1
|
Guerbette T, Ciesielski V, Brien M, Catheline D, Viel R, Bostoën M, Perrin JB, Burel A, Janvier R, Rioux V, Lan A, Boudry G. Bioenergetic adaptations of small intestinal epithelial cells reduce cell differentiation enhancing intestinal permeability in obese mice. Mol Metab 2025; 92:102098. [PMID: 39814101 PMCID: PMC11795564 DOI: 10.1016/j.molmet.2025.102098] [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: 11/29/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
OBJECTIVE Obesity and overweight are associated with low-grade inflammation induced by adipose tissue expansion and perpetuated by altered intestinal homeostasis, including increased epithelial permeability. Intestinal epithelium functions are supported by intestinal epithelial cells (IEC) mitochondria function. However, diet-induced obesity (DIO) may impair mitochondrial activity of IEC and consequently, intestinal homeostasis. The aim of the project was to determine whether DIO alters the mitochondrial function of IEC, and what are the consequences on intestinal homeostasis. METHODS C57Bl/6J mice were fed a control diet for 22 weeks or a high fat diet (58 kcal% fat). Bioenergetic adaptations of IEC were evaluated on isolated crypts and villi from mouse jejunum. To determine the link between mitochondrial function and alterations of intestinal homeostasis in response to lipid overload, we used the jejunal epithelial cell line IPEC-J2 in vitro and mouse jejunum organoids. RESULTS Here, we report that DIO in mice induced lipid metabolism adaptations favoring lipid storage in IEC together with reduced number, altered dynamics and diminished oxidative phosphorylation activity of IEC mitochondria. Using the IPEC-J2 cell line, we showed that IEC lipid metabolism and oxidative stress machinery adaptations preceded mitochondrial bioenergetic ones. Moreover, we unraveled the intricate link between IEC energetic status and proliferation / differentiation balance since enhancing mitochondrial function with the AMPK activator AICAR in jejunal organoids reduced proliferation and initiated IEC differentiation and conversely. We confirmed that the reduced IEC mitochondrial function observed in DIO mice was associated with increased proliferation and reduced differentiation, promoting expression of the permissive Cldn2 in the jejunal epithelium of DIO mice. CONCLUSIONS Our study provides new insights into metabolic adaptations of IEC in obesity by revealing that excess lipid intake diminishes mitochondrial number in IEC, reducing IEC differentiation that contribute to increased epithelial permeability.
Collapse
Affiliation(s)
| | - Vincent Ciesielski
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Manon Brien
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Daniel Catheline
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Roselyne Viel
- Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, France-BioImaging (ANR-10-INBS-04), plateforme H2P2, Rennes, France
| | - Mégane Bostoën
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | | | - Agnès Burel
- Plateforme MRic, UMS 3480 BIOSIT, Rennes, France
| | - Régis Janvier
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Vincent Rioux
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Annaïg Lan
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Gaëlle Boudry
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France.
| |
Collapse
|
2
|
Kumar KK, Aburawi EH, Ljubisavljevic M, Leow MKS, Feng X, Ansari SA, Emerald BS. Exploring histone deacetylases in type 2 diabetes mellitus: pathophysiological insights and therapeutic avenues. Clin Epigenetics 2024; 16:78. [PMID: 38862980 PMCID: PMC11167878 DOI: 10.1186/s13148-024-01692-0] [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: 02/27/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
Diabetes mellitus is a chronic disease that impairs metabolism, and its prevalence has reached an epidemic proportion globally. Most people affected are with type 2 diabetes mellitus (T2DM), which is caused by a decline in the numbers or functioning of pancreatic endocrine islet cells, specifically the β-cells that release insulin in sufficient quantity to overcome any insulin resistance of the metabolic tissues. Genetic and epigenetic factors have been implicated as the main contributors to the T2DM. Epigenetic modifiers, histone deacetylases (HDACs), are enzymes that remove acetyl groups from histones and play an important role in a variety of molecular processes, including pancreatic cell destiny, insulin release, insulin production, insulin signalling, and glucose metabolism. HDACs also govern other regulatory processes related to diabetes, such as oxidative stress, inflammation, apoptosis, and fibrosis, revealed by network and functional analysis. This review explains the current understanding of the function of HDACs in diabetic pathophysiology, the inhibitory role of various HDAC inhibitors (HDACi), and their functional importance as biomarkers and possible therapeutic targets for T2DM. While their role in T2DM is still emerging, a better understanding of the role of HDACi may be relevant in improving insulin sensitivity, protecting β-cells and reducing T2DM-associated complications, among others.
Collapse
Affiliation(s)
- Kukkala Kiran Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Elhadi Husein Aburawi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Milos Ljubisavljevic
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Melvin Khee Shing Leow
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
- Dept of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Xu Feng
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates.
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates.
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
3
|
Sun Q, Bibi S, Xue Y, Du M, Chew B, Zhu MJ. Dietary purple potato supplement attenuates DSS-induced colitis in mice: impact on mitochondrial function. J Nutr Biochem 2024; 126:109585. [PMID: 38253109 DOI: 10.1016/j.jnutbio.2024.109585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 12/18/2023] [Accepted: 01/17/2024] [Indexed: 01/24/2024]
Abstract
Inflammatory bowel disease (IBD) is a condition characterized by disrupted intestinal barrier function, abnormal immune response, and mucosal structure loss. This study evaluated the beneficial role of purple potato (PP) supplementation against IBD symptoms using a murine model of dextran sulfate sodium (DSS)-induced colitis, and further explored the underlying mechanisms. Six-week-old C57BL/6J male mice were randomized into two groups and fed a standard rodent diet with or without 10% PP powder for 7 weeks. At the 5th week of dietary supplements, mice in each group were further divided into two subgroups and were either induced with or without 2.5% DSS induction for 7 days, followed by 7 days of recovery. Data showed that PP supplementation ameliorated the disease activity index in DSS-treated mice and reversed the colonic structure loss, mucosal damage, macrophage infiltration, and pro-inflammatory cytokine secretion induced by DSS in the colonic tissue. PP supplementation also restored the levels of tight junction proteins and caudal type homeobox 2 in DSS-treated mice. Furthermore, dietary PP enhanced peroxisome proliferator-activated receptor-γ coactivator-1α signaling pathway, mitochondrial biogenesis, mitochondrial proteostasis, and protein-folding capacity. In summary, dietary PP ameliorated DSS-induced colitis and improved gut structures and barrier function, which was associated with improved mitochondrial function. These results support further investigation of PP as a potential dietary intervention for IBD.
Collapse
Affiliation(s)
- Qi Sun
- School of Food Science, Washington State University, Pullman, WA, USA
| | - Shima Bibi
- School of Food Science, Washington State University, Pullman, WA, USA
| | - Yansong Xue
- School of Food Science, Washington State University, Pullman, WA, USA
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Boon Chew
- School of Food Science, Washington State University, Pullman, WA, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, USA.
| |
Collapse
|
4
|
Jiang W, Hu T, Ye C, Hu M, Yu Q, Sun L, Liang J, Chen Y. Formononetin attenuates high glucose-induced neurotoxicity by negatively regulating oxidative stress and mitochondrial dysfunction in Schwann cells via activation of SIRT3. Food Chem Toxicol 2023; 182:114156. [PMID: 37944786 DOI: 10.1016/j.fct.2023.114156] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023]
Abstract
High glucose induces Schwann cells death and neurotoxicity. Formononetin was originally found in Astragalus membranaceus and showed anti-tumor and anti-neuroinflammation properties. The aim of this study is to explore the molecular mechanism underlying the neuroprotective effects of formononetin and identify its direct protein target. The effects of formononetin on oxidative stress and mitochondrial dysfunction in Schwann cells induced by high glucose were investigated. High glucose treatment significantly induced oxidative stress, mitochondrial dysfunction and apoptosis in Schwann cells, while these effects were partially or completely prevented by co-treatment with formononetin. Mechanistically, we found that SIRT3/PGC-1α/SOD2 pathway was activated by formononetin under high glucose conditions as evidenced by western blotting. Knockdown of SIRT3 by siRNA delivery reversed the protective effects of formononetin on high glucose-induced Schwann cells injury and changes in expression profile of SIRT3 downstream target genes. Molecular docking, thermal shift assay and surface plasmon resonance assay revealed a direct binding between formononetin and SIRT3. Taken together, we identified a novel SIRT3 activator formononetin and revealed its beneficial effects on high glucose-induced neurotoxicity, suggesting that targeting SIRT3 in Schwann cells may be a new approach for treatment of peripheral nerve regeneration related diseases such as diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Wen Jiang
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Hubei University, Wuhan, 430062, China
| | - Ting Hu
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Hubei University, Wuhan, 430062, China
| | - Chen Ye
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Hubei University, Wuhan, 430062, China
| | - Man Hu
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Hubei University, Wuhan, 430062, China
| | - Qingqing Yu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Lijuan Sun
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Hubei University, Wuhan, 430062, China
| | - Jichao Liang
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Hubei University, Wuhan, 430062, China.
| | - Yong Chen
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
5
|
Nagahisa T, Kosugi S, Yamaguchi S. Interactions between Intestinal Homeostasis and NAD + Biology in Regulating Incretin Production and Postprandial Glucose Metabolism. Nutrients 2023; 15:nu15061494. [PMID: 36986224 PMCID: PMC10052115 DOI: 10.3390/nu15061494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
The intestine has garnered attention as a target organ for developing new therapies for impaired glucose tolerance. The intestine, which produces incretin hormones, is the central regulator of glucose metabolism. Glucagon-like peptide-1 (GLP-1) production, which determines postprandial glucose levels, is regulated by intestinal homeostasis. Nicotinamide phosphoribosyltransferase (NAMPT)-mediated nicotinamide adenine dinucleotide (NAD+) biosynthesis in major metabolic organs such as the liver, adipose tissue, and skeletal muscle plays a crucial role in obesity- and aging-associated organ derangements. Furthermore, NAMPT-mediated NAD+ biosynthesis in the intestines and its upstream and downstream mediators, adenosine monophosphate-activated protein kinase (AMPK) and NAD+-dependent deacetylase sirtuins (SIRTs), respectively, are critical for intestinal homeostasis, including gut microbiota composition and bile acid metabolism, and GLP-1 production. Thus, boosting the intestinal AMPK-NAMPT-NAD+-SIRT pathway to improve intestinal homeostasis, GLP-1 production, and postprandial glucose metabolism has gained significant attention as a novel strategy to improve impaired glucose tolerance. Herein, we aimed to review in detail the regulatory mechanisms and importance of intestinal NAMPT-mediated NAD+ biosynthesis in regulating intestinal homeostasis and GLP-1 secretion in obesity and aging. Furthermore, dietary and molecular factors regulating intestinal NAMPT-mediated NAD+ biosynthesis were critically explored to facilitate the development of new therapeutic strategies for postprandial glucose dysregulation.
Collapse
Affiliation(s)
- Taichi Nagahisa
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shotaro Kosugi
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shintaro Yamaguchi
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
6
|
Sun Q, Bravo Iniguez A, Tian Q, Du M, Zhu MJ. PGC-1α in mediating mitochondrial biogenesis and intestinal epithelial differentiation promoted by purple potato extract. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
7
|
Guerbette T, Boudry G, Lan A. Mitochondrial function in intestinal epithelium homeostasis and modulation in diet-induced obesity. Mol Metab 2022; 63:101546. [PMID: 35817394 PMCID: PMC9305624 DOI: 10.1016/j.molmet.2022.101546] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
Background Systemic low-grade inflammation observed in diet-induced obesity has been associated with dysbiosis and disturbance of intestinal homeostasis. This latter relies on an efficient epithelial barrier and coordinated intestinal epithelial cell (IEC) renewal that are supported by their mitochondrial function. However, IEC mitochondrial function might be impaired by high fat diet (HFD) consumption, notably through gut-derived metabolite production and fatty acids, that may act as metabolic perturbators of IEC. Scope of review This review presents the current general knowledge on mitochondria, before focusing on IEC mitochondrial function and its role in the control of intestinal homeostasis, and featuring the known effects of nutrients and metabolites, originating from the diet or gut bacterial metabolism, on IEC mitochondrial function. It then summarizes the impact of HFD on mitochondrial function in IEC of both small intestine and colon and discusses the possible link between mitochondrial dysfunction and altered intestinal homeostasis in diet-induced obesity. Major conclusions HFD consumption provokes a metabolic shift toward fatty acid β-oxidation in the small intestine epithelial cells and impairs colonocyte mitochondrial function, possibly through downstream consequences of excessive fatty acid β-oxidation and/or the presence of deleterious metabolites produced by the gut microbiota. Decreased levels of ATP and concomitant O2 leaks into the intestinal lumen could explain the alterations of intestinal epithelium dynamics, barrier disruption and dysbiosis that contribute to the loss of epithelial homeostasis in diet-induced obesity. However, the effect of HFD on IEC mitochondrial function in the small intestine remains unknown and the precise mechanisms by which HFD induces mitochondrial dysfunction in the colon have not been elucidated so far.
Collapse
Affiliation(s)
| | - Gaëlle Boudry
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France.
| | - Annaïg Lan
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France
| |
Collapse
|
8
|
Berberine remodels adipose tissue to attenuate metabolic disorders by activating sirtuin 3. Acta Pharmacol Sin 2022; 43:1285-1298. [PMID: 34417576 PMCID: PMC9061715 DOI: 10.1038/s41401-021-00736-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022]
Abstract
Adipose tissue remodelling is considered a critical pathophysiological hallmark of obesity and related metabolic diseases. Berberine (BBR), a natural isoquinoline alkaloid, has potent anti-hyperlipidaemic and anti-hyperglycaemic effects. This study aimed to explore the role of BBR in modulating adipose tissue remodelling and the underlying mechanisms. BBR protected high fat diet (HFD)-fed mice against adiposity, insulin resistance and hyperlipidemia. BBR alleviated adipose tissue inflammation and fibrosis by inhibiting macrophage infiltration, pro-inflammatory macrophage polarization and the abnormal deposition of extracellular matrix, and the effect was mediated by BBR directly binding and activating the deacetylase Sirtuin 3 (SIRT3) and suppressing the activation of the mitogen-activated protein kinases and nuclear factor-κB signalling pathways. Furthermore, BBR decreased microRNA-155-5p secretion by macrophages, which in turn ameliorated liver injury. Moreover, BBR mitigated inflammatory responses in both LPS-stimulated macrophages and TNF-α-treated adipocytes and suppressed macrophage migration towards adipocytes by activating SIRT3. Collectively, this study revealed that BBR improved adipose tissue remodelling, and subsequently inhibited the secretion of microRNA-155-5p by macrophages, which alleviated adiposity, insulin resistance and liver injury in obese mice. The modulation of adipose tissue remodelling by activating SIRT3 could contribute to the anti-hyperlipidemic and anti-hyperglycemic effects of BBR.
Collapse
|
9
|
Arora P, Andersen D, Moll JM, Danneskiold-Samsøe NB, Xu L, Zhou B, Kladis G, Rausch P, Workman CT, Kristiansen K, Brix S. Small Intestinal Tuft Cell Activity Associates With Energy Metabolism in Diet-Induced Obesity. Front Immunol 2021; 12:629391. [PMID: 34122403 PMCID: PMC8195285 DOI: 10.3389/fimmu.2021.629391] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/26/2021] [Indexed: 01/06/2023] Open
Abstract
Little is known about the involvement of type 2 immune response-promoting intestinal tuft cells in metabolic regulation. We here examined the temporal changes in small intestinal tuft cell number and activity in response to high-fat diet-induced obesity in mice and investigated the relation to whole-body energy metabolism and the immune phenotype of the small intestine and epididymal white adipose tissue. Intake of high fat diet resulted in a reduction in overall numbers of small intestinal epithelial and tuft cells and reduced expression of the intestinal type 2 tuft cell markers Il25 and Tslp. Amongst >1,700 diet-regulated transcripts in tuft cells, we observed an early association between body mass expansion and increased expression of the gene encoding the serine protease inhibitor neuroserpin. By contrast, tuft cell expression of genes encoding gamma aminobutyric acid (GABA)-receptors was coupled to Tslp and Il25 and reduced body mass gain. Combined, our results point to a possible role for small intestinal tuft cells in energy metabolism via coupled regulation of tuft cell type 2 markers and GABA signaling receptors, while being independent of type 2 immune cell involvement. These results pave the way for further studies into interventions that elicit anti-obesogenic circuits via small intestinal tuft cells.
Collapse
Affiliation(s)
- Pankaj Arora
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Daniel Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Janne Marie Moll
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Liqin Xu
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
- BGI-Shenzhen, Shenzhen, China
| | | | - Georgios Kladis
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Philipp Rausch
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Christopher T. Workman
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- BGI-Shenzhen, Shenzhen, China
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
10
|
Quiñones M, Hernández-Bautista R, Beiroa D, Heras V, Torres-Leal FL, Lam BYH, Senra A, Fernø J, Gómez-Valadés AG, Schwaninger M, Prevot V, Yeo G, Claret M, López M, Diéguez C, Al-Massadi O, Nogueiras R. Sirt3 in POMC neurons controls energy balance in a sex- and diet-dependent manner. Redox Biol 2021; 41:101945. [PMID: 33744652 PMCID: PMC8005845 DOI: 10.1016/j.redox.2021.101945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuin homologs of the yeast Sir2 gene that has emerged as an important player in the regulation of energy metabolism in peripheral tissues. However, its role in the hypothalamus has not been explored. Herein, we show that the genetic inhibition of SIRT3 in the hypothalamic arcuate nucleus (ARC) induced a negative energy balance and improvement of several metabolic parameters. These effects are specific for POMC neurons, because ablation of SIRT3 in POMC, but not in AgRP neurons, decreased body weight and adiposity, increased energy expenditure and brown adipose tissue (BAT) activity, and induced browning in white adipose tissue (WAT). Notably, the depletion of SIRT3 in POMC neurons caused these effects in male mice fed a chow diet but failed to affect energy balance in males fed a high fat diet and females under both type of diets. Overall, we provide the first evidence pointing for a key role of SIRT3 in POMC neurons in the regulation of energy balance.
Collapse
Affiliation(s)
- Mar Quiñones
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| | - René Hernández-Bautista
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Daniel Beiroa
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Violeta Heras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Francisco L Torres-Leal
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; Metabolic Diseases, Exercise and Nutrition (DOMEN) Research Group, Federal University of Piauí, Teresina, Brazil
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Ana Senra
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Alicia García Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), F-59000, Lille, France
| | - Giles Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036, Barcelona, Spain; School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Omar Al-Massadi
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana S/n, 15706, Santiago de Compostela, Spain.
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
11
|
Han D, Wang B, Cui X, He W, zhang Y, Jiang Q, Wang F, Liu Z, Shen D. ICS II protects against cardiac hypertrophy by regulating metabolic remodelling, not by inhibiting autophagy. J Cell Mol Med 2021. [PMCID: PMC7812268 DOI: 10.1111/jcmm.16175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cardiac hypertrophy is characterized by a shift in metabolic substrate utilization. Therefore, the regulation of ketone body uptake and metabolism may have beneficial effects on heart injuries that induce cardiac remodelling. In this study, we investigated whether icariside II (ICS II) protects against cardiac hypertrophy in mice and cardiomyocytes. To create cardiac hypertrophy animal and cell models, mice were subjected to transverse aortic constriction (TAC), and embryonic rat cardiomyocytes (H9C2) were stimulated with angiotensin II, a neurohumoral stressor. Both the in vivo and in vitro results suggest that ICS II treatment ameliorated pressure overload–induced cardiac hypertrophy and preserved heart function. In addition, apoptosis and oxidative stress were reduced in the presence of ICS II. Moreover, ICS II inhibited excess autophagy in TAC‐induced hearts and angiotensin II–stimulated cardiomyocytes. Mechanistically, we found that ICS II administration regulated SIRT3 expression in cardiac remodelling. SIRT3 activation increased ketone body transportation and utilization. Collectively, our data show that ICS II attenuated cardiac hypertrophy by modulating ketone body and fatty acid metabolism, and that this was likely due to the activation of the SIRT3‐AMPK pathway. ICS II treatment may provide a new therapeutic strategy for improving myocardial metabolism in cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Dongjian Han
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Bo Wang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Xinyue Cui
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Weiwei He
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Yi zhang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Qingjiao Jiang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Fuhang Wang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Zhiyu Liu
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Deliang Shen
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| |
Collapse
|
12
|
Wallenius V, Elias E, Elebring E, Haisma B, Casselbrant A, Larraufie P, Spak E, Reimann F, le Roux CW, Docherty NG, Gribble FM, Fändriks L. Suppression of enteroendocrine cell glucagon-like peptide (GLP)-1 release by fat-induced small intestinal ketogenesis: a mechanism targeted by Roux-en-Y gastric bypass surgery but not by preoperative very-low-calorie diet. Gut 2020; 69:1423-1431. [PMID: 31753852 PMCID: PMC7347417 DOI: 10.1136/gutjnl-2019-319372] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Food intake normally stimulates release of satiety and insulin-stimulating intestinal hormones, such as glucagon-like peptide (GLP)-1. This response is blunted in obese insulin resistant subjects, but is rapidly restored following Roux-en-Y gastric bypass (RYGB) surgery. We hypothesised this to be a result of the metabolic changes taking place in the small intestinal mucosa following the anatomical rearrangement after RYGB surgery, and aimed at identifying such mechanisms. DESIGN Jejunal mucosa biopsies from patients undergoing RYGB surgery were retrieved before and after very-low calorie diet, at time of surgery and 6 months postoperatively. Samples were analysed by global protein expression analysis and Western blotting. Biological functionality of these findings was explored in mice and enteroendocrine cells (EECs) primary mouse jejunal cell cultures. RESULTS The most prominent change found after RYGB was decreased jejunal expression of the rate-limiting ketogenic enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (mHMGCS), corroborated by decreased ketone body levels. In mice, prolonged high-fat feeding induced the expression of mHMGCS and functional ketogenesis in jejunum. The effect of ketone bodies on gut peptide secretion in EECs showed a ∼40% inhibition of GLP-1 release compared with baseline. CONCLUSION Intestinal ketogenesis is induced by high-fat diet and inhibited by RYGB surgery. In cell culture, ketone bodies inhibited GLP-1 release from EECs. Thus, we suggest that this may be a mechanism by which RYGB can remove the inhibitory effect of ketone bodies on EECs, thereby restituting the responsiveness of EECs resulting in increased meal-stimulated levels of GLP-1 after surgery.
Collapse
Affiliation(s)
- Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Sahlgrenska, University of Gothenburg, Gothenburg, Sweden .,Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Östra, University of Gothenburg, Gothenburg, Sweden
| | - Erik Elias
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Sahlgrenska, University of Gothenburg, Gothenburg, Sweden,Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Sahlgrenska, University of Gothenburg, Gothenburg, Sweden
| | - Erik Elebring
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Sahlgrenska, University of Gothenburg, Gothenburg, Sweden
| | - Bauke Haisma
- Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Anna Casselbrant
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Sahlgrenska, University of Gothenburg, Gothenburg, Sweden
| | - Pierre Larraufie
- Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Emma Spak
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Sahlgrenska, University of Gothenburg, Gothenburg, Sweden
| | - Frank Reimann
- Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, University College of Dublin, Dublin, Ireland
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute, University College of Dublin, Dublin, Ireland
| | - Fiona M Gribble
- Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Lars Fändriks
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital/Sahlgrenska, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
13
|
Schiffer TA, Lundberg JO, Weitzberg E, Carlström M. Modulation of mitochondria and NADPH oxidase function by the nitrate-nitrite-NO pathway in metabolic disease with focus on type 2 diabetes. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165811. [PMID: 32339643 DOI: 10.1016/j.bbadis.2020.165811] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria play fundamental role in maintaining cellular metabolic homeostasis, and metabolic disorders including type 2 diabetes (T2D) have been associated with mitochondrial dysfunction. Pathophysiological mechanisms are coupled to increased production of reactive oxygen species and oxidative stress, together with reduced bioactivity/signaling of nitric oxide (NO). Novel strategies restoring these abnormalities may have therapeutic potential in order to prevent or even treat T2D and associated cardiovascular and renal co-morbidities. A diet rich in green leafy vegetables, which contains high concentrations of inorganic nitrate, has been shown to reduce the risk of T2D. To this regard research has shown that in addition to the classical NO synthase (NOS) dependent pathway, nitrate from our diet can work as an alternative precursor for NO and other bioactive nitrogen oxide species via serial reductions of nitrate (i.e. nitrate-nitrite-NO pathway). This non-conventional pathway may act as an efficient back-up system during various pathological conditions when the endogenous NOS system is compromised (e.g. acidemia, hypoxia, ischemia, aging, oxidative stress). A number of experimental studies have demonstrated protective effects of nitrate supplementation in models of obesity, metabolic syndrome and T2D. Recently, attention has been directed towards the effects of nitrate/nitrite on mitochondrial functions including beiging/browning of white adipose tissue, PGC-1α and SIRT3 dependent AMPK activation, GLUT4 translocation and mitochondrial fusion-dependent improvements in glucose homeostasis, as well as dampening of NADPH oxidase activity. In this review, we examine recent research related to the effects of bioactive nitrogen oxide species on mitochondrial function with emphasis on T2D.
Collapse
Affiliation(s)
- Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Dávalos-Salas M, Montgomery MK, Reehorst CM, Nightingale R, Ng I, Anderton H, Al-Obaidi S, Lesmana A, Scott CM, Ioannidis P, Kalra H, Keerthikumar S, Tögel L, Rigopoulos A, Gong SJ, Williams DS, Yoganantharaja P, Bell-Anderson K, Mathivanan S, Gibert Y, Hiebert S, Scott AM, Watt MJ, Mariadason JM. Deletion of intestinal Hdac3 remodels the lipidome of enterocytes and protects mice from diet-induced obesity. Nat Commun 2019; 10:5291. [PMID: 31757939 PMCID: PMC6876593 DOI: 10.1038/s41467-019-13180-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 10/23/2019] [Indexed: 12/22/2022] Open
Abstract
Histone deacetylase 3 (Hdac3) regulates the expression of lipid metabolism genes in multiple tissues, however its role in regulating lipid metabolism in the intestinal epithelium is unknown. Here we demonstrate that intestine-specific deletion of Hdac3 (Hdac3IKO) protects mice from diet induced obesity. Intestinal epithelial cells (IECs) from Hdac3IKO mice display co-ordinate induction of genes and proteins involved in mitochondrial and peroxisomal β-oxidation, have an increased rate of fatty acid oxidation, and undergo marked remodelling of their lipidome, particularly a reduction in long chain triglycerides. Many HDAC3-regulated fatty oxidation genes are transcriptional targets of the PPAR family of nuclear receptors, Hdac3 deletion enhances their induction by PPAR-agonists, and pharmacological HDAC3 inhibition induces their expression in enterocytes. These findings establish a central role for HDAC3 in co-ordinating PPAR-regulated lipid oxidation in the intestinal epithelium, and identify intestinal HDAC3 as a potential therapeutic target for preventing obesity and related diseases. Histone deacetylase 3 (HDAC3) is a regulator of lipid homeostasis in several tissues, however, its role in intestinal lipid metabolism was not yet known. Here the authors study intestine specific HDAC3 knock out mice and report that these animals have increased fatty acid oxidation and undergo remodeling of the intestinal epithelial cell lipidome.
Collapse
Affiliation(s)
- Mercedes Dávalos-Salas
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Magdalene K Montgomery
- Department of Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Camilla M Reehorst
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Rebecca Nightingale
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Irvin Ng
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Holly Anderton
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Sheren Al-Obaidi
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Analia Lesmana
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Cameron M Scott
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Paul Ioannidis
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Hina Kalra
- La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Shivakumar Keerthikumar
- La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Lars Tögel
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Angela Rigopoulos
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Sylvia J Gong
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia
| | - David S Williams
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia.,Department of Pathology, Austin Health, Melbourne, Victoria, Australia
| | | | - Kim Bell-Anderson
- Faculty of Science, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Suresh Mathivanan
- La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Yann Gibert
- Department of Medicine, Deakin University, Geelong, Victoria, Australia
| | | | - Andrew M Scott
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia.,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Matthew J Watt
- Department of Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia.
| | - John M Mariadason
- Olivia Newton John Cancer Research Institute, Melbourne, Victoria, Australia. .,La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia. .,Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
15
|
Langhans W. Serendipity and spontaneity - Critical components in 40 years of academia. Physiol Behav 2019; 204:76-85. [PMID: 30753847 DOI: 10.1016/j.physbeh.2019.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 10/27/2022]
Abstract
I was flattered and felt tremendously honored to receive the 2018 Distinguished Career Award (DCA) from SSIB, the society that I always considered my scientific home, my family. Preparing the award lecture, I reflected about defining features of my career. This paper summarizes this very personal retrospective. As you will read, serendipity and more or less spontaneous decisions; i.e., some luck to be in the right place at the right time, and spontaneity to grab an opportunity when it presented itself, played a major role, and not necessarily a thorough analysis of my life situation at various junctions of my career path. Luck also often had the name of a fantastic tutor or mentor, or came in the form of enlightening discussions with a friend. Science is teamwork, which emphasizes how important collaborators, post-docs, students and technicians are. Although deep thinking was not necessarily crucial for my career path, a thorough examination is of course necessary when analyzing data, which were often most important when they did not confirm my hypothesis. Science is also hard work considering how much time one spends, but it never seemed like work to me because I had always this desire to find out how things in the organism work, and I always felt privileged to be able to pursue my "hobby" and even get a decent pay for it. In short, being a scientist is probably one of the most rewarding professional activities that life can offer.
Collapse
Affiliation(s)
- Wolfgang Langhans
- Physiology and Behavior Laboratory, Department of Health Sciences and Technology, ETH Zurich, Schorenstr. 16, 8603 Schwerzenbach, Switzerland.
| |
Collapse
|
16
|
Kaufman S, Arnold M, Diaz AA, Neubauer H, Wolfrum S, Köfeler H, Langhans W, Krieger JP. Roux-en-Y gastric bypass surgery reprograms enterocyte triglyceride metabolism and postprandial secretion in rats. Mol Metab 2019; 23:51-59. [PMID: 30905616 PMCID: PMC6480308 DOI: 10.1016/j.molmet.2019.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/03/2019] [Accepted: 03/07/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Roux-en-Y gastric bypass (RYGB) surgery produces rapid and persistent reductions in plasma triglyceride (TG) levels associated with fewer cardiovascular events. The mechanisms of the reduction in systemic TG levels remain unclear. We hypothesized that RYGB reduces intestinal TG secretion via altered enterocyte lipid handling. METHODS RYGB or Sham surgery was performed in diet-induced obese, insulin-resistant male Sprague-Dawley rats. First, we tested whether RYGB reduced test meal-induced TG levels in the intestinal lymph, a direct readout of enterocyte lipid secretion. Second, we examined whether RYGB modified TG enterocyte secretion at the single lipid level and in comparison to other lipid subclasses, applying mass spectrometry lipidomics to the intestinal lymph of RYGB and Sham rats (0-21 days after surgery). Third, we explored whether RYGB modulated the metabolic characteristics of primary enterocytes using transcriptional and functional assays relevant to TG absorption, reesterification, storage in lipid droplets, and oxidation. RESULTS RYGB reduced overall postprandial TG concentrations compared to Sham surgery in plasma and intestinal lymph similarly. RYGB reduced lymphatic TG concentrations more than other lipid subclasses, and shifted the remaining TG pool towards long-chain, unsaturated species. In enterocytes of fasted RYGB rats, lipid uptake was transcriptionally (Fatp4, Fabp2, Cd36) and functionally reduced compared to Sham, whereas TG reesterification genes were upregulated. CONCLUSION Our results show that RYGB substantially reduces intestinal TG secretion and modifies enterocyte lipid absorption and handling in rats. These changes likely contribute to the improvements in the plasma TG profile observed after RYGB in humans.
Collapse
Affiliation(s)
- Sharon Kaufman
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland.
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | | | - Heike Neubauer
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, 88397 Biberach/Riss, Germany
| | - Susanne Wolfrum
- Laboratory of Organic Chemistry, ETH Zurich, Zurich, Switzerland
| | - Harald Köfeler
- Core Facility Mass Spectrometry Lipidomics Research Center Graz, Austria
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | | |
Collapse
|
17
|
Wang T, Wang G, Zhang Y, Zhang J, Cao W, Chen X. Effect of lentivirus-mediated overexpression or silencing of MnSOD on apoptosis of resveratrol-treated fibroblast-like synoviocytes in rheumatoid arthritis. Eur J Pharmacol 2019; 844:65-72. [DOI: 10.1016/j.ejphar.2018.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022]
|
18
|
Striatal dopamine 2 receptor upregulation during development predisposes to diet-induced obesity by reducing energy output in mice. Proc Natl Acad Sci U S A 2018; 115:10493-10498. [PMID: 30254156 DOI: 10.1073/pnas.1800171115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dopaminergic signaling in the striatum, particularly at dopamine 2 receptors (D2R), has been a topic of active investigation in obesity research in the past decades. However, it still remains unclear whether variations in striatal D2Rs modulate the risk for obesity and if so in which direction. Human studies have yielded contradictory findings that likely reflect a complex nonlinear relationship, possibly involving a combination of causal effects and compensatory changes. Animal work indicates that although chronic obesogenic diets reduce striatal D2R function, striatal D2R down-regulation does not lead to obesity. In this study, we evaluated the consequences of striatal D2R up-regulation on body-weight gain susceptibility and energy balance in mice. We used a mouse model of D2R overexpression (D2R-OE) in which D2Rs were selectively up-regulated in striatal medium spiny neurons. We uncover a pathological mechanism by which striatal D2R-OE leads to reduced brown adipose tissue thermogenesis, reduced energy expenditure, and accelerated obesity despite reduced eating. We also show that D2R-OE restricted to development is sufficient to promote obesity and to induce energy-balance deficits. Together, our findings indicate that striatal D2R-OE during development persistently increases the propensity for obesity by reducing energy output in mice. This suggests that early alterations in the striatal dopamine system could represent a key predisposition factor toward obesity.
Collapse
|
19
|
Ramachandran D, Clara R, Fedele S, Michel L, Burkard J, Kaufman S, Diaz AA, Weissfeld N, De Bock K, Prip-Buus C, Langhans W, Mansouri A. Enhancing enterocyte fatty acid oxidation in mice affects glycemic control depending on dietary fat. Sci Rep 2018; 8:10818. [PMID: 30018405 PMCID: PMC6050244 DOI: 10.1038/s41598-018-29139-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 07/06/2018] [Indexed: 12/15/2022] Open
Abstract
Studies indicate that modulating enterocyte metabolism might affect whole body glucose homeostasis and the development of diet-induced obesity (DIO). We tested whether enhancing enterocyte fatty acid oxidation (FAO) could protect mice from DIO and impaired glycemic control. To this end, we used mice expressing a mutant form of carnitine palmitoyltransferase-1a (CPT1mt), insensitive to inhibition by malonyl-CoA, in their enterocytes (iCPT1mt) and fed them low-fat control diet (CD) or high-fat diet (HFD) chronically. CPT1mt expression led to an upregulation of FAO in the enterocytes. On CD, iCPT1mt mice had impaired glycemic control and showed concomitant activation of lipogenesis, glycolysis and gluconeogenesis in their enterocytes. On HFD, both iCPT1mt and control mice developed DIO, but iCPT1mt mice showed improved glycemic control and reduced visceral fat mass. Together these data indicate that modulating enterocyte metabolism in iCPT1mt mice affects glycemic control in a body weight-independent, but dietary fat-dependent manner.
Collapse
Affiliation(s)
| | - Rosmarie Clara
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Shahana Fedele
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Ladina Michel
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Johannes Burkard
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Sharon Kaufman
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | | | - Nadja Weissfeld
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Katrien De Bock
- Excercise and Health Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Carina Prip-Buus
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR, 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Abdelhak Mansouri
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|