1
|
Bailoo JD, Bergeson SE, Ponomarev I, Willms JO, Kisby BR, Cornwall GA, MacDonald CC, Lawrence JJ, Ganapathy V, Sivaprakasam S, Panthagani P, Trasti S, Varholick JA, Findlater M, Deonarine A. A bespoke water T-maze apparatus and protocol: an optimized, reliable, and repeatable method for screening learning, memory, and executive functioning in laboratory mice. Front Behav Neurosci 2024; 18:1492327. [PMID: 39720305 PMCID: PMC11666379 DOI: 10.3389/fnbeh.2024.1492327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/16/2024] [Indexed: 12/26/2024] Open
Abstract
The Morris Water Maze (MWM) is the most commonly used assay for evaluating learning and memory in laboratory mice. Despite its widespread use, contemporary reviews have highlighted substantial methodological variation in experimental protocols and that the associated testing procedures are acutely (each trial) and chronically (testing across days) stressful; stress impairs attention, memory consolidation and the retrieval of learned information. Moreover, the interpretation of behavior within the MWM is often difficult because of wall hugging, non-spatial swim strategies, floating, and jumping off the escape platform. Together, these issues may compromise the reproducibility, generalizability, and predictability of experimental results, as well as animal welfare. To address these issues, and as an initial proof-of-principle, we first narrowed the spatial dimensions of the MWM by using a T-insert, which constrained and reduced the overall length of time/distance that the animal must swim in order to navigate to the escape platform, thus reducing stress and off-task behavior. Given the robust performance observed across spatial acquisition (learning and memory) as well as during reversal learning (executive function), we further reduced (by 43%) the overall distance and time that the animal must swim in order to find the escape platform in a bespoke standalone Water T-Maze (WTM). We show, across five experiments, procedural refinements to our protocol and demonstrate robust, reliable and reproducible indicators of learning, memory and executive functioning in a task that is also significantly more efficient (3 days of testing within the WTM vs. 11 days of testing within the MWM). Taken together, our WTM apparatus and protocol are a significant improvement over other water-based apparatuses and protocols for evaluating learning, memory, and executive functioning in laboratory mice.
Collapse
Affiliation(s)
- Jeremy Davidson Bailoo
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Susan E. Bergeson
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Joshua O. Willms
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Brent R. Kisby
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Gail A. Cornwall
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Clinton C. MacDonald
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Sathish Sivaprakasam
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Praneetha Panthagani
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Scott Trasti
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Justin A. Varholick
- Department of Biology, University of Florida, Gainesville, FL, United States
| | - Michael Findlater
- Department of Chemistry and Biochemistry, University of California Merced, Merced, CA, United States
| | - Amrika Deonarine
- Department of Civil, Environmental, and Construction Engineering, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
2
|
Ambroziak W, Nencini S, Pohle J, Zuza K, Pino G, Lundh S, Araujo-Sousa C, Goetz LIL, Schrenk-Siemens K, Manoj G, Herrera MA, Acuna C, Siemens J. Thermally induced neuronal plasticity in the hypothalamus mediates heat tolerance. Nat Neurosci 2024:10.1038/s41593-024-01830-0. [PMID: 39653806 DOI: 10.1038/s41593-024-01830-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/25/2024] [Indexed: 12/19/2024]
Abstract
Heat acclimation is an adaptive process that improves physiological performance and supports survival in the face of increasing environmental temperatures, but the underlying mechanisms are not well understood. Here we identified a discrete group of neurons in the mouse hypothalamic preoptic area (POA) that rheostatically increase their activity over the course of heat acclimation, a property required for mice to become heat tolerant. In non-acclimated mice, peripheral thermoafferent pathways via the parabrachial nucleus activate POA neurons and mediate acute heat-defense mechanisms. However, long-term heat exposure promotes the POA neurons to gain intrinsically warm-sensitive activity, independent of thermoafferent parabrachial input. This newly gained cell-autonomous warm sensitivity is required to recruit peripheral heat tolerance mechanisms in acclimated animals. This pacemaker-like, warm-sensitive activity is driven by a combination of increased sodium leak current and enhanced utilization of the NaV1.3 ion channel. We propose that this salient neuronal plasticity mechanism adaptively drives acclimation to promote heat tolerance.
Collapse
Affiliation(s)
- Wojciech Ambroziak
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department of Translational Disease Understanding, Grünenthal GmbH, Aachen, Germany
| | - Sara Nencini
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Istituto Italiano di Tecnologia, Genoa, Italy
| | - Jörg Pohle
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department of Translational Disease Understanding, Grünenthal GmbH, Aachen, Germany
| | - Kristina Zuza
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Gabriela Pino
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Sofia Lundh
- Department of Pathology and Imaging, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Carolina Araujo-Sousa
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Larissa I L Goetz
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | | | - Gokul Manoj
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Mildred A Herrera
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Claudio Acuna
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Jan Siemens
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
3
|
Dos Santos BG, Brisnovali NF, Goedeke L. Biochemical basis and therapeutic potential of mitochondrial uncoupling in cardiometabolic syndrome. Biochem J 2024; 481:1831-1854. [PMID: 39630236 DOI: 10.1042/bcj20240005] [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: 08/05/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
Mild uncoupling of oxidative phosphorylation is an intrinsic property of all mitochondria, allowing for adjustments in cellular energy metabolism to maintain metabolic homeostasis. Small molecule uncouplers have been extensively studied for their potential to increase metabolic rate, and recent research has focused on developing safe and effective mitochondrial uncoupling agents for the treatment of obesity and cardiometabolic syndrome (CMS). Here, we provide a brief overview of CMS and cover the recent mechanisms by which chemical uncouplers regulate CMS-associated risk-factors and comorbidities, including dyslipidemia, insulin resistance, steatotic liver disease, type 2 diabetes, and atherosclerosis. Additionally, we review the current landscape of uncoupling agents, focusing on repurposed FDA-approved drugs and compounds in advanced preclinical or early-stage clinical development. Lastly, we discuss recent molecular insights by which chemical uncouplers enhance cellular energy expenditure, highlighting their potential as a new addition to the current CMS drug landscape, and outline several limitations that need to be addressed before these agents can successfully be introduced into clinical practice.
Collapse
Affiliation(s)
- Bernardo Gindri Dos Santos
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| | - Niki F Brisnovali
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| | - Leigh Goedeke
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
- Department of Medicine (Endocrinology), The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| |
Collapse
|
4
|
Wang R, Gomez Salazar M, Pruñonosa Cervera I, Coutts A, French K, Pinto MM, Gohlke S, García-Martín R, Blüher M, Schofield CJ, Kourtzelis I, Stimson RH, Bénézech C, Christian M, Schulz TJ, Gudmundsson EF, Jennings LL, Gudnason VG, Chavakis T, Morton NM, Emilsson V, Michailidou Z. Adipocyte deletion of the oxygen-sensor PHD2 sustains elevated energy expenditure at thermoneutrality. Nat Commun 2024; 15:7483. [PMID: 39209825 PMCID: PMC11362468 DOI: 10.1038/s41467-024-51718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Enhancing thermogenic brown adipose tissue (BAT) function is a promising therapeutic strategy for metabolic disease. However, predominantly thermoneutral modern human living conditions deactivate BAT. We demonstrate that selective adipocyte deficiency of the oxygen-sensor HIF-prolyl hydroxylase (PHD2) gene overcomes BAT dormancy at thermoneutrality. Adipocyte-PHD2-deficient mice maintain higher energy expenditure having greater BAT thermogenic capacity. In human and murine adipocytes, a PHD inhibitor increases Ucp1 levels. In murine brown adipocytes, antagonising the major PHD2 target, hypoxia-inducible factor-(HIF)-2a abolishes Ucp1 that cannot be rescued by PHD inhibition. Mechanistically, PHD2 deficiency leads to HIF2 stabilisation and binding of HIF2 to the Ucp1 promoter, thus enhancing its expression in brown adipocytes. Serum proteomics analysis of 5457 participants in the deeply phenotyped Age, Gene and Environment Study reveal that serum PHD2 associates with increased risk of metabolic disease. Here we show that adipose-PHD2-inhibition is a therapeutic strategy for metabolic disease and identify serum PHD2 as a disease biomarker.
Collapse
Affiliation(s)
- Rongling Wang
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mario Gomez Salazar
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Iris Pruñonosa Cervera
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Amanda Coutts
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Karen French
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Marlene Magalhaes Pinto
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Ruben García-Martín
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC (CNB-CSIC), Campus-UAM, Madrid, Spain
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research University of Oxford, Oxford, UK
| | - Ioannis Kourtzelis
- Hull York Medical School, York Biomedical Research Institute, University of York, York, UK
| | - Roland H Stimson
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark Christian
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | | | - Lori L Jennings
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Vilmundur G Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Triantafyllos Chavakis
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Nicholas M Morton
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Zoi Michailidou
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK.
| |
Collapse
|
5
|
Henckel MM, Chun JH, Knaub LA, Pott GB, James GE, Hunter KS, Shandas R, Walker LA, Reusch JEB, Keller AC. Perivascular adipose tissue remodeling impairs vasoreactivity in thermoneutral-housed rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593330. [PMID: 38798439 PMCID: PMC11118269 DOI: 10.1101/2024.05.09.593330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Objective Vascular pathology, characterized by impaired vasoreactivity and mitochondrial respiration, differs between the sexes. Housing rats under thermoneutral (TN) conditions causes vascular dysfunction and perturbed metabolism. We hypothesized that perivascular adipose tissue (PVAT), a vasoregulatory adipose depot with brown adipose tissue (BAT) phenotype, remodels to a white adipose (WAT) phenotype in rats housed at TN, driving diminished vasoreactivity in a sex-dependent manner. Methods Male and female Wistar rats were housed at either room temperature (RT) or TN. Endpoints included changes in PVAT morphology, vasoreactivity in vessels with intact PVAT or transferred to PVAT of the oppositely-housed animal, vessel stiffness, vessel mitochondrial respiration and cellular signaling. Results Remodeling of PVAT was observed in rats housed at TN; animals in this environment showed PVAT whitening and displayed diminished aortae vasodilation (p<0.05), different between the sexes. Juxtaposing PVAT from RT rats onto aortae from TN rats in females corrected vasodilation (p<0.05); this did not occur in males. In aortae of all animals housed at TN, mitochondrial respiration was significantly diminished in lipid substrate experiments (p<0.05), and there was significantly less expression of peNOS (p<0.001). Conclusions These data are consistent with TN-induced remodeling of PVAT, notably associated with sex-specific blunting of vasoreactivity, diminished mitochondrial respiration, and altered cellular signaling.
Collapse
Affiliation(s)
- Melissa M Henckel
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Ji Hye Chun
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Leslie A Knaub
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Gregory B Pott
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | | | - Kendall S Hunter
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Robin Shandas
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Lori A Walker
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jane E-B Reusch
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Amy C Keller
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| |
Collapse
|
6
|
Neri D, Ramos-Lobo AM, Lee S, Lafond A, Zeltser LM. Rearing mice at 22°C programs increased capacity to respond to chronic exposure to cold but not high fat diet. Mol Metab 2023; 73:101740. [PMID: 37211277 PMCID: PMC10248272 DOI: 10.1016/j.molmet.2023.101740] [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: 03/13/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023] Open
Abstract
OBJECTIVE Rodent models raised at environmental temperatures of 21-22 °C are increasingly switched to thermoneutral housing conditions in adulthood to better capture human physiology. We quantified the developmental effects of rearing mice at an ambient temperature of 22 °C vs. 30 °C on metabolic responses to cold and high fat diet (HFD) in adulthood. METHODS Mice were reared from birth to 8 weeks of age at 22 °C or 30 °C, when they were acclimated to single housing at the same temperature for 2-3 weeks in indirect calorimetry cages. Energy expenditure attributable to basal metabolic rate, physical activity, thermic effect of food, and adaptive cold- or diet-induced thermogenesis was calculated. Responses to cooling were evaluated by decreasing the ambient temperature from 22 °C to 14 °C, while responses to HFD feeding were assessed at 30 °C. Influences of rearing temperature on thermogenic responses that emerge over hours, days and weeks were assessed by maintaining mice in the indirect calorimetry cages throughout the study. RESULTS At an ambient temperature of 22 °C, total energy expenditure (TEE) was 12-16% higher in mice reared at 22 °C as compared to 30 °C. Rearing temperature had no effect on responses in the first hours or week of the 14 °C challenge. Differences emerged in the third week, when TEE increased an additional 10% in mice reared at 22 °C, but mice reared at 30 °C could not sustain this level of cold-induced thermogenesis. Rearing temperature only affected responses to HFD during the first week, due to differences in the timing but not the strength of metabolic adaptations. CONCLUSION Rearing at 22 °C does not have a lasting effect on metabolic adaptations to HFD at thermoneutrality, but it programs an enhanced capacity to respond to chronic cold challenges in adulthood. These findings highlight the need to consider rearing temperature when using mice to model cold-induced thermogenesis.
Collapse
Affiliation(s)
- Daniele Neri
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Angela M Ramos-Lobo
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Seoeun Lee
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alexandre Lafond
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lori M Zeltser
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
7
|
Hylander BL, Qiao G, Cortes Gomez E, Singh P, Repasky EA. Housing temperature plays a critical role in determining gut microbiome composition in research mice: Implications for experimental reproducibility. Biochimie 2023; 210:71-81. [PMID: 36693616 PMCID: PMC10953156 DOI: 10.1016/j.biochi.2023.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Preclinical mouse models are widely used for studying mechanisms of disease and responses to therapeutics, however there is concern about the lack of experimental reproducibility and failure to predict translational success. The gut microbiome has emerged as a regulator of metabolism and immunological processes in health and disease. The gut microbiome of mice differs by supplier and this affects experimental outcomes. We have previously reported that the mandated, mildly cool housing temperature for research mice (22°-26 °C) induces chronic adrenergic stress which suppresses anti-tumor immunity and promotes tumor growth compared to thermoneutral housing (30 °C). Therefore, we wondered how housing temperature affects the microbiome. Here, we demonstrate that the gut microbiome of BALB/c mice is easily modulated by a few degrees difference in temperature. Our results reveal significant differences between the gut microbiome of mice housed at 22°-23 °C vs. 30 °C. Although the genera vary, we consistently observed an enrichment of members of the family Lachnospiraceae when mice are housed at 22°-23 °C. These findings demonstrate that adrenergic stress and need for increased energy harvest to support thermogenesis, in addition to other factors such as diet, modulates the gut microbiome and this could be one mechanism by which housing temperature affects experimental outcomes. Additionally, tumor growth in mice housed at 30 °C also increases the proportion of Lachnospiraceae. The idea that stress can alter the gut microbiome and cause differences in experimental outcomes is applicable to mouse studies in general and is a variable that has significant potential to affect experimental reproducibility.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Guanxi Qiao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Eduardo Cortes Gomez
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Prashant Singh
- Genomics Shared Resource, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
8
|
Vialard F, Allaeys I, Dong G, Phan MP, Singh U, Hébert MJ, Dieudé M, Langlais D, Boilard E, Labbé DP, Olivier M. Thermoneutrality and severe malaria: investigating the effect of warmer environmental temperatures on the inflammatory response and disease progression. Front Immunol 2023; 14:1128466. [PMID: 37350957 PMCID: PMC10283000 DOI: 10.3389/fimmu.2023.1128466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Most studies using murine disease models are conducted at housing temperatures (20 - 22°C) that are sub-optimal (ST) for mice, eliciting changes in metabolism and response to disease. Experiments performed at a thermoneutral temperature (TT; 28 - 31°C) have revealed an altered immune response to pathogens and experimental treatments in murine disease model that have implications for their translation to clinical research. How such conditions affect the inflammatory response to infection with Plasmodium berghei ANKA (PbA) and disease progression is unknown. We hypothesized that changes in environmental temperature modulate immune cells and modify host response to malaria disease. To test this hypothesis, we conducted experiments to determine: (1) the inflammatory response to malarial agents injection in a peritonitis model and (2) disease progression in PbA-infected mice at TT compared to ST. Methods In one study, acclimatized mice were injected intraperitoneally with native hemozoin (nHZ) or Leishmania at TT (28 - 31°C) or ST, and immune cells, cytokine, and extracellular vesicle (EV) profiles were determined from the peritoneal cavity (PEC) fluid. In another study, PbA-infected mice were monitored until end-point (i.e. experimental malaria score ≥4). Results We found that Leishmania injection resulted in decreased cell recruitment and higher phagocytosis of nHZ in mice housed at TT. We found 398 upregulated and 293 downregulated proinflammatory genes in mice injected with nHZ, at both temperatures. We report the presence of host-derived EVs never reported before in a murine parasitic murine model at both temperatures. We observed metabolic changes in mice housed at TT, but these did not result to noticeable changes in disease progression compared to ST. Discussion To our knowledge, these experiments are the first to investigate the effect of thermoneutrality on a malaria murine model. We found important metabolic difference in mice housed at TT. Our results offer insights on how thermoneutrality might impact a severe malaria murine model and directions for more targeted investigations.
Collapse
Affiliation(s)
- Fiorella Vialard
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Isabelle Allaeys
- Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC, Canada
| | - George Dong
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Minh Phuong Phan
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Urvashi Singh
- Department of Human Genetics, McGill University Genome Centre, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Marie Josée Hébert
- Centre de Recherche, Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
| | - Mélanie Dieudé
- Centre de Recherche, Centre Hospitalier de l’Université de Montréal, Montréal, QC, Canada
- Département Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - David Langlais
- Department of Human Genetics, McGill University Genome Centre, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Eric Boilard
- Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC, Canada
| | - David P. Labbé
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Urology, Department of Surgery, McGill University, Montréal, QC, Canada
| | - Martin Olivier
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Zhang L, Koller J, Gopalasingam G, Herzog H. NPFF signalling is critical for thermosensory and dietary regulation of thermogenesis. Neuropeptides 2022; 96:102292. [PMID: 36155087 DOI: 10.1016/j.npep.2022.102292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
Thermogenesis is a centrally regulated physiological process integral for thermoregulation and energy homeostasis. However, the mechanisms and pathways involved remain poorly understood. Importantly, in this study we uncovered that in an environment of 28 °C that is within the mouse thermoneutral zone, lack of NPFF signalling leads to significant increases in energy expenditure, resting metabolic rate and brown adipose tissue (BAT) thermogenesis, which is associated with decreased body weight gain and lean tissue mass. Interestingly, when exposed to a high-fat diet (HFD) at 28 °C, Npff-/- mice lost the high energy expenditure phenotype observed under chow condition and exhibited an impaired diet-induced thermogenesis. On the other hand, under conditions of increasing levels of thermal demands, Npff-/- mice exhibited an elevated BAT thermogenesis at mild cold condition (22 °C), but initiated comparable BAT thermogenic responses as WT mice when thermal demand increased, such as an exposure to 4 °C. Together, these results reveal NPFF signalling as a novel and critical player in the control of thermogenesis, where it regulates thermosensory thermogenesis at warm condition and adjusts thermoregulation under positive energy balance to regulate diet-induced thermogenesis.
Collapse
Affiliation(s)
- Lei Zhang
- Neuroscience Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical Campus, School of Clinical Medicine, UNSW Medicine and Health, UNSW SYDNEY, NSW 2052, Australia.
| | - Julia Koller
- Neuroscience Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical Campus, School of Clinical Medicine, UNSW Medicine and Health, UNSW SYDNEY, NSW 2052, Australia
| | - Gopana Gopalasingam
- Neuroscience Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical Campus, School of Clinical Medicine, UNSW Medicine and Health, UNSW SYDNEY, NSW 2052, Australia
| |
Collapse
|
10
|
Keller AC, Chun JH, Knaub L, Henckel M, Hull S, Scalzo R, Pott G, Walker L, Reusch J. Thermoneutrality induces vascular dysfunction and impaired metabolic function in male Wistar rats: a new model of vascular disease. J Hypertens 2022; 40:2133-2146. [PMID: 35881464 PMCID: PMC9553250 DOI: 10.1097/hjh.0000000000003153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Cardiovascular disease is of paramount importance, yet there are few relevant rat models to investigate its pathology and explore potential therapeutics. Housing at thermoneutral temperature (30 °C) is being employed to humanize metabolic derangements in rodents. We hypothesized that housing rats in thermoneutral conditions would potentiate a high-fat diet, resulting in diabetes and dysmetabolism, and deleteriously impact vascular function, in comparison to traditional room temperature housing (22 °C). METHODS Male Wistar rats were housed at either room temperature or thermoneutral temperatures for 16 weeks on either a low or high-fat diet. Glucose and insulin tolerance tests were conducted at the beginning and end of the study. At the study's conclusion, vasoreactivity and mitochondrial respiration of aorta and carotid were conducted. RESULTS We observed diminished vasodilation in vessels from thermoneutral rats ( P < 0.05), whereas high-fat diet had no effect. This effect was also observed in endothelium-denuded aorta in thermoneutral rats ( P < 0.05). Vasoconstriction was significantly elevated in aorta of thermoneutral rats ( P < 0.05). Diminished nitric oxide synthase activity and nitrotyrosine, and elevated glutathione activity were observed in aorta from rats housed under thermoneutral conditions, indicating a climate of lower nitric oxide and excess reactive oxygen species in aorta. Thermoneutral rat aorta also demonstrated less mitochondrial respiration with lipid substrates compared with the controls ( P < 0.05). CONCLUSION Our data support that thermoneutrality causes dysfunctional vasoreactivity, decreased lipid mitochondrial metabolism, and modified cellular signaling. These are critical observations as thermoneutrality is becoming prevalent for translational research models. This new model of vascular dysfunction may be useful for dissection of targetable aspects of cardiovascular disease and is a novel and necessary model of disease.
Collapse
Affiliation(s)
- Amy C. Keller
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | | | - L.A. Knaub
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - M.M. Henckel
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - S.E. Hull
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - R.L. Scalzo
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - G.B. Pott
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - L.A. Walker
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - J.E.B. Reusch
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| |
Collapse
|
11
|
Chun JH, Henckel MM, Knaub LA, Hull SE, Pott GB, Ramirez DG, Reusch JEB, Keller AC. (-)-Epicatechin Reverses Glucose Intolerance in Rats Housed at Thermoneutrality. PLANTA MEDICA 2022; 88:735-744. [PMID: 35777366 PMCID: PMC9343939 DOI: 10.1055/a-1843-9855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/04/2022] [Indexed: 06/09/2023]
Abstract
Diabetes is a life-threatening and debilitating disease with pathological hallmarks, including glucose intolerance and insulin resistance. Plant compounds are a source of novel and effective therapeutics, and the flavonoid (-)-epicatechin, common to popular foods worldwide, has been shown to improve carbohydrate metabolism in both clinical studies and preclinical models. We hypothesized that (-)-epicatechin would alleviate thermoneutral housing-induced glucose intolerance. Male rats were housed at either thermoneutral (30 °C) or room temperature (24 °C) for 16 weeks and gavaged with either 1 mg/kg body weight or vehicle for the last 15 days before sacrifice. Rats housed at thermoneutrality had a significantly elevated serum glucose area under the curve (p < 0.05) and reduced glucose-mediated insulin secretion. In contrast, rats at thermoneutrality treated with (-)-epicatechin had improved glucose tolerance and increased insulin secretion (p < 0.05). Insulin tolerance tests revealed no differences in insulin sensitivity in any of the four groups. Pancreatic immunohistochemistry staining showed significantly greater islet insulin positive cells in animals housed at thermoneutrality. In conclusion, (-)-epicatechin improved carbohydrate tolerance via increased insulin secretion in response to glucose challenge without a change in insulin sensitivity.
Collapse
Affiliation(s)
- Ji Hye Chun
- Aquillius Corp., San Diego, CA, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Melissa M. Henckel
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Leslie A. Knaub
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Sara E. Hull
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Greg B. Pott
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - David G. Ramirez
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Jane E.-B. Reusch
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Amy C. Keller
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| |
Collapse
|
12
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
13
|
Chun JH, Henckel MM, Knaub LA, Hull SE, Pott GB, Walker LA, Reusch JEB, Keller AC. (-)-Epicatechin Improves Vasoreactivity and Mitochondrial Respiration in Thermoneutral-Housed Wistar Rat Vasculature. Nutrients 2022; 14:nu14051097. [PMID: 35268072 PMCID: PMC8912787 DOI: 10.3390/nu14051097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/12/2022] [Accepted: 02/19/2022] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular disease (CVD) is a global health concern. Vascular dysfunction is an aspect of CVD, and novel treatments targeting vascular physiology are necessary. In the endothelium, eNOS regulates vasodilation and mitochondrial function; both are disrupted in CVD. (−)-Epicatechin, a botanical compound known for its vasodilatory, eNOS, and mitochondrial-stimulating properties, is a potential therapy in those with CVD. We hypothesized that (−)-epicatechin would support eNOS activity and mitochondrial respiration, leading to improved vasoreactivity in a thermoneutral-derived rat model of vascular dysfunction. We housed Wistar rats at room temperature or in thermoneutral conditions for a total of 16 week and treated them with 1mg/kg body weight (−)-epicatechin for 15 day. Vasoreactivity, eNOS activity, and mitochondrial respiration were measured, in addition to the protein expression of upstream cellular signaling molecules including AMPK and CaMKII. We observed a significant improvement of vasodilation in those housed in thermoneutrality and treated with (−)-epicatechin (p < 0.05), as well as dampened mitochondrial respiration (p < 0.05). AMPK and CaMKIIα and β expression were lessened with (−)-epicatechin treatment in those housed at thermoneutrality (p < 0.05). The opposite was observed with animals housed at room temperature supplemented with (−)-epicatechin. These data illustrate a context-dependent vascular response to (−)-epicatechin, a candidate for CVD therapeutic development.
Collapse
Affiliation(s)
- Ji Hye Chun
- Microtek, Inc., San Diego, CA 92127, USA;
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA; (M.M.H.); (L.A.K.); (S.E.H.); (G.B.P.); (J.E.-B.R.)
| | - Melissa M. Henckel
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA; (M.M.H.); (L.A.K.); (S.E.H.); (G.B.P.); (J.E.-B.R.)
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Leslie A. Knaub
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA; (M.M.H.); (L.A.K.); (S.E.H.); (G.B.P.); (J.E.-B.R.)
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sara E. Hull
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA; (M.M.H.); (L.A.K.); (S.E.H.); (G.B.P.); (J.E.-B.R.)
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Greg B. Pott
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA; (M.M.H.); (L.A.K.); (S.E.H.); (G.B.P.); (J.E.-B.R.)
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lori A. Walker
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Jane E.-B. Reusch
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA; (M.M.H.); (L.A.K.); (S.E.H.); (G.B.P.); (J.E.-B.R.)
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amy C. Keller
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA; (M.M.H.); (L.A.K.); (S.E.H.); (G.B.P.); (J.E.-B.R.)
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Correspondence: ; Tel.: +1-303-724-3921
| |
Collapse
|
14
|
Li M, Speakman JR. Setting Ambient Temperature Conditions to Optimize Translation of Molecular Work from the Mouse to Human: The "Goldilocks Solution". Methods Mol Biol 2022; 2448:235-250. [PMID: 35167101 DOI: 10.1007/978-1-0716-2087-8_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Temperature has a profound effect on many aspects of murine physiology. This raises the question of the best temperature at which mice should be housed to maximize the translational potential to humans. The temperatures at which mice have been routinely kept for studies of molecular physiology (20-21 °C) maximize the comfort of animal handling staff. There is a widespread movement suggesting we should perform experiments instead on mice housed at 30 °C. This often produces very different outcomes. Here we analyze the basis of this suggestion and show that while 20-21 °C is too cold, 30 °C is probably too hot. Rather we suggest an intermediate temperature "the Goldilocks solution" of 25-26 °C is probably optimal. This should be combined with providing animals with nesting material so that they can construct nests to generate microclimates that are within their own control. Providing copious nesting material has additional spin-off advantages in terms of increasing environmental enrichment. Ultimately, however, advocating a single temperature to mimic human physiology is plagued by the problem that humans vary widely in the temperature environments they experience, with consequences for human disease. Hence studying responses at a range of temperatures may provide the greatest insights and translational potential.
Collapse
Affiliation(s)
- Min Li
- Shenzhen Key Laboratory for Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen, Institutes of Advanced Technology, Shenzhen, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
- School of Biological Sciences, University of Aberdeen, Scotland, UK
| | - John R Speakman
- Shenzhen Key Laboratory for Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen, Institutes of Advanced Technology, Shenzhen, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- University of the Chinese Academy of Sciences, Beijing, China.
- School of Biological Sciences, University of Aberdeen, Scotland, UK.
| |
Collapse
|
15
|
Brzęk P, Gębczyński A, Selewestruk P, Książek A, Sadowska J, Konarzewski M. Significance of variation in basal metabolic rate in laboratory mice for translational experiments. J Comp Physiol B 2021; 192:161-169. [PMID: 34595579 PMCID: PMC8816319 DOI: 10.1007/s00360-021-01410-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/24/2021] [Accepted: 09/20/2021] [Indexed: 12/03/2022]
Abstract
The basal metabolic rate (BMR) accounts for 60–70% of the daily energy expenditure (DEE) in sedentary humans and at least 50% of the DEE in laboratory mice in the thermoneutral zone. Surprisingly, however, the significance of the variation in the BMR is largely overlooked in translational research using such indices as physical activity level (PAL), i.e., the ratio of DEE/BMR. In particular, it is unclear whether emulation of human PAL in mouse models should be carried out within or below the thermoneutral zone. It is also unclear whether physical activity within the thermoneutral zone is limited by the capacity to dissipate heat generated by exercise and obligatory metabolic processes contributing to BMR. We measured PAL and spontaneous physical activity (SPA) in laboratory mice from two lines, divergently selected towards either high or low level of BMR, and acclimated to 30 °C (i.e., the thermoneutral zone), 23 or 4 °C. The mean PAL did not differ between both lines in the mice acclimated to 30 °C but became significantly higher in the low BMR mouse line at the lower ambient temperatures. Acclimation to 30 °C reduced the mean locomotor activity but did not affect the significant difference observed between the selected lines. We conclude that carrying out experiments within the thermoneutral zone can increase the consistency of translational studies aimed at the emulation of human energetics, without affecting the variation in physical activity correlated with BMR.
Collapse
Affiliation(s)
- Paweł Brzęk
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245, Białystok, Poland.
| | - Andrzej Gębczyński
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245, Białystok, Poland
| | - Piotr Selewestruk
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245, Białystok, Poland
| | - Aneta Książek
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245, Białystok, Poland
| | - Julita Sadowska
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245, Białystok, Poland
| | - Marek Konarzewski
- Faculty of Biology, University of Białystok, Ciołkowskiego 1J, 15-245, Białystok, Poland
| |
Collapse
|
16
|
Löffler MC, Betz MJ, Blondin DP, Augustin R, Sharma AK, Tseng YH, Scheele C, Zimdahl H, Mark M, Hennige AM, Wolfrum C, Langhans W, Hamilton BS, Neubauer H. Challenges in tackling energy expenditure as obesity therapy: From preclinical models to clinical application. Mol Metab 2021; 51:101237. [PMID: 33878401 PMCID: PMC8122111 DOI: 10.1016/j.molmet.2021.101237] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A chronic imbalance of energy intake and energy expenditure results in excess fat storage. The obesity often caused by this overweight is detrimental to the health of millions of people. Understanding both sides of the energy balance equation and their counter-regulatory mechanisms is critical to the development of effective therapies to treat this epidemic. SCOPE OF REVIEW Behaviors surrounding ingestion have been reviewed extensively. This review focuses more specifically on energy expenditure regarding bodyweight control, with a particular emphasis on the organs and attractive metabolic processes known to reduce bodyweight. Moreover, previous and current attempts at anti-obesity strategies focusing on energy expenditure are highlighted. Precise measurements of energy expenditure, which consist of cellular, animal, and human models, as well as measurements of their translatability, are required to provide the most effective therapies. MAJOR CONCLUSIONS A precise understanding of the components surrounding energy expenditure, including tailored approaches based on genetic, biomarker, or physical characteristics, must be integrated into future anti-obesity treatments. Further comprehensive investigations are required to define suitable treatments, especially because the complex nature of the human perspective remains poorly understood.
Collapse
Affiliation(s)
- Mona C Löffler
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Matthias J Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Denis P Blondin
- Department of Medicine, Division of Neurology, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, QC, Canada
| | - Robert Augustin
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Anand K Sharma
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Section on Integrative Physiology and Metabolism, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Heike Zimdahl
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Michael Mark
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Anita M Hennige
- Therapeutic Area CardioMetabolism & Respiratory, Boehringer Ingelheim International GmbH, Biberach, Germany
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | - Bradford S Hamilton
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Heike Neubauer
- Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany.
| |
Collapse
|
17
|
Cheong LY, Xu A. Intercellular and inter-organ crosstalk in browning of white adipose tissue: molecular mechanism and therapeutic complications. J Mol Cell Biol 2021; 13:466-479. [PMID: 34185049 PMCID: PMC8530522 DOI: 10.1093/jmcb/mjab038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/27/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022] Open
Abstract
Adipose tissue (AT) is highly plastic and heterogeneous in response to environmental and nutritional changes. The development of heat-dissipating beige adipocytes in white AT (WAT) through a process known as browning (or beiging) has garnered much attention as a promising therapeutic strategy for obesity and its related metabolic complications. This is due to its inducibility in response to thermogenic stimulation and its association with improved metabolic health. WAT consists of adipocytes, nerves, vascular endothelial cells, various types of immune cells, adipocyte progenitor cells, and fibroblasts. These cells contribute to the formation of beige adipocytes through the release of protein factors that significantly influence browning capacity. In addition, inter-organ crosstalk is also important for beige adipocyte biogenesis. Here, we summarize recent findings on fat depot-specific differences, secretory factors participating in intercellular and inter-organ communications that regulate the recruitment of thermogenic beige adipocytes, as well as challenges in targeting beige adipocytes as a potential anti-obese therapy.
Collapse
Affiliation(s)
- Lai Yee Cheong
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
18
|
Moreno-Fernandez ME, Sharma V, Stankiewicz TE, Oates JR, Doll JR, Damen MSMA, Almanan MATA, Chougnet CA, Hildeman DA, Divanovic S. Aging mitigates the severity of obesity-associated metabolic sequelae in a gender independent manner. Nutr Diabetes 2021; 11:15. [PMID: 34099626 PMCID: PMC8184786 DOI: 10.1038/s41387-021-00157-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 05/10/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Understanding gender-associated bias in aging and obesity-driven metabolic derangements has been hindered by the inability to model severe obesity in female mice. METHODS Here, using chow- or high fat diet (HFD)-feeding regimens at standard (TS) and thermoneutral (TN) housing temperatures, the latter to model obesity in female mice, we examined the impact of gender and aging on obesity-associated metabolic derangements and immune responsiveness. Analysis included quantification of: (i) weight gain and adiposity; (ii) the development and severity of glucose dysmetabolism and non-alcoholic fatty liver disease (NAFLD); and (iii) induction of inflammatory pathways related to metabolic dysfunction. RESULTS We show that under chow diet feeding regimen, aging was accompanied by increased body weight and white adipose tissue (WAT) expansion in a gender independent manner. HFD feeding regimen in aged, compared to young, male mice at TS, resulted in attenuated glucose dysmetabolism and hepatic steatosis. However, under TS housing conditions only aged, but not young, HFD fed female mice developed obesity. At TN however, both young and aged HFD fed female mice developed severe obesity. Independent of gender or housing conditions, aging attenuated the severity of metabolic derangements in HFD-fed obese mice. Tempered severity of metabolic derangements in aged mice was associated with increased splenic frequency of regulatory T (Treg) cells, Type I regulatory (Tr1)-like cells and circulating IL-10 levels and decreased vigor of HFD-driven induction of inflammatory pathways in adipose and liver tissues. CONCLUSION Our findings suggest that aging-associated altered immunological profile and inflammatory vigor may play a dominant role in the attenuation of obesogenic diet-driven metabolic dysfunction.
Collapse
Affiliation(s)
- Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Vishakha Sharma
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Jarren R Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - Jessica R Doll
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Maha A T A Almanan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - Claire A Chougnet
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - David A Hildeman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Center for Transplant Immunology, and Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center Cincinnati, Ohio, 45229, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA.
- Division of Immunobiology Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA.
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA.
| |
Collapse
|
19
|
Zolkipli-Cunningham Z, Naviaux JC, Nakayama T, Hirsch CM, Monk JM, Li K, Wang L, Le TP, Meinardi S, Blake DR, Naviaux RK. Metabolic and behavioral features of acute hyperpurinergia and the maternal immune activation mouse model of autism spectrum disorder. PLoS One 2021; 16:e0248771. [PMID: 33735311 PMCID: PMC7971557 DOI: 10.1371/journal.pone.0248771] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Since 2012, studies in mice, rats, and humans have suggested that abnormalities in purinergic signaling may be a final common pathway for many genetic and environmental causes of autism spectrum disorder (ASD). The current study in mice was conducted to characterize the bioenergetic, metabolomic, breathomic, and behavioral features of acute hyperpurinergia triggered by systemic injection of the purinergic agonist and danger signal, extracellular ATP (eATP). Responses were studied in C57BL/6J mice in the maternal immune activation (MIA) model and controls. Basal metabolic rates and locomotor activity were measured in CLAMS cages. Plasma metabolomics measured 401 metabolites. Breathomics measured 98 volatile organic compounds. Intraperitoneal eATP dropped basal metabolic rate measured by whole body oxygen consumption by 74% ± 6% (mean ± SEM) and rectal temperature by 6.2˚ ± 0.3˚C in 30 minutes. Over 200 metabolites from 37 different biochemical pathways where changed. Breathomics showed an increase in exhaled carbon monoxide, dimethylsulfide, and isoprene. Metabolomics revealed an acute increase in lactate, citrate, purines, urea, dopamine, eicosanoids, microbiome metabolites, oxidized glutathione, thiamine, niacinamide, and pyridoxic acid, and decreased folate-methylation-1-carbon intermediates, amino acids, short and medium chain acyl-carnitines, phospholipids, ceramides, sphingomyelins, cholesterol, bile acids, and vitamin D similar to some children with ASD. MIA animals were hypersensitive to postnatal exposure to eATP or poly(IC), which produced a rebound increase in body temperature that lasted several weeks before returning to baseline. Acute hyperpurinergia produced metabolic and behavioral changes in mice. The behaviors and metabolic changes produced by ATP injection were associated with mitochondrial functional changes that were profound but reversible.
Collapse
Affiliation(s)
- Zarazuela Zolkipli-Cunningham
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Jane C. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Tomohiro Nakayama
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Charlotte M. Hirsch
- Department of Chemistry, University of California, Irvine (UCI), Irvine, CA, United States of America
| | - Jonathan M. Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Thuy P. Le
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| | - Simone Meinardi
- Department of Chemistry, University of California, Irvine (UCI), Irvine, CA, United States of America
| | - Donald R. Blake
- Department of Chemistry, University of California, Irvine (UCI), Irvine, CA, United States of America
| | - Robert K. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA, United States of America
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, United States of America
| |
Collapse
|
20
|
Therapeutic potential of mitochondrial uncouplers for the treatment of metabolic associated fatty liver disease and NASH. Mol Metab 2021; 46:101178. [PMID: 33545391 PMCID: PMC8085597 DOI: 10.1016/j.molmet.2021.101178] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/18/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Background Mitochondrial uncouplers shuttle protons across the inner mitochondrial membrane via a pathway that is independent of adenosine triphosphate (ATP) synthase, thereby uncoupling nutrient oxidation from ATP production and dissipating the proton gradient as heat. While initial toxicity concerns hindered their therapeutic development in the early 1930s, there has been increased interest in exploring the therapeutic potential of mitochondrial uncouplers for the treatment of metabolic diseases. Scope of review In this review, we cover recent advances in the mechanisms by which mitochondrial uncouplers regulate biological processes and disease, with a particular focus on metabolic associated fatty liver disease (MAFLD), nonalcoholic hepatosteatosis (NASH), insulin resistance, and type 2 diabetes (T2D). We also discuss the challenges that remain to be addressed before synthetic and natural mitochondrial uncouplers can successfully enter the clinic. Major conclusions Rodent and non-human primate studies suggest that a myriad of small molecule mitochondrial uncouplers can safely reverse MAFLD/NASH with a wide therapeutic index. Despite this, further characterization of the tissue- and cell-specific effects of mitochondrial uncouplers is needed. We propose targeting the dosing of mitochondrial uncouplers to specific tissues such as the liver and/or developing molecules with self-limiting properties to induce a subtle and sustained increase in mitochondrial inefficiency, thereby avoiding systemic toxicity concerns.
Collapse
|
21
|
Vialard F, Olivier M. Thermoneutrality and Immunity: How Does Cold Stress Affect Disease? Front Immunol 2020; 11:588387. [PMID: 33329571 PMCID: PMC7714907 DOI: 10.3389/fimmu.2020.588387] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
One of the major challenges the scientific community faces today is the lack of translational data generated from mouse trials for human health application. Housing temperature-dependent chronic cold stress in laboratory rodents is one of the key factors contributing to lack of translatability because it reveals major metabolic differences between humans and rodents. While humans tend to operate at temperatures within their thermoneutral zone, most laboratory rodents are housed at temperatures below this zone and have an increased energy demand to generate heat. This has an impact on the immune system of mice and thus affects results obtained using murine models of human diseases. A limited number of studies and reviews have shown that results obtained on mice housed at thermoneutrality were different from those obtained from mice housed in traditional housing conditions. Most of those studies, focused on obesity and cancer, found that housing mice at thermoneutrality changed the outcomes of the diseases negatively and positively, respectively. In this review, we describe how thermoneutrality impacts the immune system of rodents generally and in the context of different disease models. We show that thermoneutrality exacerbates cardiovascular and auto-immune diseases; alleviates asthma and Alzheimer’s disease; and, changes gut microbiome populations. We also show that thermoneutrality can have exacerbating or alleviating effects on the outcome of infectious diseases. Thus, we join the call of others in this field to urge researchers to refine murine models of disease and increase their translational capacity by considering housing at thermoneutrality for trials involving rodents.
Collapse
Affiliation(s)
- Fiorella Vialard
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
22
|
Wee NKY, Nguyen AD, Enriquez RF, Zhang L, Herzog H, Baldock PA. Neuropeptide Y Regulation of Energy Partitioning and Bone Mass During Cold Exposure. Calcif Tissue Int 2020; 107:510-523. [PMID: 32804252 DOI: 10.1007/s00223-020-00745-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/04/2020] [Indexed: 10/23/2022]
Abstract
The maintenance of whole body energy homeostasis is critical to survival and mechanisms exist whereby an organism can adapt to its environment and the stresses placed upon it. Environmental temperature and thermogenesis are key components known to affect energy balance. However, little is known about how these processes are balanced against the overall energy balance. We show that even mild cold exposure has a significant effect on energy expenditure and UCP-1 levels which increase by 43% and 400%, respectively, when wild-type (WT) mice at thermoneutral (29 °C) were compared to mice at room temperature (22 °C) conditions. Interestingly, bone mass was lower in cold-stressed WT mice with significant reductions in femoral bone mineral content (- 19%) and bone volume (- 13%). Importantly, these cold-induced skeletal changes were absent in mice lacking NPY, one of the main controllers of energy homeostasis, highlighting the critical role of NPY in this process. However, energy expenditure was significantly greater in cold-exposed NPY null mice, indicating that suppression of non-thermogenic tissues, like bone, contributes to the adaptive responses to cold exposure. Altogether, this work identifies NPY as being crucial in coordinating energy and bone homeostasis where it suppresses energy expenditure, UCP-1 levels and lowers bone mass under conditions of cold exposure.
Collapse
Affiliation(s)
- Natalie K Y Wee
- Bone Biology Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Amy D Nguyen
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Ronaldo F Enriquez
- Bone Biology Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Lei Zhang
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
- School of Medical Sciences, University of NSW, Sydney, NSW, Australia
| | - Paul A Baldock
- Bone Biology Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
- School of Medical Sciences, University of NSW, Sydney, NSW, Australia.
- School of Medicine Sydney, University of Notre Dame Australia, Sydney, Australia.
| |
Collapse
|
23
|
Bastías-Pérez M, Serra D, Herrero L. Dietary Options for Rodents in the Study of Obesity. Nutrients 2020; 12:nu12113234. [PMID: 33105762 PMCID: PMC7690621 DOI: 10.3390/nu12113234] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/05/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity and its associated metabolic diseases are currently a priority research area. The increase in global prevalence at different ages is having an enormous economic and health impact. Genetic and environmental factors play a crucial role in the development of obesity, and diet is one of the main factors that contributes directly to the obesogenic phenotype. Scientific evidence has shown that increased fat intake is associated with the increase in body weight that triggers obesity. Rodent animal models have been extremely useful in the study of obesity since weight gain can easily be induced with a high-fat diet. Here, we review the dietary patterns and physiological mechanisms involved in the dynamics of energy balance. We report the main dietary options for the study of obesity and the variables to consider in the use of a high-fat diet, and assess the progression of obesity and diet-induced thermogenesis.
Collapse
Affiliation(s)
- Marianela Bastías-Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; (M.B.-P.); (D.S.)
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; (M.B.-P.); (D.S.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; (M.B.-P.); (D.S.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
24
|
Lang GP, Ndongson-Dongmo B, Lajqi T, Brodhun M, Han Y, Wetzker R, Frasch MG, Bauer R. Impact of ambient temperature on inflammation-induced encephalopathy in endotoxemic mice-role of phosphoinositide 3-kinase gamma. J Neuroinflammation 2020; 17:292. [PMID: 33028343 PMCID: PMC7541275 DOI: 10.1186/s12974-020-01954-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is an early and frequent event of infection-induced systemic inflammatory response syndrome. Phosphoinositide 3-kinase γ (PI3Kγ) is linked to neuroinflammation and inflammation-related microglial activity. In homeotherms, variations in ambient temperature (Ta) outside the thermoneutral zone lead to thermoregulatory responses, mainly driven by a gradually increasing sympathetic activity, and may affect disease severity. We hypothesized that thermoregulatory response to hypothermia (reduced Ta) aggravates SAE in PI3Kγ-dependent manner. METHODS Experiments were performed in wild-type, PI3Kγ knockout, and PI3Kγ kinase-dead mice, which were kept at neutral (30 ± 0.5 °C) or moderately lowered (26 ± 0.5 °C) Ta. Mice were exposed to lipopolysaccharide (LPS, 10 μg/g, from Escherichia coli serotype 055:B5, single intraperitoneal injection)-evoked systemic inflammatory response (SIR) and monitored 24 h for thermoregulatory response and blood-brain barrier integrity. Primary microglial cells and brain tissue derived from treated mice were analyzed for inflammatory responses and related cell functions. Comparisons between groups were made with one-way or two-way analysis of variance, as appropriate. Post hoc comparisons were made with the Holm-Sidak test or t tests with Bonferroni's correction for adjustments of multiple comparisons. Data not following normal distribution was tested with Kruskal-Wallis test followed by Dunn's multiple comparisons test. RESULTS We show that a moderate reduction of ambient temperature triggers enhanced hypothermia of mice undergoing LPS-induced systemic inflammation by aggravated SAE. PI3Kγ deficiency enhances blood-brain barrier injury and upregulation of matrix metalloproteinases (MMPs) as well as an impaired microglial phagocytic activity. CONCLUSIONS Thermoregulatory adaptation in response to ambient temperatures below the thermoneutral range exacerbates LPS-induced blood-brain barrier injury and neuroinflammation. PI3Kγ serves a protective role in suppressing release of MMPs, maintaining microglial motility and reinforcing phagocytosis leading to improved brain tissue integrity. Thus, preclinical research targeting severe brain inflammation responses is seriously biased when basic physiological prerequisites of mammal species such as preferred ambient temperature are ignored.
Collapse
Affiliation(s)
- Guang-Ping Lang
- Institute of Molecular Cell Biology, Jena University Hospital, Friedrich Schiller University, Hans-Knöll-Straße 2, D-07745 Jena, Germany
- Joint International Research Laboratory of Ethnomedicine and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, 563006 China
| | - Bernadin Ndongson-Dongmo
- Institute of Molecular Cell Biology, Jena University Hospital, Friedrich Schiller University, Hans-Knöll-Straße 2, D-07745 Jena, Germany
- Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Trim Lajqi
- Institute of Molecular Cell Biology, Jena University Hospital, Friedrich Schiller University, Hans-Knöll-Straße 2, D-07745 Jena, Germany
- Department of Neonatology, University Children’s Hospital, Heidelberg, Germany
| | - Michael Brodhun
- Department of Pathology, Helios-Klinikum Erfurt, Erfurt, Germany
| | - Yingying Han
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | | | - Reinhard Bauer
- Institute of Molecular Cell Biology, Jena University Hospital, Friedrich Schiller University, Hans-Knöll-Straße 2, D-07745 Jena, Germany
| |
Collapse
|
25
|
Deemer SE, Davis RAH, Roberts BM, Smith DL, Koutnik AP, Poff AM, D’Agostino DP, Plaisance EP. Exogenous Dietary Ketone Ester Decreases Body Weight and Adiposity in Mice Housed at Thermoneutrality. Obesity (Silver Spring) 2020; 28:1447-1455. [PMID: 32618116 PMCID: PMC7501155 DOI: 10.1002/oby.22855] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 04/06/2020] [Accepted: 04/14/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The aim of this study was to examine the effects of a ketone ester (KE)-supplemented diet on energy expenditure (EE) and adiposity in mice housed at 23 °C versus thermoneutrality (30 °C), in which sympathetic nervous system activity is diminished. METHODS Thirty-two 10-week-old male C57BL/6J mice were assigned to 1 of 4 groups (n = 8 per group): 30% KE diet + 23 °C (KE23), control (CON) diet + 23 °C (CON23), 30% KE diet + 30 °C (KE30), or CON diet + 30 °C (CON30). CON mice were pair-fed to the average intake of mice consuming the KE diet (ad libitum) for 8 weeks. Body composition and components of energy balance were measured at completion of the study. RESULTS CON23 (mean ± SD, 26.0 ± 1.6 g) and CON30 (29.7 ± 1.4 g) mice weighed more than KE groups (P < 0.03 for both) and were also different from each other (CON23 vs. CON30, P < 0.01). However, KE23 (23.4 ± 2.7 g) and KE30 (23.1 ± 1.9 g) mice were not different in body weight. As expected, food intake at 30 °C (2.0 ± 0.3 g/d) was lower than at 23 °C (2.6 ± 0.3 g/d, P < 0.01). Diet did not influence resting and total EE, but mice housed at 30 °C had lower EE compared with mice at 23 °C (P < 0.01). CONCLUSIONS Dietary KEs attenuate body weight gain at standard (23 °C) and thermoneutral (30 °C) housing temperatures, and this effect is not mediated by increased EE under these conditions.
Collapse
Affiliation(s)
- Sarah E. Deemer
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rachel A. H. Davis
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brandon M. Roberts
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew P. Koutnik
- Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Angela M. Poff
- Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | | | - Eric P. Plaisance
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
26
|
Abstract
Regulatory guidelines mandate housing for laboratory mice at temperatures below their thermoneutral zone, creating chronic cold stress. However, increases in housing temperature could alter immune responses. We hypothesized housing mice at temperatures within their thermoneutral zone would improve sepsis survival and alter immune responses. Male C57BL/6 mice were housed at 22°C or 30°C after cecal ligation and puncture (CLP) for 10 days. Survival of mice housed at 30°C (78%) after CLP was significantly increased compared with mice housed at 22°C (40%). Experimental groups were repeated with mice euthanized at 0, 12, 24, and 48 h post-surgery to examine select immune parameters. Raising housing temperature minimally altered systemic, peritoneal, or splenic cell counts. However, IL-6 levels in plasma and peritoneal lavage fluid were significantly lower at 12 h post-surgery in mice housed at 30°C compared with 22°C. Bacterial colony counts from peritoneal lavage fluid were significantly lower in mice housed at 30°C and in vivo studies suggested this was the result of increased phagocytosis by neutrophils. As previously demonstrated, adoptive transfer of fibrocytes significantly increased sepsis survival compared with saline at 22°C. However, there was no additive effect when adoptive transfer was performed at 30°C. Overall, the results demonstrated that thermoneutral housing improves survival after CLP by increasing local phagocytic activity and technical revisions may be necessary to standardize the severity of the model across different housing temperatures. These findings stress the pronounced impact housing temperature has on the CLP model and the importance of reporting housing temperature.
Collapse
|
27
|
Škop V, Guo J, Liu N, Xiao C, Hall KD, Gavrilova O, Reitman ML. Mouse Thermoregulation: Introducing the Concept of the Thermoneutral Point. Cell Rep 2020; 31:107501. [PMID: 32294435 PMCID: PMC7243168 DOI: 10.1016/j.celrep.2020.03.065] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/18/2019] [Accepted: 03/19/2020] [Indexed: 12/21/2022] Open
Abstract
Human and mouse thermal physiology differ due to dissimilar body sizes. Unexpectedly, in mice we found no ambient temperature zone where both metabolic rate and body temperature were constant. Body temperature began increasing once cold-induced thermogenesis was no longer required. This result reproduced in male, female, C57BL/6J, 129, chow-fed, diet-induced obese, and ob/ob mice as well as Trpv1-/-;Trpm8-/-;Trpa1-/- mice lacking thermal sensory channels. During the resting-light phase, the energy expenditure minimum spanned ∼4°C of ambient temperature, whereas in the active-dark phase it approximated a point. We propose the concept of a thermoneutral point (TNP), a discrete ambient temperature below which energy expenditure increases and above which body temperature increases. Humans do not have a TNP. As studied, the mouse TNP is ∼29°C in light phase and ∼33°C in dark phase. These observations inform how thermoneutrality is defined and how mice are used to model human energy physiology and drug development.
Collapse
Affiliation(s)
- Vojtěch Škop
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Juen Guo
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Naili Liu
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Cuiying Xiao
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Kevin D Hall
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Bastías-Pérez M, Zagmutt S, Soler-Vázquez MC, Serra D, Mera P, Herrero L. Impact of Adaptive Thermogenesis in Mice on the Treatment of Obesity. Cells 2020; 9:E316. [PMID: 32012991 PMCID: PMC7072509 DOI: 10.3390/cells9020316] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/20/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity and associated metabolic diseases have become a priority area of study due to the exponential increase in their prevalence and the corresponding health and economic impact. In the last decade, brown adipose tissue has become an attractive target to treat obesity. However, environmental variables such as temperature and the dynamics of energy expenditure could influence brown adipose tissue activity. Currently, most metabolic studies are carried out at a room temperature of 21 °C, which is considered a thermoneutral zone for adult humans. However, in mice this chronic cold temperature triggers an increase in their adaptive thermogenesis. In this review, we aim to cover important aspects related to the adaptation of animals to room temperature, the influence of housing and temperature on the development of metabolic phenotypes in experimental mice and their translation to human physiology. Mice studies performed in chronic cold or thermoneutral conditions allow us to better understand underlying physiological mechanisms for successful, reproducible translation into humans in the fight against obesity and metabolic diseases.
Collapse
Affiliation(s)
- Marianela Bastías-Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - M Carmen Soler-Vázquez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
29
|
Franco NH, Gerós A, Oliveira L, Olsson IAS, Aguiar P. ThermoLabAnimal – A high-throughput analysis software for non-invasive thermal assessment of laboratory mice. Physiol Behav 2019; 207:113-121. [DOI: 10.1016/j.physbeh.2019.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 11/28/2022]
|
30
|
McKie GL, Medak KD, Knuth CM, Shamshoum H, Townsend LK, Peppler WT, Wright DC. Housing temperature affects the acute and chronic metabolic adaptations to exercise in mice. J Physiol 2019; 597:4581-4600. [PMID: 31297830 DOI: 10.1113/jp278221] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/14/2019] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Mice are commonly housed at room temperatures below their thermoneutral zone meaning they are exposed to chronic thermal stress. Endurance exercise induces browning and mitochondrial biogenesis in white adipose tissue of rodents, but there are conflicting reports of this phenomenon in humans. We hypothesized that the ambient room temperature at which mice are housed could partially explain these discrepant reports between humans and rodents. We housed mice at room temperature or thermoneutrality and studied their physiological responses to acute and chronic exercise. We found that thermoneutral housing altered running behaviour and glucose homeostasis, and further, that exercise-induced markers of mitochondrial biogenesis and the browning of white adipose tissue were reduced in mice housed at thermoneutrality. ABSTRACT Mice are often housed at temperatures below their thermoneutral zone resulting in compensatory increases in thermogenesis. Despite this, many studies report housing mice at room temperature (RT), likely for the convenience of the researchers studying them. As such, the conflicting reports between humans and rodents regarding the ability of exercise to increase mitochondrial and thermogenic markers in white adipose tissue may be explained by the often-overlooked variable, housing temperature. To test this hypothesis, we housed male C57BL/6 mice at RT (22°C) or thermoneutrality (TN) (29°C) with or without access to a voluntary running wheel for 6 weeks or subjected them to an acute exhaustive bout of treadmill running. We examined the gene expression and protein content of select mitochondrial and thermogenic markers in skeletal muscle, epididymal white adipose tissue (eWAT), inguinal white adipose tissue (iWAT) and brown adipose tissue (BAT). We also assessed adipocyte morphology and indices of glucose homeostasis. Housing temperature influenced glucose tolerance and insulin action in vivo, yet the beneficial effects of exercise, both acute and chronic, remained intact in eWAT, BAT and skeletal muscle irrespective of housing temperature. Housing mice at TN led to an attenuation of some of the effects of exercise on iWAT. Collectively, we present data characterizing the acute and chronic metabolic adaptations to exercise at different housing temperatures and demonstrate, for the first time, that temperature influences the ability of exercise to increase markers of mitochondrial biogenesis and the browning of white adipose tissue.
Collapse
Affiliation(s)
- Greg L McKie
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Kyle D Medak
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Carly M Knuth
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Hesham Shamshoum
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Logan K Townsend
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Willem T Peppler
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - David C Wright
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
31
|
Woollard KJ, Murphy AJ. The Endless Summer: Thermoneutrality Prevents Monocytosis and Reduces Atherosclerosis. Circ Res 2019; 121:596-598. [PMID: 28860315 DOI: 10.1161/circresaha.117.311721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Kevin J Woollard
- From the Division of Immunology and Inflammation, Imperial College London, United Kingdom (K.J.W); and Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.)
| | - Andrew J Murphy
- From the Division of Immunology and Inflammation, Imperial College London, United Kingdom (K.J.W); and Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.J.M.)
| |
Collapse
|
32
|
Ndongson-Dongmo B, Lang GP, Mece O, Hechaichi N, Lajqi T, Hoyer D, Brodhun M, Heller R, Wetzker R, Franz M, Levy FO, Bauer R. Reduced ambient temperature exacerbates SIRS-induced cardiac autonomic dysregulation and myocardial dysfunction in mice. Basic Res Cardiol 2019; 114:26. [DOI: 10.1007/s00395-019-0734-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
|
33
|
Morton LD, Sanders M, Reagan WJ, Crabbs TA, McVean M, Funk KA. Confounding Factors in the Interpretation of Preclinical Studies. Int J Toxicol 2019; 38:228-234. [PMID: 30975012 DOI: 10.1177/1091581819837157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A number of issues may arise during the conduct of a study which can complicate interpretation of in vitro and in vivo datasets. Speakers discussed the implications of differing interpretations and how to avoid complicating factors during study planning and execution. Consideration needs to be given to study design factors including defining objectives, consideration of expected pharmacological effects, dose selection and drug kinetics, species used, and vehicle selection. In addition, the effects of vivarium temperature effects on various endpoints, how to control variables affecting clinical pathology, and how early death animals, common background findings, and artifacts can affect histopathology interpretation all play into the final interpretation of study data.
Collapse
Affiliation(s)
| | | | - William J Reagan
- 3 Pfizer Global Research and Development Eastern, Groton, CT, USA
| | | | - Maralee McVean
- 5 PreClinical Research Services, Inc, Fort Collins, CO, USA
| | | |
Collapse
|
34
|
Keijer J, Li M, Speakman JR. What is the best housing temperature to translate mouse experiments to humans? Mol Metab 2019; 25:168-176. [PMID: 31003945 PMCID: PMC6599456 DOI: 10.1016/j.molmet.2019.04.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Ambient temperature impinges on energy metabolism in a body size dependent manner. This has implications for the housing temperature at which mice are best compared to humans. In 2013, we suggested that, for comparative studies, solitary mice are best housed at 23-25 °C, because this is 3-5 °C below the mouse thermoneutral zone and humans routinely live 3-5 °C below thermoneutrality, and because this generates a ratio of DEE to BMR of 1.6-1.9, mimicking the ratio found in free-living humans. METHODS Recently, Fischer et al. (2017) challenged this estimate. By studying mice at 21 °C and at 30 °C (but notably not at 23-25 °C) they concluded that 30 °C is the optimal housing temperature. Here, we measured energy metabolism of C57BL/6 mice over a range of temperatures, between 21.4 °C and 30.2 °C. RESULTS We observed a ratio of DEE to BMR of 1.7 at 27.6 °C and of 1.8 at 25.5 °C, suggesting that this is the best temperature range for housing C57BL/6 mice to mimic human thermal relations. We used a 24 min average to calculate the ratio, similar to that used in human studies, while the ratio calculated by Fisher et al. dependent on short, transient metabolic declines. CONCLUSION We concur with Fisher et al. and others that 21 °C is too cool, but we continue to suggest that 30 °C is too warm. We support this with other data. Finally, to mimic living environments of all humans, and not just those in controlled Western environments, mouse experimentation at various temperatures is likely required.
Collapse
Affiliation(s)
- Jaap Keijer
- Human and Animal Physiology, Wageningen University, De Elst 1, PO box 338, 6700 AH, Wageningen, the Netherlands.
| | - Min Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK; CAS Centre of Excellence in Animal Evolution and Genetics, Kunming, China.
| |
Collapse
|
35
|
Conceição EPS, Madden CJ, Morrison SF. Neurons in the rat ventral lateral preoptic area are essential for the warm-evoked inhibition of brown adipose tissue and shivering thermogenesis. Acta Physiol (Oxf) 2019; 225:e13213. [PMID: 30365209 PMCID: PMC6686665 DOI: 10.1111/apha.13213] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/25/2018] [Accepted: 10/19/2018] [Indexed: 12/31/2022]
Abstract
AIM To determine the role of neurons in the ventral part of the lateral preoptic area (vLPO) in CNS thermoregulation. METHODS In vivo electrophysiological and neuropharmacological were used to evaluate the contribution of neurons in the vLPO to the regulation of brown adipose tissue (BAT) thermogenesis and muscle shivering in urethane/chloralose-anaesthetized rats. RESULTS Nanoinjections of NMDA targeting the medial preoptic area (MPA) and the vLPO suppressed the cold-evoked BAT sympathetic activity (SNA), reduced the BAT temperature (TBAT ), expired CO2 , mean arterial pressure (MAP), and heart rate. Inhibition of vLPO neurons with muscimol or AP5/CNQX elicited increases in BAT SNA, TBAT , tachycardia, and small elevations in MAP. The BAT thermogenesis evoked by AP5/CNQX in vLPO was inhibited by the activation of MPA neurons. The inhibition of BAT SNA by vLPO neurons does not require a GABAergic input to dorsomedial hypothalamus (DMH), but MPA provides a GABAergic input to DMH. The activation of vLPO neurons inhibits the BAT thermogenesis evoked by NMDA in the rostral raphe pallidus (rRPa), but not that after bicuculline in rRPa. The BAT thermogenesis elicited by vLPO inhibition is dependent on glutamatergic inputs to DMH and rRPa, but these excitatory inputs do not arise from MnPO neurons. The activation of neurons in the vLPO also inhibits cold- and prostaglandin-evoked muscle shivering, and vLPO inhibition is sufficient to evoke shivering. CONCLUSION The vLPO contains neurons that are required for the warm ambient-evoked inhibition of muscle shivering and of BAT thermogenesis, mediated through a direct or indirect GABAergic input to rRPa from vLPO.
Collapse
Affiliation(s)
- Ellen P S Conceição
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | - Christopher J Madden
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | - Shaun F Morrison
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
36
|
Madden CJ, Morrison SF. Central nervous system circuits that control body temperature. Neurosci Lett 2019; 696:225-232. [PMID: 30586638 PMCID: PMC6397692 DOI: 10.1016/j.neulet.2018.11.027] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/14/2018] [Accepted: 11/19/2018] [Indexed: 02/01/2023]
Abstract
Maintenance of mammalian core body temperature within a narrow range is a fundamental homeostatic process to optimize cellular and tissue function, and to improve survival in adverse thermal environments. Body temperature is maintained during a broad range of environmental and physiological challenges by central nervous system circuits that process thermal afferent inputs from the skin and the body core to control the activity of thermoeffectors. These include thermoregulatory behaviors, cutaneous vasomotion (vasoconstriction and, in humans, active vasodilation), thermogenesis (shivering and brown adipose tissue), evaporative heat loss (salivary spreading in rodents, and human sweating). This review provides an overview of the central nervous system circuits for thermoregulatory reflex regulation of thermoeffectors.
Collapse
Affiliation(s)
- Christopher J Madden
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States.
| | - Shaun F Morrison
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
37
|
Small L, Gong H, Yassmin C, Cooney GJ, Brandon AE. Thermoneutral housing does not influence fat mass or glucose homeostasis in C57BL/6 mice. J Endocrinol 2018; 239:313-324. [PMID: 30400016 DOI: 10.1530/joe-18-0279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022]
Abstract
One major factor affecting physiology often overlooked when comparing data from animal models and humans is the effect of ambient temperature. The majority of rodent housing is maintained at ~22°C, the thermoneutral temperature for lightly clothed humans. However, mice have a much higher thermoneutral temperature of ~30°C, consequently data collected at 22°C in mice could be influenced by animals being exposed to a chronic cold stress. The aim of this study was to investigate the effect of housing temperature on glucose homeostasis and energy metabolism of mice fed normal chow or a high-fat, obesogenic diet (HFD). Male C57BL/6J(Arc) mice were housed at standard temperature (22°C) or at thermoneutrality (29°C) and fed either chow or a 60% HFD for 13 weeks. The HFD increased fat mass and produced glucose intolerance as expected but this was not exacerbated in mice housed at thermoneutrality. Changing the ambient temperature, however, did alter energy expenditure, food intake, lipid content and glucose metabolism in skeletal muscle, liver and brown adipose tissue. Collectively, these findings demonstrate that mice regulate energy balance at different housing temperatures to maintain whole-body glucose tolerance and adiposity irrespective of the diet. Despite this, metabolic differences in individual tissues were apparent. In conclusion, dietary intervention in mice has a greater impact on adiposity and glucose metabolism than housing temperature although temperature is still a significant factor in regulating metabolic parameters in individual tissues.
Collapse
Affiliation(s)
- Lewin Small
- Diabetes and Metabolism Division, Garvan Institute, Sydney, New South Wales, Australia
| | - Henry Gong
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| | - Christian Yassmin
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| | - Gregory J Cooney
- Diabetes and Metabolism Division, Garvan Institute, Sydney, New South Wales, Australia
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| | - Amanda E Brandon
- Diabetes and Metabolism Division, Garvan Institute, Sydney, New South Wales, Australia
- The University of Sydney, School of Medical Sciences, Charles Perkins Centre, Sydney, New South Wales, Australia
| |
Collapse
|
38
|
Hankenson FC, Marx JO, Gordon CJ, David JM. Effects of Rodent Thermoregulation on Animal Models in the Research Environment. Comp Med 2018; 68:425-438. [PMID: 30458902 DOI: 10.30802/aalas-cm-18-000049] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
To best promote animal wellbeing and the efficacy of biomedical models, scientific, husbandry, and veterinary professionals must consider the mechanisms, influences, and outcomes of rodent thermoregulation in contemporary research environments. Over the last 2 decades, numerous studies have shown that laboratory mice and rats prefer temperatures that are several degrees warmer than the environments in which they typically are housed within biomedical facilities. Physiologic changes to rodents that are cage-housed under standard temperatures (20 to 26 °C) are attributed to 'cold stress' and include alterations in metabolism, cardiovascular parameters, respiration, and immunologic function. This review article describes common behavioral and physiologic adaptations of laboratory mice and rats to cold stress within modern vivaria, with emphasis on environmental enrichment and effects of anesthesia and procedural support efforts. In addition, potential interventions and outcomes for rodents are presented, relative to the importance of repeating and reproducing experiments involving laboratory rodent research models of human disease.
Collapse
Affiliation(s)
- F Claire Hankenson
- Campus Animal Resources, Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - James O Marx
- University Laboratory Animal Resources, Department of Pathobiology, School of Veterinary Medicine; University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher J Gordon
- Toxicity Assessment Division, Neurotoxicology Branch, United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - John M David
- Comparative Medicine, Pfizer, La Jolla, California, USA
| |
Collapse
|
39
|
Robbins A, Tom CATMB, Cosman MN, Moursi C, Shipp L, Spencer TM, Brash T, Devlin MJ. Low temperature decreases bone mass in mice: Implications for humans. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 167:557-568. [PMID: 30187469 DOI: 10.1002/ajpa.23684] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Humans exhibit significant ecogeographic variation in bone size and shape. However, it is unclear how significantly environmental temperature influences cortical and trabecular bone, making it difficult to recognize adaptation versus acclimatization in past populations. There is some evidence that cold-induced bone loss results from sympathetic nervous system activation and can be reduced by nonshivering thermogenesis (NST) via uncoupling protein (UCP1) in brown adipose tissue (BAT). Here we test two hypotheses: (1) low temperature induces impaired cortical and trabecular bone acquisition and (2) UCP1, a marker of NST in BAT, increases in proportion to degree of low-temperature exposure. METHODS We housed wildtype C57BL/6J male mice in pairs at 26 °C (thermoneutrality), 22 °C (standard), and 20 °C (cool) from 3 weeks to 6 or 12 weeks of age with access to food and water ad libitum (N = 8/group). RESULTS Cool housed mice ate more but had lower body fat at 20 °C versus 26 °C. Mice at 20 °C had markedly lower distal femur trabecular bone volume fraction, thickness, and connectivity density and lower midshaft femur cortical bone area fraction versus mice at 26 °C (p < .05 for all). UCP1 expression in BAT was inversely related to temperature. DISCUSSION These results support the hypothesis that low temperature was detrimental to bone mass acquisition. Nonshivering thermogenesis in brown adipose tissue increased in proportion to low-temperature exposure but was insufficient to prevent bone loss. These data show that chronic exposure to low temperature impairs bone architecture, suggesting climate may contribute to phenotypic variation in humans and other hominins.
Collapse
Affiliation(s)
- Amy Robbins
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | | | - Miranda N Cosman
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Cleo Moursi
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Lillian Shipp
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Taylor M Spencer
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Timothy Brash
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Maureen J Devlin
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
40
|
Kovalčíková A, Gyurászová M, Gardlík R, Boriš M, Celec P, Tóthová Ľ. The effects of sucrose on urine collection in metabolic cages. Lab Anim 2018; 53:180-189. [PMID: 30045671 DOI: 10.1177/0023677218781674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Representative urine collection that respects the standards of animal welfare is still an issue in experimental nephrology. The commonly used metabolic cages induce stress in rodents. In mice, the volume of collected urine is sometimes insufficient for further analysis. The aim of this experiment was to analyse the effects of time of day, temperature and 2%, 5% or 10% sucrose solutions on diuresis, weight change and liquid intake of adult mice placed in metabolic cages for urine collection. Mice were placed in metabolic cages for 12 h during the day or night at standard ambient (22℃) and thermoneutral (28℃) temperatures. To determine the effect of acclimatisation, mice were placed in metabolic cages for five consecutive days. Diuresis increased with concentrations of sucrose. Body weight reduction was most rapid in the group given tap water and decreased with increasing sucrose concentrations. A drastic drop in body weight was observed in mice placed in metabolic cages for four consecutive days with access to tap water and food, indicating that time spent in metabolic cages should be kept to a minimum, as prolonged confinement in metabolic cages can be harmful to mice. The administration of concentrated sucrose solutions can potentially aid in mouse urine collection by reducing the time spent in metabolic cages. Sucrose supplementation increased the albumin/creatinine ratio. However, without showing estimates of glomerular filtration rate, renal haemodynamics, plasma electrolytes and urinary electrolyte excretions, the results of this study do not provide any conclusion about the effect of sucrose on renal function.
Collapse
Affiliation(s)
- Alexandra Kovalčíková
- 1 Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Marianna Gyurászová
- 1 Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Roman Gardlík
- 1 Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,2 Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Martin Boriš
- 3 Institute of Electronics and Photonics, Faculty of Electrical Engineering and Information Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Peter Celec
- 1 Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,2 Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,4 Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Ľubomíra Tóthová
- 1 Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,5 Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
41
|
Brown SDM, Holmes CC, Mallon AM, Meehan TF, Smedley D, Wells S. High-throughput mouse phenomics for characterizing mammalian gene function. Nat Rev Genet 2018; 19:357-370. [PMID: 29626206 PMCID: PMC6582361 DOI: 10.1038/s41576-018-0005-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We are entering a new era of mouse phenomics, driven by large-scale and economical generation of mouse mutants coupled with increasingly sophisticated and comprehensive phenotyping. These studies are generating large, multidimensional gene-phenotype data sets, which are shedding new light on the mammalian genome landscape and revealing many hitherto unknown features of mammalian gene function. Moreover, these phenome resources provide a wealth of disease models and can be integrated with human genomics data as a powerful approach for the interpretation of human genetic variation and its relationship to disease. In the future, the development of novel phenotyping platforms allied to improved computational approaches, including machine learning, for the analysis of phenotype data will continue to enhance our ability to develop a comprehensive and powerful model of mammalian gene-phenotype space.
Collapse
Affiliation(s)
| | - Chris C Holmes
- Nuffield Department of Medicine and Department of Statistics, University of Oxford, Oxford, UK.
| | | | - Terrence F Meehan
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.
| | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Mesenchymal stem cells (MSCs) located in the bone marrow have the capacity to differentiate into multiple cell lineages, including osteoblast and adipocyte. Adipocyte density within marrow is inversely associated with bone mass during aging and in some pathological conditions, contributing to the prevailing view that marrow adipocytes play a largely negative role in bone metabolism. However, a negative association between marrow adipocytes and bone balance is not universal. Although MAT levels appear tightly regulated, establishing the precise physiological significance of MAT has proven elusive. Here, we review recent literature aimed at delineating the function of MAT. RECENT FINDINGS An important physiological function of MAT may be to provide an expandable/contractible fat depot, which is critical for minimization of energy requirements for sustaining optimal hematopoiesis. Because the energy requirements for storing fat are negligible compared to those required to maintain hematopoiesis, even small reductions in hematopoietic tissue volume to match a reduced requirement for hematopoiesis could represent an important reduction in energy cost. Such a physiological function would require tight coupling between hematopoietic stem cells and MSCs to regulate the balance between MAT and hematopoiesis. Kit-ligand, an important regulator of proliferation, differentiation, and survival of hematopoietic cells, may function as a prototypic factor coupling MAT and hematopoiesis. Crosstalk between hematopoietic and mesenchymal cells in the bone marrow may contribute to establishing the balance between MAT levels and hematopoiesis.
Collapse
Affiliation(s)
- Russell T Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA
| | - Stephen A Martin
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA.
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
43
|
Sepa-Kishi DM, Katsnelson G, Bikopoulos G, Iqbal A, Ceddia RB. Cold acclimation reduces hepatic protein Kinase B and AMP-activated protein kinase phosphorylation and increases gluconeogenesis in Rats. Physiol Rep 2018; 6:e13592. [PMID: 29504286 PMCID: PMC5835512 DOI: 10.14814/phy2.13592] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023] Open
Abstract
This study investigated the molecular and metabolic responses of the liver to cold-induced thermogenesis. To accomplish that, male Wistar rats were exposed to cold (4°C) for 7 days. Livers were then extracted and used for the determination of glucose and fatty acid oxidation, glycogen content, the expression and content of proteins involved in insulin signaling, as well as in the regulation of gluconeogenesis and de novo lipid synthesis. Despite being hyperphagic, cold-acclimated rats displayed normoglycemia with reduced insulinemia, which suggests improved whole-body insulin sensitivity. However, liver protein kinase B (AKT) and glycogen synthase kinase 3 (GSK3) phosphorylations were markedly reduced along with the expressions of the insulin receptor (IR) and its substrates IRS1 and IRS2, whereas glycogen synthase (GS) phosphorylation increased. Thus, major signaling steps of the glycogen synthesis pathway in the liver were inhibited. Furthermore, glucagonemia and hepatic glucose and fatty acid oxidation were increased, whereas liver glycogen content was reduced by cold acclimation. This was accompanied by significantly elevated expressions of the gluconeogenic transcription regulators CRTC2, PGC-1α, and FoxO1, as well as of major gluconeogenic enzymes (G6Pase, FBP1, and PEPCK). Conversely, phosphorylation and contents of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) content were markedly downregulated in livers of cold-acclimated rats. In conclusion, cold acclimation suppressed hepatic glycogen synthesis and promoted profound metabolic changes in the liver so the organ could sustain its ability to regulate whole-body glucose and lipid metabolism under conditions of high-energy demand in thermogenic tissues.
Collapse
Affiliation(s)
- Diane M Sepa-Kishi
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Glen Katsnelson
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - George Bikopoulos
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Ayesha Iqbal
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Rolando B Ceddia
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
44
|
Ohrnberger SA, Hambly C, Speakman JR, Valencak TG. Limits to sustained energy intake XXIX: the case of the golden hamster (Mesocricetus auratus). J Exp Biol 2018; 221:jeb.183749. [DOI: 10.1242/jeb.183749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/11/2018] [Indexed: 01/11/2023]
Abstract
Golden hamster females have the shortest known gestation period among placental mammals and at the same time raise very large litters of up to 16 offspring, which are born in a naked and blind state and are able to pick up food from days 12-14 only. We quantified energy metabolism and milk production in female golden hamsters raising offspring under cold (8°C), normal (22°C) and hot (30°C) ambient temperature conditions. We monitored energy intake, subcutaneous body temperature, daily energy expenditure, litter size and pup masses over the course of lactation. Our results show that, in line with the concept of heat dissipation limitation, female golden hamsters had the largest energy intake under the coldest conditions and a significantly lower intake at 30° (partial for influence of ambient temperature: F2,403=5.6; p= 0.004). Metabolisable energy intake as well as milk energy output showed the same pattern and were significantly different between the temperatures (partial for milk energy production: F1,40= 86.4; p<0.0001). With consistently higher subcutaneous temperatures in the reproductive females (F1,813= 36.77; p<0.0001) compared to baseline females. These data suggest that raising offspring in golden hamsters comes at the cost of producing large amounts of body heat up to a level constraining energy intake, similar to that observed in some laboratory mice. Notably, we observed that females seemed to adjust litter size according to their milk production with the smallest litters (3.4±0.7 pups) being raised by hot exposed mothers. Future research is needed to unravel the mechanism by which females assess their own milk production capabilities and how this may be linked to litter size at different ambient temperatures. Golden hamsters reach 8-10 times resting metabolic rate (RMR) when raising offspring under cold conditions, which is compatible with the findings from laboratory mice and other rodents.
Collapse
Affiliation(s)
- S. A. Ohrnberger
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| | - C. Hambly
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB 24 2 TZ, UK
| | - J. R. Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB 24 2 TZ, UK
- Institute of Genetics and Developmental Biology, State Key Laboratory of Molecular Developmental Biology, Chinese Academy of Sciences, Beichen Xi Lu, Chaoyang, Beijing, People's Republic of China
| | - T. G. Valencak
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| |
Collapse
|
45
|
Vaicik MK, Blagajcevic A, Ye H, Morse MC, Yang F, Goddi A, Brey EM, Cohen RN. The Absence of Laminin α4 in Male Mice Results in Enhanced Energy Expenditure and Increased Beige Subcutaneous Adipose Tissue. Endocrinology 2018; 159:356-367. [PMID: 28973559 PMCID: PMC5761598 DOI: 10.1210/en.2017-00186] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/05/2017] [Indexed: 01/27/2023]
Abstract
Laminin α4 (LAMA4) is located in the extracellular basement membrane that surrounds each individual adipocyte. Here we show that LAMA4 null (Lama4-/-) mice exhibit significantly higher energy expenditure (EE) relative to wild-type (WT) mice at room temperature and when exposed to a cold challenge, despite similar levels of food intake and locomotor activity. The Lama4-/- mice are resistant to age- and diet-induced obesity. Expression of uncoupling protein 1 is higher in subcutaneous white adipose tissue of Lama4-/- mice relative to WT animals on either a chow diet or a high-fat diet. In contrast, uncoupling protein 1 expression was not increased in brown adipose tissue. Lama4-/- mice exhibit significantly improved insulin sensitivity compared with WT mice, suggesting improved metabolic function. Overall, these data provide critical evidence for a role of the basement membrane in EE, weight gain, and systemic insulin sensitivity.
Collapse
Affiliation(s)
- Marcella K. Vaicik
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois 60141
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616
| | - Alen Blagajcevic
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Honggang Ye
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Mallory C. Morse
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Feipeng Yang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616
| | - Anna Goddi
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Eric M. Brey
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois 60141
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616
| | - Ronald N. Cohen
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
46
|
Cold acclimation causes fiber type-specific responses in glucose and fat metabolism in rat skeletal muscles. Sci Rep 2017; 7:15430. [PMID: 29133865 PMCID: PMC5684227 DOI: 10.1038/s41598-017-15842-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/02/2017] [Indexed: 01/03/2023] Open
Abstract
This study investigated fiber type-specific metabolic responses and the molecular mechanisms that regulate glucose and fat metabolism in oxidative and glycolytic muscles upon cold acclimation. Male Wistar rats were exposed to cold (4 °C) for 7 days, and then glycogen synthesis and content, glucose and palmitate oxidation, and the molecular mechanisms underlying these metabolic pathways were assessed in soleus (Sol), extensor digitorum longus (EDL), and epitrochlearis (Epit) muscles. Cold acclimation increased glycogen synthesis, glycogen content, glucose oxidation, and reduced glycogen synthase (GS) phosphorylation only in Sol muscles. Protein kinase B (AKT), glycogen synthase kinase 3 (GSK3), and AMP-activated protein kinase (AMPK) phosphorylation increased in all three muscles upon cold acclimation. Cold acclimation increased palmitate oxidation, gene expression of the transcriptional co-activator Pgc-1α, lipoprotein lipase (Lpl), fatty acid transporter (Cd36), and Sarco/endoplasmic reticulum Ca2+-ATPase (Serca) in Sol, EDL, and Epit muscles. Sarcolipin was only detected and had its content increased in Sol muscles. In conclusion, cold-induced thermogenesis activated similar signaling pathways in oxidative and glycolytic muscles, but the metabolic fate of glucose differed in skeletal muscles with distinct fiber type composition. Furthermore, only muscles rich in type I fibers appeared to have the capacity for sarcolipin-mediated SERCA uncoupling.
Collapse
|
47
|
Kato GA, Shichijo H, Takahashi T, Shinohara A, Morita T, Koshimoto C. Protein restriction does not affect body temperature pattern in female mice. Exp Anim 2017. [PMID: 28626157 PMCID: PMC5682344 DOI: 10.1538/expanim.17-0035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Daily torpor is a physiological adaptation in mammals and birds characterized by a
controlled reduction of metabolic rate and body temperature during the resting phase of
circadian rhythms. In laboratory mice, daily torpor is induced by dietary caloric
restriction. However, it is not known which nutrients are related to daily torpor
expression. To determine whether dietary protein is a key factor in inducing daily torpor
in mice, we fed mice a protein-restricted (PR) diet that included only one-quarter of the
amount of protein but the same caloric level as a control (C) diet. We assigned six
non-pregnant female ICR mice to each group and recorded their body weights and core body
temperatures for 4 weeks. Body weights in the C group increased, but those in the PR group
remained steady or decreased. Mice in both groups did not show daily torpor, but most mice
in a food-restricted group (n=6) supplied with 80% of the calories given to the C group
exhibited decreased body weights and frequently displayed daily torpor. This suggests that
protein restriction is not a trigger of daily torpor; torpid animals can conserve their
internal energy, but torpor may not play a significant role in conserving internal
protein. Thus, opportunistic daily torpor in mice may function in energy conservation
rather than protein saving.
Collapse
Affiliation(s)
- Goro A Kato
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan.,Division of Bio-resources, Department of Biotechnology, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan.,Present address: Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, University of Kyushu, 3-1-1 Maidashi, Fukuoka, Fukuoka 812-8582, Japan
| | - Hiroki Shichijo
- Division of Bio-resources, Department of Biotechnology, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan
| | - Toshihiro Takahashi
- Department of Animal and Grassland Sciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-Nishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Akio Shinohara
- Division of Bio-resources, Department of Biotechnology, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan
| | - Tetsuo Morita
- Department of Animal and Grassland Sciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-Nishi, Miyazaki, Miyazaki 889-2192, Japan
| | - Chihiro Koshimoto
- Division of Bio-resources, Department of Biotechnology, Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan
| |
Collapse
|
48
|
Gordon CJ. The mouse thermoregulatory system: Its impact on translating biomedical data to humans. Physiol Behav 2017; 179:55-66. [PMID: 28533176 PMCID: PMC6196327 DOI: 10.1016/j.physbeh.2017.05.026] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/04/2017] [Accepted: 05/18/2017] [Indexed: 01/01/2023]
Abstract
The laboratory mouse has become the predominant test species in biomedical research. The number of papers that translate or extrapolate data from mouse to human has grown exponentially since the year 2000. There are many physiological and anatomical factors to consider in the process of extrapolating data from one species to another. Body temperature is, of course, a critical determinant in extrapolation because it has a direct impact on metabolism, cardiovascular function, drug efficacy, pharmacokinetics of toxins and drugs, and many other effects. While most would consider the thermoregulatory system of mice to be sufficiently stable and predictable as to not be a cause for concern, the thermal physiology of mice does in fact present unique challenges to the biomedical researcher. A variable and unstable core temperature, high metabolic rate, preference for warm temperatures, large surface area: body mass ratio, and high rate of thermal conductance, are some of the key factors of mice that can affect the interpretation and translation of data to humans. It is the intent of this brief review to enlighten researchers studying interspecies translation of biomedical data on the salient facets of the mouse thermal physiology and show how extrapolation in fields such as physiology, psychology, nutrition, pharmacology, toxicology, and pathology.
Collapse
Affiliation(s)
- Christopher J Gordon
- Toxicity Assessment Division, National Health Effects and Environmental Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, United States.
| |
Collapse
|
49
|
Giles DA, Moreno-Fernandez ME, Stankiewicz TE, Graspeuntner S, Cappelletti M, Wu D, Mukherjee R, Chan CC, Lawson MJ, Klarquist J, Sünderhauf A, Softic S, Kahn CR, Stemmer K, Iwakura Y, Aronow BJ, Karns R, Steinbrecher KA, Karp CL, Sheridan R, Shanmukhappa SK, Reynaud D, Haslam DB, Sina C, Rupp J, Hogan SP, Divanovic S. Thermoneutral housing exacerbates nonalcoholic fatty liver disease in mice and allows for sex-independent disease modeling. Nat Med 2017; 23:829-838. [PMID: 28604704 PMCID: PMC5596511 DOI: 10.1038/nm.4346] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/22/2017] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a common prelude to cirrhosis and hepatocellular carcinoma, is the most common chronic liver disease worldwide. Defining the molecular mechanisms underlying the pathogenesis of NAFLD has been hampered by a lack of animal models that closely recapitulate the severe end of the disease spectrum in humans, including bridging hepatic fibrosis. Here we demonstrate that a novel experimental model employing thermoneutral housing, as opposed to standard housing, resulted in lower stress-driven production of corticosterone, augmented mouse proinflammatory immune responses and markedly exacerbated high-fat diet (HFD)-induced NAFLD pathogenesis. Disease exacerbation at thermoneutrality was conserved across multiple mouse strains and was associated with augmented intestinal permeability, an altered microbiome and activation of inflammatory pathways that are associated with the disease in humans. Depletion of Gram-negative microbiota, hematopoietic cell deletion of Toll-like receptor 4 (TLR4) and inactivation of the IL-17 axis resulted in altered immune responsiveness and protection from thermoneutral-housing-driven NAFLD amplification. Finally, female mice, typically resistant to HFD-induced obesity and NAFLD, develop full disease characteristics at thermoneutrality. Thus, thermoneutral housing provides a sex-independent model of exacerbated NAFLD in mice and represents a novel approach for interrogation of the cellular and molecular mechanisms underlying disease pathogenesis.
Collapse
Affiliation(s)
- Daniel A Giles
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Monica Cappelletti
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David Wu
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rajib Mukherjee
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Calvin C Chan
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Matthew J Lawson
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jared Klarquist
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Annika Sünderhauf
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Samir Softic
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Bruce J Aronow
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rebekah Karns
- Division of Gastroenterology Hepatology and Nutrition, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kris A Steinbrecher
- Division of Gastroenterology Hepatology and Nutrition, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Rachel Sheridan
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shiva K Shanmukhappa
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David B Haslam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christian Sina
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Senad Divanovic
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
50
|
Zuriaga MA, Fuster JJ, Gokce N, Walsh K. Humans and Mice Display Opposing Patterns of "Browning" Gene Expression in Visceral and Subcutaneous White Adipose Tissue Depots. Front Cardiovasc Med 2017; 4:27. [PMID: 28529941 PMCID: PMC5418233 DOI: 10.3389/fcvm.2017.00027] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/18/2017] [Indexed: 01/12/2023] Open
Abstract
Visceral adiposity is much more strongly associated with cardiometabolic disease in humans than subcutaneous adiposity. Browning, the appearance of brown-like adipocytes in the white adipose tissue (WAT), has been shown to protect mice against metabolic dysfunction, suggesting the possibility of new therapeutic approaches to treat obesity and type 2 diabetes. In mice, subcutaneous WAT depots express higher levels of browning genes when compared with visceral WAT, further suggesting that differences in WAT browning could contribute to the differences in the pathogenicity of the two depots. However, the expression of browning genes in different WAT depots of human has not been characterized. Here, it is shown that the expression of browning genes is higher in visceral than in subcutaneous WAT in humans, a pattern that is opposite to what is observed in mice. These results suggest that caution should be applied in extrapolating the results of murine browning gene expression studies to human pathophysiology.
Collapse
Affiliation(s)
- Maria A. Zuriaga
- Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Jose J. Fuster
- Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Noyan Gokce
- Cardiovascular Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Kenneth Walsh
- Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|