GC)-induced bone loss is the most prevalent form of secondary osteoporosis. Previous studies demonstrated that long-term incubation of dexamethasone (DEX) induced oxidative stress and mitochondrial dysfunctions, consequently leading to apoptosis of differentiated osteoblasts. This DEX-induced cell death might be the main causes of bone loss. We previously described that DEX induced biphasic mitochondrial alternations. As GC affects mitochondrial physiology through several different possible routes, the short-term and long-term effects of GC treatment on mitochondria in the osteoblast have not been carefully characterized. Here, we examined the expression levels of genes that are associated with mitochondrial functions at several different time points after incubation with DEX. Mitochondrial biogenesis-mediated genes nuclear respiratory factor 1 (Nrf1) and Nrf2 were upregulated after 4-h incubation, and then declined after 24-h incubation, suggesting that mitochondrial biogenesis were transiently upregulated by DEX. In contrast, mitochondrial fusion gene optic atrophy 1 (Opa1) and mitofusin 2 (Mfn2) started to be elevated as the biogenesis started to decrease. Finally, the mitochondrial fission increased and apoptosis becomes prominent. Agree with the mitochondrial biphasic alterations hypothesis, the results suggested an early increase of mitochondrial activities and biogenesis upon DEX stimulation to the osteoblasts. The oxidative phosphorylation and inducible nitric oxide synthase levels increased results in oxidative stress accumulation, leading to mitochondrial fusion, and subsequently fission and triggering the apoptosis. Our results indicated that the primary effects of GC on mitochondria are promoting their functions and biogenesis. Mitochondrial breakdown and the activation of the apoptotic pathways appeared to be the secondary effect after long-term treatment.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Orthopedics, Taoyuan General Hospital, Ministry of Health and Welfare; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, Taiwan
| | - Chih-Yuan Jen
- Department of Surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Yu-Hsu Chen
- Department of Orthopedics, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan; Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Shao-Yu Peng
- Department of Animal Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Shinn-Chih Wu
- Department of Animal Science and Technology; Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chao-Ling Yao
- Graduate School of Biotechnology and Bioengineering; Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan, Taiwan
| |
Collapse
|
14
|
Hara S, Furukawa F, Mukai K, Yazawa T, Kitano T. Peroxisome proliferator-activated receptor alpha is involved in the temperature-induced sex differentiation of a vertebrate. Sci Rep 2020; 10:11672. [PMID: 32669596 PMCID: PMC7363821 DOI: 10.1038/s41598-020-68594-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/30/2020] [Indexed: 11/09/2022] Open
Abstract
Medaka (Oryzias latipes) is a teleost fish with an XX/XY sex determination system, similar to that of mammals. However, under high temperature conditions, XX medaka is masculinised by elevation of cortisol, the major teleost glucocorticoid. In this study, to identify novel factors in the gonads acting downstream from cortisol during sexual differentiation, we performed RNA sequencing (RNA-seq) analysis using the gonadal regions of larvae reared at normal temperature with and without cortisol, and at high temperature. The RNA-seq and real-time PCR analyses showed that expression of some peroxisome proliferator-activated receptor α (PPARα) signalling-targeted genes was increased by cortisol. PPARα agonist treatment induced masculinisation of XX medaka in some cases, and co-treatment of the agonist with cortisol further induced masculinisation, whereas treatment of pparaa knockout medaka with cortisol or the agonist did not induce masculinisation. This study provides the first evidence that PPARα is involved in environmental sex determination in vertebrates.
Collapse
Affiliation(s)
- Seiji Hara
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto, 860-8555, Japan
| | - Fumiya Furukawa
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto, 860-8555, Japan
| | - Koki Mukai
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto, 860-8555, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| | - Takeshi Kitano
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto, 860-8555, Japan.
| |
Collapse
|
15
|
De Bosscher K, Desmet SJ, Clarisse D, Estébanez-Perpiña E, Brunsveld L. Nuclear receptor crosstalk - defining the mechanisms for therapeutic innovation. Nat Rev Endocrinol 2020; 16:363-377. [PMID: 32303708 DOI: 10.1038/s41574-020-0349-5] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
Nuclear receptor crosstalk can be defined as the interplay between different nuclear receptors or between their overlapping signalling pathways. A subset of nuclear receptors (such as PPARs and RARs) engage in the formation of well-characterized 'typical' heterodimers with RXR. 'Atypical' heterodimers (such as GR with PPARs, or PPAR with ERR) might form a novel class of physical complexes that might be more transient in nature. These heterodimers might harbour strong transcriptional flexibility, with no strict need for DNA binding of both partners. Direct crosstalk could stem from a pairwise physical association between atypical nuclear receptor heterodimers, either via pre-existing interaction pairs or via interactions that are newly induced with small molecules; such crosstalk might constitute an uncharted space to target nuclear receptor physiological and/or pathophysiological actions. In this Review, we discuss the emerging aspects of crosstalk in the nuclear receptor field and present various mechanistic crosstalk modes with examples that support applicability of the atypical heterodimer concept. Stabilization or disruption, in a context-dependent or cell type-dependent manner, of these more transient heterodimers is expected to fuel unprecedented translational approaches to yield novel therapeutic agents to treat major human diseases with higher precision.
Collapse
Affiliation(s)
- Karolien De Bosscher
- Translational Nuclear Receptor Research, VIB Center for Medical Biotechnology, UGent Department of Biomolecular Medicine, Gent, Belgium.
| | - Sofie J Desmet
- Translational Nuclear Receptor Research, VIB Center for Medical Biotechnology, UGent Department of Biomolecular Medicine, Gent, Belgium
| | - Dorien Clarisse
- Translational Nuclear Receptor Research, VIB Center for Medical Biotechnology, UGent Department of Biomolecular Medicine, Gent, Belgium
| | - Eva Estébanez-Perpiña
- Laboratory of Structural Biology, Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine (IBUB) of the University of Barcelona (UB), Barcelona, Spain
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Eindhoven, Netherlands
| |
Collapse
|
16
|
Oh HYP, Ellero-Simatos S, Manickam R, Tan NS, Guillou H, Wahli W. Depletion of Gram-Positive Bacteria Impacts Hepatic Biological Functions During the Light Phase. Int J Mol Sci 2019; 20:E812. [PMID: 30769793 PMCID: PMC6412208 DOI: 10.3390/ijms20040812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
Living organisms display internal biological rhythms, which are an evolutionarily conserved adaptation to the environment that drives their rhythmic behavioral and physiological activities. The gut microbiota has been proposed, in association with diet, to regulate the intestinal peripheral clock. However, the effect of gut dysbiosis on liver remains elusive, despite that germfree mice show alterations in liver metabolic functions and the hepatic daily rhythm. We analyzed whether the disruption of gut microbial populations with various antibiotics would differentially impact liver functions in mice. Our results support the notion of an impact on the hepatic biological rhythm by gram-positive bacteria. In addition, we provide evidence for differential roles of gut microbiota spectra in xenobiotic metabolism that could protect against the harmful pharmacological effects of drugs. Our results underscore a possible link between liver cell proliferation and gram-positive bacteria.
Collapse
Affiliation(s)
- Hui Yun Penny Oh
- Interdisciplinary Graduate School, NTU Institute for Health Technologies, Nanyang Technological University Singapore, 50 Nanyang Avenue, Singapore 639798, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| | | | - Ravikumar Manickam
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Hervé Guillou
- INRA UMR1331, ToxAlim, 180 Chemin de Tournefeuille, 31300 Toulouse, France.
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
- INRA UMR1331, ToxAlim, 180 Chemin de Tournefeuille, 31300 Toulouse, France.
- Center for Integrative Genomics, University of Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
17
|
Schmidt M, Lax E, Zhou R, Cheishvili D, Ruder AM, Ludiro A, Lapert F, Macedo da Cruz A, Sandrini P, Calzoni T, Vaisheva F, Brandwein C, Luoni A, Massart R, Lanfumey L, Riva MA, Deuschle M, Gass P, Szyf M. Fetal glucocorticoid receptor (Nr3c1) deficiency alters the landscape of DNA methylation of murine placenta in a sex-dependent manner and is associated to anxiety-like behavior in adulthood. Transl Psychiatry 2019; 9:23. [PMID: 30655507 PMCID: PMC6336883 DOI: 10.1038/s41398-018-0348-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 11/13/2018] [Indexed: 12/28/2022] Open
Abstract
Prenatal stress defines long-term phenotypes through epigenetic programming of the offspring. These effects are potentially mediated by glucocorticoid release and by sex. We hypothesized that the glucocorticoid receptor (Gr, Nr3c1) fashions the DNA methylation profile of offspring. Consistent with this hypothesis, fetal Nr3c1 heterozygosity leads to altered DNA methylation landscape in fetal placenta in a sex-specific manner. There was a significant overlap of differentially methylated genes in fetal placenta and adult frontal cortex in Nr3c1 heterozygotes. Phenotypically, Nr3c1 heterozygotes show significantly more anxiety-like behavior than wildtype. DNA methylation status of fetal placental tissue is significantly correlated with anxiety-like behavior of the same animals in adulthood. Thus, placental DNA methylation might predict behavioral phenotypes in adulthood. Our data supports the hypothesis that Nr3c1 influences DNA methylation at birth and that DNA methylation in placenta correlates with adult frontal cortex DNA methylation and anxiety-like phenotypes.
Collapse
Affiliation(s)
- Michaela Schmidt
- Central Institute of Mental Health Mannheim (ZI), Medical Faculty of Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany.
| | - Elad Lax
- 0000 0004 1936 8649grid.14709.3bDepartment of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6 Canada ,0000 0004 1936 8649grid.14709.3bSackler Program for Epigenetics and Psychobiology, McGill University, Montreal, QC H3G 1Y6 Canada
| | - Rudy Zhou
- 0000 0004 1936 8649grid.14709.3bDepartment of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6 Canada
| | - David Cheishvili
- 0000 0004 1936 8649grid.14709.3bDepartment of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6 Canada ,0000 0004 1936 8649grid.14709.3bSackler Program for Epigenetics and Psychobiology, McGill University, Montreal, QC H3G 1Y6 Canada
| | - Arne Mathias Ruder
- 0000 0001 2190 4373grid.7700.0Central Institute of Mental Health Mannheim (ZI), Medical Faculty of Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Alessia Ludiro
- 0000 0004 1757 2822grid.4708.bDepartment of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, I-20133 Milan, Italy
| | - Florian Lapert
- 0000 0001 2190 4373grid.7700.0Central Institute of Mental Health Mannheim (ZI), Medical Faculty of Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Anna Macedo da Cruz
- 0000 0001 2190 4373grid.7700.0Central Institute of Mental Health Mannheim (ZI), Medical Faculty of Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Paolo Sandrini
- 0000 0004 1757 2822grid.4708.bDepartment of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, I-20133 Milan, Italy
| | - Teresa Calzoni
- 0000 0004 1757 2822grid.4708.bDepartment of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, I-20133 Milan, Italy
| | - Farida Vaisheva
- 0000 0004 1936 8649grid.14709.3bDepartment of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6 Canada
| | - Christiane Brandwein
- 0000 0001 2190 4373grid.7700.0Central Institute of Mental Health Mannheim (ZI), Medical Faculty of Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Alessia Luoni
- 0000 0004 1757 2822grid.4708.bDepartment of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, I-20133 Milan, Italy
| | - Renaud Massart
- 0000 0004 1936 8649grid.14709.3bSackler Program for Epigenetics and Psychobiology, McGill University, Montreal, QC H3G 1Y6 Canada ,0000 0004 0638 6979grid.417896.5Inserm, U894, Centre de Psychiatrie et Neurosciences, 75014 Paris, France
| | - Laurence Lanfumey
- 0000 0004 0638 6979grid.417896.5Inserm, U894, Centre de Psychiatrie et Neurosciences, 75014 Paris, France ,0000 0001 2188 0914grid.10992.33Université Paris Descartes, UMRS894, 75014 Paris, France
| | - Marco Andrea Riva
- 0000 0004 1757 2822grid.4708.bDepartment of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, I-20133 Milan, Italy
| | - Michael Deuschle
- 0000 0001 2190 4373grid.7700.0Central Institute of Mental Health Mannheim (ZI), Medical Faculty of Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Peter Gass
- 0000 0001 2190 4373grid.7700.0Central Institute of Mental Health Mannheim (ZI), Medical Faculty of Mannheim, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Moshe Szyf
- 0000 0004 1936 8649grid.14709.3bDepartment of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6 Canada ,0000 0004 1936 8649grid.14709.3bSackler Program for Epigenetics and Psychobiology, McGill University, Montreal, QC H3G 1Y6 Canada
| |
Collapse
|
18
|
Li S, Lan Y, Wu W, Duan X, Kong Z, Wu W, Zeng G. Peroxisome proliferator‐activated receptor γ modulates renal crystal retention associated with high oxalate concentration by regulating tubular epithelial cellular transdifferentiation. J Cell Physiol 2018; 234:2837-2850. [PMID: 30317563 DOI: 10.1002/jcp.27102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 06/29/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Shujue Li
- Department of UrologyMinimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou China
- Guangdong Key Laboratory of UrologyGuangzhou Institute of UrologyGuangzhou China
| | - Yu Lan
- Department of UrologyMinimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou China
- Guangdong Key Laboratory of UrologyGuangzhou Institute of UrologyGuangzhou China
| | - Wenzheng Wu
- Department of UrologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou China
| | - Xiaolu Duan
- Department of UrologyMinimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou China
- Guangdong Key Laboratory of UrologyGuangzhou Institute of UrologyGuangzhou China
| | - Zhenzhen Kong
- Department of UrologyMinimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou China
- Guangdong Key Laboratory of UrologyGuangzhou Institute of UrologyGuangzhou China
| | - Wenqi Wu
- Department of UrologyMinimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou China
- Guangdong Key Laboratory of UrologyGuangzhou Institute of UrologyGuangzhou China
| | - Guohua Zeng
- Department of UrologyMinimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou China
- Guangdong Key Laboratory of UrologyGuangzhou Institute of UrologyGuangzhou China
| |
Collapse
|
19
|
Bougarne N, Weyers B, Desmet SJ, Deckers J, Ray DW, Staels B, De Bosscher K. Molecular Actions of PPARα in Lipid Metabolism and Inflammation. Endocr Rev 2018; 39:760-802. [PMID: 30020428 DOI: 10.1210/er.2018-00064] [Citation(s) in RCA: 440] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a nuclear receptor of clinical interest as a drug target in various metabolic disorders. PPARα also exhibits marked anti-inflammatory capacities. The first-generation PPARα agonists, the fibrates, have however been hampered by drug-drug interaction issues, statin drop-in, and ill-designed cardiovascular intervention trials. Notwithstanding, understanding the molecular mechanisms by which PPARα works will enable control of its activities as a drug target for metabolic diseases with an underlying inflammatory component. Given its role in reshaping the immune system, the full potential of this nuclear receptor subtype as a versatile drug target with high plasticity becomes increasingly clear, and a novel generation of agonists may pave the way for novel fields of applications.
Collapse
Affiliation(s)
- Nadia Bougarne
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Basiel Weyers
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Sofie J Desmet
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Julie Deckers
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation, VIB Center for Inflammation Research, Ghent (Zwijnaarde), Belgium
| | - David W Ray
- Division of Metabolism and Endocrinology, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Bart Staels
- Université de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
- INSERM, U1011, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Karolien De Bosscher
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| |
Collapse
|
20
|
Cui H, Zheng M, Zhao G, Liu R, Wen J. Identification of differentially expressed genes and pathways for intramuscular fat metabolism between breast and thigh tissues of chickens. BMC Genomics 2018; 19:55. [PMID: 29338766 PMCID: PMC5771206 DOI: 10.1186/s12864-017-4292-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 11/10/2017] [Indexed: 11/15/2022] Open
Abstract
Background Intramuscular fat (IMF) is one of the important factors influencing meat quality, however, for chickens, the molecular regulatory mechanisms underlying this trait have not yet been clear. In this study, a systematic identification of differentially expressed genes (DEGs) and molecular regulatory mechanism related to IMF metabolism between Beijing-you chicken breast and thigh at 42 and 90 days of age was performed. Results IMF contents, Gene Ontology (GO) terms, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were analyzed, The results showed that both IMF contents in breast at 42 and 90 d were significantly lower (P < 0.05 or P < 0.01) than those in thigh. By microarray, 515 common known DEGs and 36 DEGs related to IMF metabolism were identified between the breast and thigh at 42 and 90 d. Compared to thigh, the expression levels of PPARG had significantly down-regulated (P < 0.01) in breast, but the expression levels of RXRA and CEBPB had significantly up-regulated (P < 0.01). However, the expression levels of LPL, FABP4, THRSP, RBP7, LDLR, FABP3, CPT2 and PPARGC1A had significantly down-regulated in breast (P < 0.01), supporting that PPARG and its down-stream genes had the important regulatory function to IMF deposition. In addition, based on of DEGs, KEGG analysis revealed that PPAR signaling pathway and cell junction-related pathways (focal adhesion and ECM-receptor interaction, which play a prominent role in maintaining the integrity of tissues), might contribute to the IMF metabolism in chicken. Conclusions Our data had screened the potential candidate genes associated with chicken IMF metabolism, and imply that IMF metabolism in chicken is regulated and mediated not only by related functional genes and PPAR pathway, but also by others involved in cell junctions. These findings establish the groundwork and provide new clues for deciphering the molecular mechanisms underlying IMF deposition in poultry. Further studies at the translational and posttranslational level are now required to validate the genes and pathways identified here. Electronic supplementary material The online version of this article (10.1186/s12864-017-4292-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huanxian Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Maiqing Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Guiping Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Ranran Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Jie Wen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China. .,State Key Laboratory of Animal Nutrition, Beijing, 100193, China.
| |
Collapse
|
21
|
Tanaka H, Shimizu N, Yoshikawa N. Role of skeletal muscle glucocorticoid receptor in systemic energy homeostasis. Exp Cell Res 2017; 360:24-26. [DOI: 10.1016/j.yexcr.2017.03.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/22/2017] [Indexed: 12/16/2022]
|
22
|
Gong X, Marisiddaiah R, Rubin LP. Inhibition of pulmonary β-carotene 15, 15'-oxygenase expression by glucocorticoid involves PPARα. PLoS One 2017; 12:e0181466. [PMID: 28732066 PMCID: PMC5521778 DOI: 10.1371/journal.pone.0181466] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/30/2017] [Indexed: 01/19/2023] Open
Abstract
β-carotene 15,15'-oxygenase (BCO1) catalyzes the first step in the conversion of dietary provitamin A carotenoids to vitamin A. This enzyme is expressed in a variety of developing and adult tissues, suggesting that its activity may regulate local retinoid synthesis. Vitamin A and related compounds (retinoids) are critical regulators of lung epithelial development, integrity, and injury repair. A balance between the actions of retinoids and glucocorticoids (GCs) promotes normal lung development and, in particular, alveolarization. Alterations in this balance, including vitamin A deficiency and GC excess, contribute to the development of chronic lung disorders. Consequently, we investigated if GCs counteract retinoid effects in alveolar epithelial cells by mechanisms involving BCO1-dependent local vitamin A metabolism. We demonstrate that BCO1 is expressed in human fetal lung tissue and human alveolar epithelial-like A549 cells. Our results indicate A549 cells metabolize β-carotene to retinal and retinoic acid (RA). GCs exposure using dexamethasone (DEX) decreases BCO1 mRNA and protein levels in A549 cells and reduces BCO1 promoter activity via inhibiting peroxisome proliferator-activated receptor γ (PPARγ) DNA binding. DEX also induces expression of PPARα, which in turn most likely causes a decrease in PPARγ/RXRα heterodimer binding to the bco1 gene promoter and consequent inhibition of bco1 gene expression. PPARα knockdown with siRNA abolishes DEX-induced suppression of BCO1 expression, confirming the requirement for PPARα in this DEX-mediated BCO1 mechanism. Taken together, these findings provide the first evidence that GCs regulate vitamin A (retinoid) signaling via inhibition of bco1 gene expression in a PPARα-dependent manner. These results explicate novel aspects of local GC:retinoid interactions that may contribute to alveolar tissue remodeling in chronic lung diseases that affect children and, possibly, adults.
Collapse
Affiliation(s)
- Xiaoming Gong
- Department of Pediatrics, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, United States of America
- * E-mail: (XG); (LPR)
| | - Raju Marisiddaiah
- All Children’s Research Institute, St. Petersburg, Florida, United States of America
| | - Lewis P. Rubin
- Department of Pediatrics, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, United States of America
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, United States of America
- * E-mail: (XG); (LPR)
| |
Collapse
|
23
|
Goldstein I, Baek S, Presman DM, Paakinaho V, Swinstead EE, Hager GL. Transcription factor assisted loading and enhancer dynamics dictate the hepatic fasting response. Genome Res 2016; 27:427-439. [PMID: 28031249 PMCID: PMC5340970 DOI: 10.1101/gr.212175.116] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/21/2016] [Indexed: 02/03/2023]
Abstract
Fasting elicits transcriptional programs in hepatocytes leading to glucose and ketone production. This transcriptional program is regulated by many transcription factors (TFs). To understand how this complex network regulates the metabolic response to fasting, we aimed at isolating the enhancers and TFs dictating it. Measuring chromatin accessibility revealed that fasting massively reorganizes liver chromatin, exposing numerous fasting-induced enhancers. By utilizing computational methods in combination with dissecting enhancer features and TF cistromes, we implicated four key TFs regulating the fasting response: glucocorticoid receptor (GR), cAMP responsive element binding protein 1 (CREB1), peroxisome proliferator activated receptor alpha (PPARA), and CCAAT/enhancer binding protein beta (CEBPB). These TFs regulate fuel production by two distinctly operating modules, each controlling a separate metabolic pathway. The gluconeogenic module operates through assisted loading, whereby GR doubles the number of sites occupied by CREB1 as well as enhances CREB1 binding intensity and increases accessibility of CREB1 binding sites. Importantly, this GR-assisted CREB1 binding was enhancer-selective and did not affect all CREB1-bound enhancers. Single-molecule tracking revealed that GR increases the number and DNA residence time of a portion of chromatin-bound CREB1 molecules. These events collectively result in rapid synergistic gene expression and higher hepatic glucose production. Conversely, the ketogenic module operates via a GR-induced TF cascade, whereby PPARA levels are increased following GR activation, facilitating gradual enhancer maturation next to PPARA target genes and delayed ketogenic gene expression. Our findings reveal a complex network of enhancers and TFs that dynamically cooperate to restore homeostasis upon fasting.
Collapse
Affiliation(s)
- Ido Goldstein
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Diego M Presman
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ville Paakinaho
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Erin E Swinstead
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
24
|
Katsumura M, Takagi S, Oya H, Tamura S, Saneyasu T, Honda K, Kamisoyama H. Effects of dietary heme iron and exercise training on abdominal fat accumulation and lipid metabolism in high-fat diet-fed mice. Anim Sci J 2016; 88:1100-1106. [DOI: 10.1111/asj.12734] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/29/2016] [Accepted: 09/14/2016] [Indexed: 11/30/2022]
Affiliation(s)
| | - Shoko Takagi
- Graduate School of Agricultural Science; Kobe University; Kobe Japan
| | - Hana Oya
- Graduate School of Agricultural Science; Kobe University; Kobe Japan
| | - Shohei Tamura
- Graduate School of Agricultural Science; Kobe University; Kobe Japan
| | - Takaoki Saneyasu
- Graduate School of Agricultural Science; Kobe University; Kobe Japan
| | - Kazuhisa Honda
- Graduate School of Agricultural Science; Kobe University; Kobe Japan
| | | |
Collapse
|
25
|
Videla LA, Fernández V, Vargas R, Cornejo P, Tapia G, Varela N, Valenzuela R, Arenas A, Fernández J, Hernández-Rodas MC, Riquelme B. Upregulation of rat liver PPARα-FGF21 signaling by a docosahexaenoic acid and thyroid hormone combined protocol. Biofactors 2016; 42:638-646. [PMID: 27248050 DOI: 10.1002/biof.1300] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/02/2016] [Accepted: 05/04/2016] [Indexed: 12/16/2022]
Abstract
Prevention of ischemia-reperfusion liver injury is achieved by a combined omega-3 and thyroid hormone (T3 ) protocol, which may involve peroxisome-proliferator activated receptor-α (PPAR-α)-fibroblast growth factor 21 (FGF21) signaling supporting energy requirements. Combined docosahexaenoic acid (DHA; daily doses of 300 mg/kg for 3 days) plus 0.05 mg T3 /kg given to fed rats elicited higher hepatic DHA contents and serum T3 levels, increased PPAR-α mRNA and its DNA binding, with higher mRNA expression of the PPAR-α target genes for carnitine-palmitoyl transferase 1α, acyl-CoA oxidase, and 3-hydroxyl-3-methylglutaryl-CoA synthase 2, effects that were mimicked by 0.1 mg T3 /kg given alone or by the PPAR-α agonist WY-14632. Under these conditions, the mRNA expression of retinoic X receptor-α (RXR-α) is also increased, with concomitant elevation of the hepatic mRNA and protein FGF21 levels and those of serum FGF21. It is concluded that PPAR-α-FGF21 induction by DHA combined with T3 may involve ligand activation of PPAR-α by DHA and enhanced expression of PPAR-α by T3 , with consequent upregulation of the FGF21 that is controlled by PPAR-α. Considering the beneficial effects of PPAR-α-FGF21 signaling on carbohydrate and lipid metabolism, further investigations are required to clarify its potential therapeutic applications in human metabolic disorders. © 2016 BioFactors, 42(6):638-646, 2016.
Collapse
Affiliation(s)
- Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Virginia Fernández
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Romina Vargas
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pamela Cornejo
- School of Medical Technology, Faculty of Health and Odontology, Diego Portales University, Santiago, Chile
| | - Gladys Tapia
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nelson Varela
- Department of Medical Technology, Faculty of Medicine, University of Chile, Chile
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Allan Arenas
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Javier Fernández
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | - Bárbara Riquelme
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
26
|
Ratman D, Mylka V, Bougarne N, Pawlak M, Caron S, Hennuyer N, Paumelle R, De Cauwer L, Thommis J, Rider MH, Libert C, Lievens S, Tavernier J, Staels B, De Bosscher K. Chromatin recruitment of activated AMPK drives fasting response genes co-controlled by GR and PPARα. Nucleic Acids Res 2016; 44:10539-10553. [PMID: 27576532 PMCID: PMC5159533 DOI: 10.1093/nar/gkw742] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/08/2016] [Accepted: 08/15/2016] [Indexed: 12/21/2022] Open
Abstract
Adaptation to fasting involves both Glucocorticoid Receptor (GRα) and Peroxisome Proliferator-Activated Receptor α (PPARα) activation. Given both receptors can physically interact we investigated the possibility of a genome-wide cross-talk between activated GR and PPARα, using ChIP- and RNA-seq in primary hepatocytes. Our data reveal extensive chromatin co-localization of both factors with cooperative induction of genes controlling lipid/glucose metabolism. Key GR/PPAR co-controlled genes switched from transcriptional antagonism to cooperativity when moving from short to prolonged hepatocyte fasting, a phenomenon coinciding with gene promoter recruitment of phosphorylated AMP-activated protein kinase (AMPK) and blocked by its pharmacological inhibition. In vitro interaction studies support trimeric complex formation between GR, PPARα and phospho-AMPK. Long-term fasting in mice showed enhanced phosphorylation of liver AMPK and GRα Ser211. Phospho-AMPK chromatin recruitment at liver target genes, observed upon prolonged fasting in mice, is dampened by refeeding. Taken together, our results identify phospho-AMPK as a molecular switch able to cooperate with nuclear receptors at the chromatin level and reveal a novel adaptation mechanism to prolonged fasting.
Collapse
Affiliation(s)
- Dariusz Ratman
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Viacheslav Mylka
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Nadia Bougarne
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Michal Pawlak
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Sandrine Caron
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Nathalie Hennuyer
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Réjane Paumelle
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France
| | - Lode De Cauwer
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Jonathan Thommis
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Mark H Rider
- de Duve Institute and Université catholique de Louvain, 1200 Brussels, Belgium
| | - Claude Libert
- Inflammation Research Center, VIB, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Sam Lievens
- Department of Biochemistry, Ghent University, 9000 Ghent, Belgium.,Receptor Research Laboratories, Cytokine Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium
| | - Jan Tavernier
- Department of Biochemistry, Ghent University, 9000 Ghent, Belgium.,Receptor Research Laboratories, Cytokine Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium
| | - Bart Staels
- UNIV LILLE, 59000 Lille, France.,INSERM UMR 1011, 59000 Lille, France.,European Genomic Institute for Diabetes E.G.I.D., FR 3508, 59000 Lille, France.,Institut Pasteur de Lille, 59000 Lille, France.,CHU Lille, Department of Biology, 59000 Lille, France
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, 9000 Ghent, Belgium .,Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
27
|
Rando G, Tan CK, Khaled N, Montagner A, Leuenberger N, Bertrand-Michel J, Paramalingam E, Guillou H, Wahli W. Glucocorticoid receptor-PPARα axis in fetal mouse liver prepares neonates for milk lipid catabolism. eLife 2016; 5. [PMID: 27367842 PMCID: PMC4963200 DOI: 10.7554/elife.11853] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 06/30/2016] [Indexed: 01/12/2023] Open
Abstract
In mammals, hepatic lipid catabolism is essential for the newborns to efficiently use milk fat as an energy source. However, it is unclear how this critical trait is acquired and regulated. We demonstrate that under the control of PPARα, the genes required for lipid catabolism are transcribed before birth so that the neonatal liver has a prompt capacity to extract energy from milk upon suckling. The mechanism involves a fetal glucocorticoid receptor (GR)-PPARα axis in which GR directly regulates the transcriptional activation of PPARα by binding to its promoter. Certain PPARα target genes such as Fgf21 remain repressed in the fetal liver and become PPARα responsive after birth following an epigenetic switch triggered by β-hydroxybutyrate-mediated inhibition of HDAC3. This study identifies an endocrine developmental axis in which fetal GR primes the activity of PPARα in anticipation of the sudden shifts in postnatal nutrient source and metabolic demands. DOI:http://dx.doi.org/10.7554/eLife.11853.001 Birth is a highly stressful and critical event. In the womb, babies rely on the supply of oxygen and nutrients provided by the placenta. However, once they are born they need to breathe for themselves and gain all their nutrients from suckling milk. The placenta provides a sugar-rich diet, while milk is richer in fat. Failing to cope with this change in diet leads to serious complications and sometimes death. Therefore, a better understanding of how the body adapts to these changes may shed light on pathways that are important for good health in later life. The liver plays a central role in processing the nutrients absorbed by the gut. It uses fats to produce molecules called ketone bodies, such as β-hydroxybutyrate, which are then used as fuel by other tissues and organs including the heart, muscle and the brain. A protein called PPARα controls the production of ketone bodies primarily by regulating genes that are involved in the uptake and breakdown of fat in the liver. However, little is known about how this protein affects the development of the liver. Here, Rando, Tan et al. report that mice start to produce more PPARα in the liver shortly before birth. This ultimately activates several genes that encode enzymes that break down fats. The experiments show that during labor, stress hormones called glucocorticoids directly stimulate the production of PPARα in the liver of the fetus to prepare newborn mice for harnessing energy from fat-rich milk. In the absence of PPARα, mouse liver cells are less able to break down fats after birth and so start to accumulate fat, resulting in fewer ketone bodies being produced. Rando, Tan et al. show that β-hydroxybutyrate regulates some PPARα target genes, including one called Fgf21. The activity of this gene increases only after milk suckling starts and it encodes a protein that enhances the breakdown of fats in the liver. Without PPARα, the expression levels of its target genes, including Fgf21, do not increase after birth, which promotes the build up of fats in liver cells, a condition known as liver steatosis. Overall, the results reported by Rando, Tan et al. highlight how stress during labor plays an important role in priming the body to cope with a fat-rich diet after birth. Future studies will need to determine if stress hormones and ketone bodies could be used as therapies for babies born by caesarean section with liver steatosis. DOI:http://dx.doi.org/10.7554/eLife.11853.002
Collapse
Affiliation(s)
- Gianpaolo Rando
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Chek Kun Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, , Singapore
| | - Nourhène Khaled
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Alexandra Montagner
- UMR 1331 ToxAlim Research Centre in Food Toxicology, INRA, Université de Toulouse, Toulouse, France
| | - Nicolas Leuenberger
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Justine Bertrand-Michel
- IFR 150 Plateforme Metatoul, Institut Fédératif de Recherche Bio-Médicale de Toulouse INSERM U563, Toulouse, France
| | - Eeswari Paramalingam
- Lee Kong Chian School of Medicine, Nanyang Technological University, , Singapore
| | - Hervé Guillou
- UMR 1331 ToxAlim Research Centre in Food Toxicology, INRA, Université de Toulouse, Toulouse, France
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Lee Kong Chian School of Medicine, Nanyang Technological University, , Singapore.,UMR 1331 ToxAlim Research Centre in Food Toxicology, INRA, Université de Toulouse, Toulouse, France
| |
Collapse
|
28
|
Laermans J, Depoortere I. Chronobesity: role of the circadian system in the obesity epidemic. Obes Rev 2016; 17:108-25. [PMID: 26693661 DOI: 10.1111/obr.12351] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/25/2015] [Accepted: 10/01/2015] [Indexed: 01/17/2023]
Abstract
Although obesity is considered to result from an imbalance between energy uptake and energy expenditure, the strategy of dietary changes and physical exercise has failed to tackle the global obesity epidemic. In search of alternative and more adequate treatment options, research has aimed at further unravelling the mechanisms underlying this excessive weight gain. While numerous studies are focusing on the neuroendocrine alterations that occur after bariatric Roux-en-Y gastric bypass surgery, an increasing amount of chronobiological studies have started to raise awareness concerning the pivotal role of the circadian system in the development and exacerbation of obesity. This internal timekeeping mechanism rhythmically regulates metabolic and physiological processes in order to meet the fluctuating demands in energy use and supply throughout the 24-h day. This review elaborates on the extensive bidirectional interaction between the circadian system and metabolism and explains how disruption of body clocks by means of shift work, frequent time zone travelling or non-stop consumption of calorie-dense foods can evoke detrimental metabolic alterations that contribute to obesity. Altering the body's circadian rhythms by means of time-related dietary approaches (chrononutrition) or pharmacological substances (chronobiotics) may therefore represent a novel and interesting way to prevent or treat obesity and associated comorbidities.
Collapse
Affiliation(s)
- J Laermans
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - I Depoortere
- Gut Peptide Research Lab, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Abstract
In response to stress, the central nervous system initiates a signaling cascade, which leads to the production of glucocorticoids (GCs). GCs act through the glucocorticoid receptor (GR) to coordinate the appropriate cellular response with the primary goal of mobilizing the storage forms of carbon precursors to generate a continuous glucose supply for the brain. Although GCs are critical for maintaining energy homeostasis, excessive GC stimulation leads to a number of undesirable side effects, including hyperglycemia, insulin resistance, fatty liver, obesity, and muscle wasting leading to severe metabolic dysfunction. Summarized below are the diverse metabolic roles of glucocorticoids in energy homeostasis and dysregulation, focusing specifically on glucose, lipid, and protein metabolism.
Collapse
Affiliation(s)
- Lilia Magomedova
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
30
|
PPARs: Protectors or Opponents of Myocardial Function? PPAR Res 2015; 2015:835985. [PMID: 26713088 PMCID: PMC4680114 DOI: 10.1155/2015/835985] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 12/15/2022] Open
Abstract
Over 5 million people in the United States suffer from the complications of heart failure (HF), which is a rapidly expanding health complication. Disorders that contribute to HF include ischemic cardiac disease, cardiomyopathies, and hypertension. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family. There are three PPAR isoforms: PPARα, PPARγ, and PPARδ. They can be activated by endogenous ligands, such as fatty acids, as well as by pharmacologic agents. Activators of PPARs are used for treating several metabolic complications, such as diabetes and hyperlipidemia that are directly or indirectly associated with HF. However, some of these drugs have adverse effects that compromise cardiac function. This review article aims to summarize the current basic and clinical research findings of the beneficial or detrimental effects of PPAR biology on myocardial function.
Collapse
|
31
|
Retraction. Prenatal PFOA exposure alters gene expression pathways in murine mammary gland. Toxicol Sci 2014; 145:211. [PMID: 25490953 DOI: 10.1093/toxsci/kfu253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
32
|
Lecarpentier Y, Claes V, Duthoit G, Hébert JL. Circadian rhythms, Wnt/beta-catenin pathway and PPAR alpha/gamma profiles in diseases with primary or secondary cardiac dysfunction. Front Physiol 2014; 5:429. [PMID: 25414671 PMCID: PMC4220097 DOI: 10.3389/fphys.2014.00429] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/15/2014] [Indexed: 12/13/2022] Open
Abstract
Circadian clock mechanisms are far-from-equilibrium dissipative structures. Peroxisome proliferator-activated receptors (PPAR alpha, beta/delta, and gamma) play a key role in metabolic regulatory processes, particularly in heart muscle. Links between circadian rhythms (CRs) and PPARs have been established. Mammalian CRs involve at least two critical transcription factors, CLOCK and BMAL1 (Gekakis et al., 1998; Hogenesch et al., 1998). PPAR gamma plays a major role in both glucose and lipid metabolisms and presents circadian properties which coordinate the interplay between metabolism and CRs. PPAR gamma is a major component of the vascular clock. Vascular PPAR gamma is a peripheral regulator of cardiovascular rhythms controlling circadian variations in blood pressure and heart rate through BMAL1. We focused our review on diseases with abnormalities of CRs and with primary or secondary cardiac dysfunction. Moreover, these diseases presented changes in the Wnt/beta-catenin pathway and PPARs, according to two opposed profiles. Profile 1 was defined as follows: inactivation of the Wnt/beta-catenin pathway with increased expression of PPAR gamma. Profile 2 was defined as follows: activation of the Wnt/beta-catenin pathway with decreased expression of PPAR gamma. A typical profile 1 disease is arrhythmogenic right ventricular cardiomyopathy, a genetic cardiac disease which presents mutations of the desmosomal proteins and is mainly characterized by fatty acid accumulation in adult cardiomyocytes mainly in the right ventricle. The link between PPAR gamma dysfunction and desmosomal genetic mutations occurs via inactivation of the Wnt/beta-catenin pathway presenting oscillatory properties. A typical profile 2 disease is type 2 diabetes, with activation of the Wnt/beta-catenin pathway and decreased expression of PPAR gamma. CRs abnormalities are present in numerous pathologies such as cardiovascular diseases, sympathetic/parasympathetic dysfunction, hypertension, diabetes, neurodegenerative diseases, cancer which are often closely inter-related.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Centre Hospitalier Régional de Meaux Meaux, France
| | - Victor Claes
- Department of Pharmaceutical Sciences, University of Antwerp Wilrijk, Belgium
| | - Guillaume Duthoit
- Institut de Cardiologie, Hôpital de la Pitié-Salpêtière Paris, France
| | - Jean-Louis Hébert
- Institut de Cardiologie, Hôpital de la Pitié-Salpêtière Paris, France
| |
Collapse
|
33
|
Glucocorticoids promote structural and functional maturation of foetal cardiomyocytes: a role for PGC-1α. Cell Death Differ 2014; 22:1106-16. [PMID: 25361084 PMCID: PMC4572859 DOI: 10.1038/cdd.2014.181] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 09/08/2014] [Accepted: 09/22/2014] [Indexed: 01/05/2023] Open
Abstract
Glucocorticoid levels rise dramatically in late gestation to mature foetal organs in readiness for postnatal life. Immature heart function may compromise survival. Cardiomyocyte glucocorticoid receptor (GR) is required for the structural and functional maturation of the foetal heart in vivo, yet the molecular mechanisms are largely unknown. Here we asked if GR activation in foetal cardiomyocytes in vitro elicits similar maturational changes. We show that physiologically relevant glucocorticoid levels improve contractility of primary-mouse-foetal cardiomyocytes, promote Z-disc assembly and the appearance of mature myofibrils, and increase mitochondrial activity. Genes induced in vitro mimic those induced in vivo and include PGC-1α, a critical regulator of cardiac mitochondrial capacity. SiRNA-mediated abrogation of the glucocorticoid induction of PGC-1α in vitro abolished the effect of glucocorticoid on myofibril structure and mitochondrial oxygen consumption. Using RNA sequencing we identified a number of transcriptional regulators, including PGC-1α, induced as primary targets of GR in foetal cardiomyocytes. These data demonstrate that PGC-1α is a key mediator of glucocorticoid-induced maturation of foetal cardiomyocyte structure and identify other candidate transcriptional regulators that may play critical roles in the transition of the foetal to neonatal heart.
Collapse
|
34
|
Su Y, van der Spek R, Foppen E, Kwakkel J, Fliers E, Kalsbeek A. Effects of adrenalectomy on daily gene expression rhythms in the rat suprachiasmatic and paraventricular hypothalamic nuclei and in white adipose tissue. Chronobiol Int 2014; 32:211-24. [DOI: 10.3109/07420528.2014.963198] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
35
|
Pan ZQ, Xie D, Choudhary V, Seremwe M, Tsai YY, Olala L, Chen X, Bollag WB. The effect of pioglitazone on aldosterone and cortisol production in HAC15 human adrenocortical carcinoma cells. Mol Cell Endocrinol 2014; 394:119-28. [PMID: 25038520 PMCID: PMC4237224 DOI: 10.1016/j.mce.2014.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 06/16/2014] [Accepted: 07/08/2014] [Indexed: 01/27/2023]
Abstract
Pioglitazone belongs to the class of drugs called thiazolidinediones (TZDs), which are widely used as insulin sensitizers in the treatment of diabetes. A major side effect of TZDs is fluid retention. The steroid hormone aldosterone also promotes sodium and fluid retention; however, the effect of pioglitazone on aldosterone production is controversial. We analyzed the effect of pioglitazone alone and in combination with angiotensin II (AngII) on the late rate-limiting step of adrenocortical steroidogenesis in human adrenocortical carcinoma HAC15 cells. Treatment with pioglitazone for 24 h significantly increased the expression of CYP11B2 and enhanced AngII-induced CYP11B2 expression. Despite the observed changes in mRNA levels, pioglitazone significantly inhibited AngII-induced aldosterone production and CYP11B2 protein levels. On the other hand, pioglitazone stimulated the expression of the unfolded protein response (UPR) marker DDIT3, with this effect occurring at early times and inhibitable by the PPARγ antagonist GW9962. The levels of DDIT3 (CHOP) and phospho-eIF2α (Ser51), a UPR-induced event that inhibits protein translation, were also increased. Thus, pioglitazone promotes CYP11B2 expression but nevertheless inhibits aldosterone production in AngII-treated HAC15 cells, likely by blocking global protein translation initiation through DDIT3 and phospho-eIF2α. In contrast, pioglitazone promoted AngII-induced CYP11B1 expression and cortisol production. Since cortisol enhances lipolysis, this result suggests the possibility that PPARs, activated by products of fatty acid oxidation, stimulate cortisol secretion to promote utilization of fatty acids during fasting. In turn, the ability of pioglitazone to stimulate cortisol production could potentially underlie the effects of this drug on fluid retention.
Collapse
Affiliation(s)
- Zhi-qiang Pan
- Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA; School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ding Xie
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA
| | - Vivek Choudhary
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA
| | - Mutsa Seremwe
- Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA
| | - Ying-Ying Tsai
- Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA
| | - Lawrence Olala
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA
| | - Xunsheng Chen
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA
| | - Wendy B Bollag
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA 30904, USA; Department of Physiology, Georgia Regents University (Medical College of Georgia), 1120 15th Street, Augusta, GA 30912, USA.
| |
Collapse
|
36
|
Varcoe TJ, Gatford KL, Voultsios A, Salkeld MD, Boden MJ, Rattanatray L, Kennaway DJ. Rapidly alternating photoperiods disrupt central and peripheral rhythmicity and decrease plasma glucose, but do not affect glucose tolerance or insulin secretion in sheep. Exp Physiol 2014; 99:1214-28. [DOI: 10.1113/expphysiol.2014.080630] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Tamara J. Varcoe
- Robinson Research Institute; School of Paediatrics and Reproductive Health; University of Adelaide; Adelaide SA 5005 Australia
| | - Kathryn L. Gatford
- Robinson Research Institute; School of Paediatrics and Reproductive Health; University of Adelaide; Adelaide SA 5005 Australia
| | - Athena Voultsios
- Robinson Research Institute; School of Paediatrics and Reproductive Health; University of Adelaide; Adelaide SA 5005 Australia
| | - Mark D. Salkeld
- Robinson Research Institute; School of Paediatrics and Reproductive Health; University of Adelaide; Adelaide SA 5005 Australia
| | - Michael J. Boden
- Robinson Research Institute; School of Paediatrics and Reproductive Health; University of Adelaide; Adelaide SA 5005 Australia
| | - Leewen Rattanatray
- Robinson Research Institute; School of Paediatrics and Reproductive Health; University of Adelaide; Adelaide SA 5005 Australia
| | - David J. Kennaway
- Robinson Research Institute; School of Paediatrics and Reproductive Health; University of Adelaide; Adelaide SA 5005 Australia
| |
Collapse
|
37
|
Ikeda I, Metoki K, Yamahira T, Kato M, Inoue N, Nagao K, Yanagita T, Shirakawa H, Komai M. Impact of fasting time on hepatic lipid metabolism in nutritional animal studies. Biosci Biotechnol Biochem 2014; 78:1584-91. [PMID: 25209508 DOI: 10.1080/09168451.2014.923297] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Many animal studies on improvement of lipid metabolism, using dietary components, fast the animals on the final day of the feeding. Although fasting has a significant impact on lipid metabolism, its time-dependent influence is not fully understood. We examined the effects of several fasting times on lipid metabolism. Rats fed with a semisynthetic diet for 2 wk were killed after 0 (9:00 am), 6 (7:00 am-1:00 pm), 9 (0:00 am-9:00 am), and 13 h (8:00 pm-9:00 am) of fasting. Compared to the 0 h group, marked reduction of liver weight and hepatic triacylglycerol content was observed in the 9 and 13 h groups. Activities of hepatic enzymes involved in fatty acid synthesis gradually decreased during fasting. In contrast, drastic time-dependent reduction of gene expression, of the enzymes, was observed. Expression of carnitine palmitoyltransferase mRNA was higher in the fasting groups than in the 0 h group. Our study showed that fasting has a significant impact on several parameters related to lipid metabolism in rat liver.
Collapse
Affiliation(s)
- Ikuo Ikeda
- a Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science , Tohoku University , Sendai , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Patel R, Williams-Dautovich J, Cummins CL. Minireview: new molecular mediators of glucocorticoid receptor activity in metabolic tissues. Mol Endocrinol 2014; 28:999-1011. [PMID: 24766141 DOI: 10.1210/me.2014-1062] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The glucocorticoid receptor (GR) was one of the first nuclear hormone receptors cloned and represents one of the most effective drug targets available today for the treatment of severe inflammation. The physiologic consequences of endogenous or exogenous glucocorticoid excess are well established and include hyperglycemia, insulin resistance, fatty liver, obesity, and muscle wasting. However, at the molecular and tissue-specific level, there are still many unknown protein mediators of glucocorticoid response and thus, much remains to be uncovered that will help determine whether activation of the GR can be tailored to improve therapeutic efficacy while minimizing unwanted side effects. This review summarizes recent discoveries of tissue-selective modulators of glucocorticoid signaling that are important in mediating the unwanted side effects of therapeutic glucocorticoid use, emphasizing the downstream molecular effects of GR activation in the liver, adipose tissue, muscle, and pancreas.
Collapse
Affiliation(s)
- Rucha Patel
- Department of Pharmaceutical Sciences (R.P., J.W-D., C.L.C.), University of Toronto, Toronto, Ontario, M5S 3M2, Canada; and Banting and Best Diabetes Centre (C.L.C.), Toronto, Ontario M5G 2C4 Canada
| | | | | |
Collapse
|
39
|
Sun X, Luo W, Tan X, Li Q, Zhao Y, Zhong W, Sun X, Brouwer C, Zhou Z. Increased plasma corticosterone contributes to the development of alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol 2013; 305:G849-61. [PMID: 24113770 PMCID: PMC3882437 DOI: 10.1152/ajpgi.00139.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ethanol ingestion increases endogenous glucocorticoid levels in both humans and rodents. The present study aimed to define a mechanistic link between the increased glucocorticoids and alcoholic fatty liver in mice. Plasma corticosterone levels were not affected in mice on a 2-wk ethanol diet regimen but significantly increased upon 4 wk of ethanol ingestion. Accordingly, hepatic triglyceride levels were not altered after 2 wk of ethanol ingestion but were elevated at 4 wk. Based on the observation that 2 wk of ethanol ingestion did not significantly increase endogenous corticosterone levels, we administered exogenous glucocorticoids along with the 2-wk ethanol treatment to determine whether the elevated glucocorticoid contributes to the development of alcoholic fatty liver. Mice were subjected to ethanol feeding for 2 wk with or without dexamethasone administration. Hepatic triglyceride contents were not affected by either ethanol or dexamethasone alone but were significantly increased by administration of both. Microarray and protein level analyses revealed two distinct changes in hepatic lipid metabolism in mice administered with both ethanol and dexamethasone: accelerated triglyceride synthesis by diacylglycerol O-acyltransferase 2 and suppressed fatty acid β-oxidation by long-chain acyl-CoA synthetase 1, carnitine palmitoyltransferase 1a, and acyl-CoA oxidase 1. A reduction of hepatic peroxisome proliferation activator receptor-α (PPAR-α) was associated with coadministration of ethanol and dexamethasone. These findings suggest that increased glucocorticoid levels may contribute to the development of alcoholic fatty liver, at least partially, through hepatic PPAR-α inactivation.
Collapse
Affiliation(s)
- Xiuhua Sun
- Ctr. for Translational & Biomedical Research, The Univ. of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081.
| | - Weijun Luo
- 3Bioinformatics Services Division, UNC-Charlotte, ,4Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis;
| | - Xiaobing Tan
- 1Center for Translational and Biomedical Research,
| | - Qiong Li
- 1Center for Translational and Biomedical Research,
| | - Yantao Zhao
- 5Department of Bioinformatics and Genomics, The University of North Carolina at Charlotte, North Carolina
| | - Wei Zhong
- 1Center for Translational and Biomedical Research,
| | - Xinguo Sun
- 1Center for Translational and Biomedical Research,
| | - Cory Brouwer
- 3Bioinformatics Services Division, UNC-Charlotte, ,4Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis;
| | - Zhanxiang Zhou
- 1Center for Translational and Biomedical Research, ,2Department of Nutrition, The University of North Carolina at Greensboro,
| |
Collapse
|
40
|
Gu M, Zhang Y, Fan S, Ding X, Ji G, Huang C. Extracts of Rhizoma polygonati odorati prevent high-fat diet-induced metabolic disorders in C57BL/6 mice. PLoS One 2013; 8:e81724. [PMID: 24312343 PMCID: PMC3843710 DOI: 10.1371/journal.pone.0081724] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 10/16/2013] [Indexed: 12/30/2022] Open
Abstract
Polygonatum odoratum (Mill.) Druce belongs to the genus Polygonatum family of plants. In traditional Chinese medicine, the root of Polygonatum odoratum, Rhizoma Polygonati Odorati, is used both for food and medicine to prevent and treat metabolic disorders such as hyperlipidemia, hyperglycemia, obesity and cardiovascular disease. However, there is no solid experimental evidence to support these applications, and the underlying mechanism is also needed to be elucidated. Here, we examined the effect of the extract of Rhizoma Polygonati Odorati (ER) on metabolic disorders in diet-induced C57BL/6 obese mice. In the preventive experiment, the ER blocked body weight gain, and lowered serum total cholesterol (TC), triglyceride (TG) and fasting blood glucose, improved glucose tolerance test (GTT) and insulin tolerance test (ITT), reduced the levels of serum insulin and leptin, and increased serum adiponectin levels in mice fed with a high-fat diet significantly. In the therapeutic study, we induced obesity in the mice and treated the obese mice with ER for two weeks. We found that ER treatments reduced serum TG and fasting blood glucose, and improved glucose tolerance in the mice. Gene expression analysis showed that ER increased the mRNA levels of peroxisome proliferator-activated receptors (PPAR) γ and α and their downstream target genes in mice livers, adipose tissues and HepG2 cells. Our data suggest that ER ameliorates metabolic disorders and enhances the mRNA expression of PPARs in obese C57BL/6 mice induced by high-fat diet.
Collapse
Affiliation(s)
- Ming Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaobo Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- College of Horticulture and Landscape Architecture, Southwest University, Chongqing, China
- Key Laboratory of Horticulture Science for Southern Mountainous Regions, Ministry of Education, Chongqing, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
41
|
Garratt ES, Vickers MH, Gluckman PD, Hanson MA, Burdge GC, Lillycrop KA. Tissue-specific 5' heterogeneity of PPARα transcripts and their differential regulation by leptin. PLoS One 2013; 8:e67483. [PMID: 23825665 PMCID: PMC3692471 DOI: 10.1371/journal.pone.0067483] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 05/19/2013] [Indexed: 11/18/2022] Open
Abstract
The genes encoding nuclear receptors comprise multiple 5'untranslated exons, which give rise to several transcripts encoding the same protein, allowing tissue-specific regulation of expression. Both human and mouse peroxisome proliferator activated receptor (PPAR) α genes have multiple promoters, although their function is unknown. Here we have characterised the rat PPARα promoter region and have identified three alternative PPARα transcripts, which have different transcription start sites owing to the utilisation of distinct first exons. Moreover these alternative PPARα transcripts were differentially expressed between adipose tissue and liver. We show that while the major adipose (P1) and liver (P2) transcripts were both induced by dexamethasone, they were differentially regulated by the PPARα agonist, clofibric acid, and leptin. Leptin had no effect on the adipose-specific P1 transcript, but induced liver-specific P2 promoter activity via a STAT3/Sp1 mechanism. Moreover in Wistar rats, leptin treatment between postnatal day 3-13 led to an increase in P2 but not P1 transcription in adipose tissue which was sustained into adulthood. This suggests that the expression of the alternative PPARα transcripts are in part programmed by early life exposure to leptin leading to persistent change in adipose tissue fatty acid metabolism through specific activation of a quiescent PPARα promoter. Such complexity in the regulation of PPARα may allow the expression of PPARα to be finely regulated in response to environmental factors.
Collapse
Affiliation(s)
- Emma S. Garratt
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mark H. Vickers
- Liggins Institute and the National Research Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| | - Peter D. Gluckman
- Liggins Institute and the National Research Centre for Growth and Development, University of Auckland, Auckland, New Zealand
| | - Mark A. Hanson
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Graham C. Burdge
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Karen A. Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
42
|
Castillero E, Alamdari N, Aversa Z, Gurav A, Hasselgren PO. PPARβ/δ regulates glucocorticoid- and sepsis-induced FOXO1 activation and muscle wasting. PLoS One 2013; 8:e59726. [PMID: 23555761 PMCID: PMC3605288 DOI: 10.1371/journal.pone.0059726] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Accepted: 02/17/2013] [Indexed: 01/01/2023] Open
Abstract
FOXO1 is involved in glucocorticoid- and sepsis-induced muscle wasting, in part reflecting regulation of atrogin-1 and MuRF1. Mechanisms influencing FOXO1 expression in muscle wasting are poorly understood. We hypothesized that the transcription factor peroxisome proliferator-activated receptor β/δ (PPARβ/δ) upregulates muscle FOXO1 expression and activity with a downstream upregulation of atrogin-1 and MuRF1 expression during sepsis and glucocorticoid treatment and that inhibition of PPARβ/δ activity can prevent muscle wasting. We found that activation of PPARβ/δ in cultured myotubes increased FOXO1 activity, atrogin-1 and MuRF1 expression, protein degradation and myotube atrophy. Treatment of myotubes with dexamethasone increased PPARβ/δ expression and activity. Dexamethasone-induced FOXO1 activation and atrogin-1 and MuRF1 expression, protein degradation, and myotube atrophy were inhibited by PPARβ/δ blocker or siRNA. Importantly, muscle wasting induced in rats by dexamethasone or sepsis was prevented by treatment with a PPARβ/δ inhibitor. The present results suggest that PPARβ/δ regulates FOXO1 activation in glucocorticoid- and sepsis-induced muscle wasting and that treatment with a PPARβ/δ inhibitor may ameliorate loss of muscle mass in these conditions.
Collapse
Affiliation(s)
- Estibaliz Castillero
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nima Alamdari
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Zaira Aversa
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aniket Gurav
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Per-Olof Hasselgren
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
43
|
Silvennoinen R, Escola-Gil JC, Julve J, Rotllan N, Llaverias G, Metso J, Valledor AF, He J, Yu L, Jauhiainen M, Blanco-Vaca F, Kovanen PT, Lee-Rueckert M. Acute Psychological Stress Accelerates Reverse Cholesterol Transport via Corticosterone-Dependent Inhibition of Intestinal Cholesterol Absorption. Circ Res 2012; 111:1459-69. [DOI: 10.1161/circresaha.112.277962] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rationale:
Psychological stress is associated with an increased risk of cardiovascular diseases. However, the connecting mechanisms of the stress-inducing activation of the hypothalamic-pituitary-adrenal axis with atherosclerosis are not well-understood.
Objective:
To study the effect of acute psychological stress on reverse cholesterol transport (RCT), which transfers peripheral cholesterol to the liver for its ultimate fecal excretion.
Methods and Results:
C57Bl/6J mice were exposed to restraint stress for 3 hours to induce acute psychological stress. RCT in vivo was quantified by measuring the transfer of [
3
H]cholesterol from intraperitoneally injected mouse macrophages to the lumen of the small intestine within the stress period. Surprisingly, stress markedly increased the contents of macrophage-derived [
3
H]cholesterol in the intestinal lumen. In the stressed mice, intestinal absorption of [
14
C]cholesterol was significantly impaired, the intestinal mRNA expression level of peroxisome proliferator–activated receptor-α increased, and that of the sterol influx transporter Niemann-Pick C1–like 1 decreased. The stress-dependent effects on RCT rate and peroxisome proliferator–activated receptor-α gene expression were fully mimicked by administration of the stress hormone corticosterone (CORT) to nonstressed mice, and they were blocked by the inhibition of CORT synthesis in stressed mice. Moreover, the intestinal expression of Niemann-Pick C1–like 1 protein decreased when circulating levels of CORT increased. Of note, when either peroxisome proliferator-activated receptor α or liver X receptor α knockout mice were exposed to stress, the RCT rate remained unchanged, although plasma CORT increased. This indicates that activities of both transcription factors were required for the RCT-accelerating effect of stress.
Conclusions:
Acute psychological stress accelerated RCT by compromising intestinal cholesterol absorption. The present results uncover a novel functional connection between the hypothalamic-pituitary-adrenal axis and RCT that can be triggered by a stress-induced increase in circulating CORT.
Collapse
Affiliation(s)
- Reija Silvennoinen
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Joan Carles Escola-Gil
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Josep Julve
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Noemi Rotllan
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Gemma Llaverias
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Jari Metso
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Annabel F. Valledor
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Jianming He
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Liqing Yu
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Matti Jauhiainen
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Francisco Blanco-Vaca
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Petri T. Kovanen
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| | - Miriam Lee-Rueckert
- From the Wihuri Research Institute, Helsinki, Finland (R.S., P.T.K., M.L.-R.); Departament de Bioquimica, IIB Sant Pau-CIBER de Diabetes y Enfermedades Metabolicas Asociadas-Universitat Autonoma de Barcelona, Barcelona, Spain (J.C.E.-G., J.J., N.R., G.L., F.B.-V.); Department of Chronic Disease Prevention, National Institute for Health and Welfare, Public Health Genomics Research Unit Biomedicum, Helsinki, Finland (J.M., M.J.); Department of Physiology and Immunology, School of Biology, University
| |
Collapse
|
44
|
PPARα-Independent Arterial Smooth Muscle Relaxant Effects of PPARα Agonists. PPAR Res 2012; 2012:302495. [PMID: 23008696 PMCID: PMC3447381 DOI: 10.1155/2012/302495] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 07/26/2012] [Indexed: 02/07/2023] Open
Abstract
We sought to determine direct vascular effects of peroxisome proliferator-activated receptor alpha (PPARα) agonists using isolated mouse aortas and middle cerebral arteries (MCAs). The PPARα agonists GW7647, WY14643, and gemfibrozil acutely relaxed aortas held under isometric tension and dilated pressurized MCAs with the following order of potency: GW7647≫WY14643>gemfibrozil. Responses were endothelium-independent, and the use of PPARα deficient mice demonstrated that responses were also PPARα-independent. Pretreating arteries with high extracellular K+ attenuated PPARα agonist-mediated relaxations in the aorta, but not in the MCA. In the aorta, the ATP sensitive potassium (KATP) channel blocker glibenclamide also impaired relaxations whereas the other K+ channel inhibitors, 4-aminopyridine and Iberiotoxin, had no effect. In aortas, GW7647 and WY14643 elevated cGMP levels by stimulating soluble guanylyl cyclase (sGC), and inhibition of sGC with ODQ blunted relaxations to PPARα agonists. In the MCA, dilations were inhibited by the protein kinase C (PKC) activator, phorbol 12,13-dibutyrate, and also by ODQ. Our results demonstrated acute, nonreceptor-mediated relaxant effects of PPARα agonists on smooth muscle of mouse arteries. Responses to PPARα agonists in the aorta involved KATP channels and sGC, whereas in the MCA the PKC and sGC pathways also appeared to contribute to the response.
Collapse
|
45
|
Giordano R, Guaraldi F, Berardelli R, Karamouzis I, D'Angelo V, Marinazzo E, Picu A, Ghigo E, Arvat E. Glucose metabolism in patients with subclinical Cushing's syndrome. Endocrine 2012; 41:415-23. [PMID: 22391939 DOI: 10.1007/s12020-012-9628-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/01/2012] [Indexed: 01/21/2023]
Abstract
This clinical review will summarize the available data regarding the effect of either physiological or increased glucocorticoid concentrations on glucose metabolism and insulin-sensitivity, in order to clarify the role, if any, of subclinical Cushing's syndrome (SCS), a status of altered hypothalamic-pituitary-adrenal axis secretion in the absence of the classical signs or symptoms of overt cortisol excess, in patients with adrenal incidentalomas (AI) and diabetes mellitus type 2. Focusing on patients with SCS associated to AI, while there is convincing evidence in the literature that even a mild hyper cortisolemia is associated with alterations of glucose metabolism, evidence is insufficient to conclude that the simple correction of chronic, even mild, hypercortisolism can completely revert metabolic, mainly glycemic alterations. At the same time, considering the variability of the prevalence of Cushing's syndrome in patients with diabetes mellitus type 2 reported in the literature, no agreement does exist whether screening for CS can be useful and recommended in those patients.
Collapse
Affiliation(s)
- Roberta Giordano
- Division of Endocrinology, Diabetology and Metabolism, Department of Internal Medicine and Department of Clinical and Biological Sciences, University of Turin, Corso Dogliotti 14, 10126, Torino, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The circadian system of mammals is composed of a hierarchy of oscillators that function at the cellular, tissue, and systems levels. A common molecular mechanism underlies the cell-autonomous circadian oscillator throughout the body, yet this clock system is adapted to different functional contexts. In the central suprachiasmatic nucleus (SCN) of the hypothalamus, a coupled population of neuronal circadian oscillators acts as a master pacemaker for the organism to drive rhythms in activity and rest, feeding, body temperature, and hormones. Coupling within the SCN network confers robustness to the SCN pacemaker, which in turn provides stability to the overall temporal architecture of the organism. Throughout the majority of the cells in the body, cell-autonomous circadian clocks are intimately enmeshed within metabolic pathways. Thus, an emerging view for the adaptive significance of circadian clocks is their fundamental role in orchestrating metabolism.
Collapse
Affiliation(s)
- Jennifer A Mohawk
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111, USA.
| | | | | |
Collapse
|
47
|
Abstract
Stress activates the hypothalamic-pituitary-adrenal (HPA) axis and sympathetic nervous system (SNS), resulting in cardiovascular responses. The endocannabinoid system (ECS), a ubiquitously expressed lipid signalling system, modulates both HPA and SNS activity. The purpose of this review is to explore the possible involvement/role of the ECS in the cardiovascular response to stress. The ECS has numerous cardiovascular effects including modulation of blood pressure, heart rate, the baroreflex, and direct vascular actions. It is also involved in a protective manner in response to stressors in cardiac preconditioning, and various stressors (for example, pain, orthostasis and social stress) increase plasma levels of endocannabinoids. Given the multitude of vascular effects of endocannabinoids, this is bound to have consequences. Beneficial effects of ECS upregulation could include cardioprotection, vasodilatation, CB(2)-mediated anti-inflammatory effects and activation of peroxisome proliferator-activated receptors. Negative effects of endocannabinoids could include mediation of the effects of glucocorticoids, CB(1)-mediated metabolic changes, and metabolism to vasoconstrictor products. It is also likely that there is a central role for the ECS in modulating cardiovascular activity via the HPA and SNS. However, much more work is required to fully integrate the role of the ECS in mediating many of the physiological responses to stress, including cardiovascular responses.
Collapse
Affiliation(s)
- Saoirse E O'Sullivan
- School of Graduate Entry Medicine and Health, University of Nottingham, Nottingham, UK.
| | | | | |
Collapse
|
48
|
Antolino-Lobo I, Meulenbelt J, van den Berg M, van Duursen MB. A mechanistic insight into 3,4-methylenedioxymethamphetamine (“ecstasy”)-mediated hepatotoxicity. Vet Q 2011; 31:193-205. [DOI: 10.1080/01652176.2011.642534] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
49
|
Brunton PJ, Russell JA. Neuroendocrine control of maternal stress responses and fetal programming by stress in pregnancy. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1178-91. [PMID: 21216265 DOI: 10.1016/j.pnpbp.2010.12.023] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Revised: 12/30/2010] [Accepted: 12/31/2010] [Indexed: 02/07/2023]
Abstract
The major changes in highly dynamic neuroendocrine systems that are essential for establishing and maintaining pregnancy are outlined from studies on rodents. These changes optimise the internal environment to provide the life support system for the placenta, embryo and fetus. These include automatic prevention of further pregnancy, blood volume expansion, increased appetite and energy storage. The brain regulates these changes, in response to steroid (estrogens, progesterone) and peptide (lactogens, relaxin) hormone signals. Activation of inhibitory endogenous opioid mechanisms in the brain in late pregnancy restrains premature secretion of oxytocin, and attenuates hypothalamo-pituitary-adrenal (HPA) responses to stress. This opioid mechanism is activated by allopregnanolone, a neuroactive progesterone metabolite. The significance of reduced HPA axis responses in shifting maternal metabolic balance, and in protecting the fetuses from adverse programming of HPA axis stress responsiveness and anxious behaviour in later life is critically discussed. Experimental studies showing sex-dependent fetal programming by maternal stress or glucocorticoid exposure in late pregnancy are reviewed. The possibility of over-writing programming in offspring through neurosteroid administration is discussed. The impact of maternal stress on placental function is considered in the context of reconciling studies that show offspring programming by stress in very early or late pregnancy produce similar phenotypes in the offspring.
Collapse
Affiliation(s)
- Paula J Brunton
- Laboratory of Neuroendocrinology, Centre for Integrative Physiology, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH89XD, UK
| | | |
Collapse
|
50
|
Abstract
Maternal stress experience is associated with neurodevelopmental disorders including schizophrenia and autism. Recent studies have examined mechanisms by which changes in the maternal milieu may be transmitted to the developing embryo and potentially translated into programming of the epigenome. Animal models of prenatal stress have identified important sex- and temporal-specific effects on offspring stress responsivity. As dysregulation of stress pathways is a common feature in most neuropsychiatric diseases, molecular and epigenetic analyses at the maternal-embryo interface, especially in the placenta, may provide unique insight into identifying much-needed predictive biomarkers. In addition, as most neurodevelopmental disorders present with a sex bias, examination of sex differences in the inheritance of phenotypic outcomes may pinpoint gene targets and specific windows of vulnerability in neurodevelopment, which have been disrupted. This review discusses the association and possible contributing mechanisms of prenatal stress in programming offspring stress pathway dysregulation and the importance of sex.
Collapse
Affiliation(s)
- Tracy L Bale
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania Philadelphia, PA 19104, USA.
| |
Collapse
|