1
|
Furrer R, Handschin C. Biomarkers of aging: from molecules and surrogates to physiology and function. Physiol Rev 2025; 105:1609-1694. [PMID: 40111763 DOI: 10.1152/physrev.00045.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Many countries face an unprecedented challenge in aging demographics. This has led to an exponential growth in research on aging, which, coupled to a massive financial influx of funding in the private and public sectors, has resulted in seminal insights into the underpinnings of this biological process. However, critical validation in humans has been hampered by the limited translatability of results obtained in model organisms, additionally confined by the need for extremely time-consuming clinical studies in the ostensible absence of robust biomarkers that would allow monitoring in shorter time frames. In the future, molecular parameters might hold great promise in this regard. In contrast, biomarkers centered on function, resilience, and frailty are available at the present time, with proven predictive value for morbidity and mortality. In this review, the current knowledge of molecular and physiological aspects of human aging, potential antiaging strategies, and the basis, evidence, and potential application of physiological biomarkers in human aging are discussed.
Collapse
|
2
|
Newport ME, Wilson P, Lowes S, Behrends M, Coons A, Bowman J, Bates HE. Photoperiod influences visceral adiposity and the adipose molecular clock independent of temperature in wild-derived Peromyscus leucopus. FASEB Bioadv 2025; 7:e70006. [PMID: 40330430 PMCID: PMC12050962 DOI: 10.1096/fba.2024-00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 05/08/2025] Open
Abstract
Physiology is closely synchronized to daily and seasonal light/dark cycles. Humans artificially extend daylight and experience irregular light schedules, resulting in dysregulation of metabolism and body mass. In rodents, winter-like conditions (cold and short photoperiod) can alter energy balance and adipose tissue mass. To determine if photoperiod alone, independent of temperature, is a strong enough signal to regulate adiposity, we compared the effects of long and short photoperiod at thermoneutrality on adiposity and WAT gene expression in photoperiod-sensitive, F1 generation wild-derived adult male white-footed mice (Peromyscus leucopus). Mice were housed in long-day (16:8 light:dark) or short-day (8:16 light:dark) photoperiod conditions at thermoneutrality (27°C) for 4 weeks with the extended light being provided through artificial lighting. Photoperiod did not impact body weight or calorie consumption. However, mice housed in long photoperiod with extended artificial light selectively developed greater visceral WAT mass without changing subcutaneous WAT or interscapular BAT mass. This was accompanied by a decrease in Adrβ3 and Ucp1 mRNA expression in visceral WAT with no change in Pgc1a, Lpl, or Hsl. Expression of Per1, Per2, and Nr1d1 mRNA in visceral WAT differed between long and short photoperiods over time when aligned to circadian time but not onset of darkness, indicating alterations in clock gene expression with photoperiod. These findings suggest that extended photoperiod through artificial light can promote visceral fat accumulation alone, independent of temperature, supporting that artificial light may play a role in obesity.
Collapse
Affiliation(s)
| | - Paul Wilson
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Shanna Lowes
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Marthe Behrends
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Alexis Coons
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Jeff Bowman
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
- Wildlife Research and Monitoring SectionOntario Ministry of Natural ResourcesPeterboroughOntarioCanada
| | - Holly E. Bates
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| |
Collapse
|
3
|
Abd El Hay MY, Kamm GB, Tlaie Boria A, Siemens J. Diverging roles of TRPV1 and TRPM2 in warm-temperature detection. eLife 2025; 13:RP95618. [PMID: 40215103 PMCID: PMC11991700 DOI: 10.7554/elife.95618] [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] [Indexed: 04/14/2025] Open
Abstract
The perception of innocuous temperatures is crucial for thermoregulation. The TRP ion channels TRPV1 and TRPM2 have been implicated in warmth detection, yet their precise roles remain unclear. A key challenge is the low prevalence of warmth-sensitive sensory neurons, comprising fewer than 10% of rodent dorsal root ganglion (DRG) neurons. Using calcium imaging of >20,000 cultured mouse DRG neurons, we uncovered distinct contributions of TRPV1 and TRPM2 to warmth sensitivity. TRPV1's absence - and to a lesser extent absence of TRPM2 - reduces the number of neurons responding to warmth. Additionally, TRPV1 mediates the rapid, dynamic response to a warmth challenge. Behavioural tracking in a whole-body thermal preference assay revealed that these cellular differences shape nuanced thermal behaviours. Drift diffusion modelling of decision-making in mice exposed to varying temperatures showed that TRPV1 deletion impairs evidence accumulation, reducing the precision of thermal choice, while TRPM2 deletion increases overall preference for warmer environments that wildtype mice avoid. It remains unclear whether TRPM2 in DRG sensory neurons or elsewhere mediates thermal preference. Our findings suggest that different aspects of thermal information, such as stimulation speed and temperature magnitude, are encoded by distinct TRP channel mechanisms.
Collapse
Affiliation(s)
- Muad Y Abd El Hay
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
- Ernst Strüngmann Institute for Neuroscience in Cooperation with the Max Planck SocietyFrankfurt am MainGermany
| | - Gretel B Kamm
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
| | - Alejandro Tlaie Boria
- Ernst Strüngmann Institute for Neuroscience in Cooperation with the Max Planck SocietyFrankfurt am MainGermany
| | - Jan Siemens
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| |
Collapse
|
4
|
Young M, Ceddia RP, Thompson-Gray A, Reyes D, Cassada JB, Ayala JE, McGuinness OP, Collins S, Hamm HE. Sex differences in metabolic regulation by Gi/o-coupled receptor modulation of exocytosis. Front Pharmacol 2025; 16:1544456. [PMID: 40176888 PMCID: PMC11962901 DOI: 10.3389/fphar.2025.1544456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/07/2025] [Indexed: 04/05/2025] Open
Abstract
Background Presynaptic Gi/o coupled GPCRs can act as negative feedback regulators of neurotransmitter release via Gβγ effector modulation through two mechanisms: decreased calcium influx and direct inhibition of membrane fusion by soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE). Previously, we discovered that truncation of the last three C-terminal amino acids of SNAP25 (SNAP25Δ3) prevents Gβγ-SNARE interaction, effectively removing the braking mechanism on neurotransmitter release. We have demonstrated enhanced metabolic protection in male SNAP25Δ3/Δ3 mice housed at room temperature (22°C), including increased adipose tissue beiging and glucose uptake and enhanced insulin sensitivity, rendering them resistant to diet-induced obesity (DIO). When male SNAP25Δ3/Δ3 mice were housed at thermoneutrality (30°C), all metabolic protection was abolished, suggesting sympathetic tone is important for the phenotypes. Methods We housed male and female mice at either standard room temperature (21°C) or at thermoneutrality (30°C) and fed them a high fat diet (HFD) for 8 weeks. Glucose tolerance tests were performed before and after the 8 weeks of HFD along with body composition analyses. Organs were then dissected for mass analysis as well as immunohistochemistry. Additionally, we ovariectomized female mice to investigate the role of sex hormones in our phenotypes. Finally, we housed mice in Sable Promethion chambers at various environmental temperatures to investigate the effect of environmental temperature on basal metabolic rates. Results We found SNAP25Δ3/Δ3 female mice exhibited the same metabolic protection at RT (22°C) and displayed enhanced metabolic protection from DIO compared to standard chow just as males did. However, female SNAP25Δ3/Δ3 mice display persistent metabolic protection even when housed at thermoneutrality. In this study, we investigate the mechanisms behind this sex dependent persistent phenotype. Thermoneutral set point did not differ between sexes nor genotype, suggesting that metabolic protection is not due to a difference in hypothalamic temperature regulation. Metabolic protection in SNAP25Δ3/Δ3 persisted in ovariectomized mice despite increased weight gain compared to mice receiving sham operations. Conclusion This study has identified that there is not a sex-dependent difference for thermoneutral set point in mice. Additionally, there is a sex hormone independent mechanism driving the persistent metabolic protection of female SNAP25Δ3/Δ3 mice housed in thermoneutrality.
Collapse
Affiliation(s)
- Montana Young
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Ryan P. Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Analisa Thompson-Gray
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - David Reyes
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Jackson B. Cassada
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Julio E. Ayala
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, IN, United States
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, IN, United States
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, IN, United States
| | - Heidi E. Hamm
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
5
|
Vöröslakos M, Zhang Y, McClain K, Huszár R, Rothstein A, Buzsáki G. ThermoMaze behavioral paradigm for assessing immobility-related brain events in rodents. eLife 2025; 12:RP90347. [PMID: 40052764 PMCID: PMC11888600 DOI: 10.7554/elife.90347] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
Brain states fluctuate between exploratory and consummatory phases of behavior. These state changes affect both internal computation and the organism's responses to sensory inputs. Understanding neuronal mechanisms supporting exploratory and consummatory states and their switching requires experimental control of behavioral shifts and collecting sufficient amounts of brain data. To achieve this goal, we developed the ThermoMaze, which exploits the animal's natural warmth-seeking homeostatic behavior. By decreasing the floor temperature and selectively heating unmarked areas, we observed that mice avoided the aversive state by exploring the maze and finding the warm spot. In its design, the ThermoMaze is analogous to the widely used water maze but without the inconvenience of a wet environment and, therefore, allows the collection of physiological data in many trials. We combined the ThermoMaze with electrophysiology recording, and report that spiking activity of hippocampal CA1 neurons during sharp-wave ripple events encode the position of mice. Thus, place-specific firing is not confined to locomotion and associated theta oscillations but persist during waking immobility and sleep at the same location. The ThermoMaze will allow for detailed studies of brain correlates of immobility, preparatory-consummatory transitions, and open new options for studying behavior-mediated temperature homeostasis.
Collapse
Affiliation(s)
| | - Yunchang Zhang
- Neuroscience Institute, New York UniversityNew YorkUnited States
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Kathryn McClain
- Neuroscience Institute, New York UniversityNew YorkUnited States
| | - Roman Huszár
- Neuroscience Institute, New York UniversityNew YorkUnited States
| | - Aryeh Rothstein
- Neuroscience Institute, New York UniversityNew YorkUnited States
| | - György Buzsáki
- Neuroscience Institute, New York UniversityNew YorkUnited States
- Department of Neurology, School of Medicine, New York UniversityNew YorkUnited States
| |
Collapse
|
6
|
Deota S, Pendergast JS, Kolthur-Seetharam U, Esser KA, Gachon F, Asher G, Dibner C, Benitah SA, Escobar C, Muoio DM, Zhang EE, Hotamışlıgil GS, Bass J, Takahashi JS, Rabinowitz JD, Lamia KA, de Cabo R, Kajimura S, Longo VD, Xu Y, Lazar MA, Verdin E, Zierath JR, Auwerx J, Drucker DJ, Panda S. The time is now: accounting for time-of-day effects to improve reproducibility and translation of metabolism research. Nat Metab 2025; 7:454-468. [PMID: 40097742 DOI: 10.1038/s42255-025-01237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
The constant expansion of the field of metabolic research has led to more nuanced and sophisticated understanding of the complex mechanisms that underlie metabolic functions and diseases. Collaborations with scientists of various fields such as neuroscience, immunology and drug discovery have further enhanced the ability to probe the role of metabolism in physiological processes. However, many behaviours, endocrine and biochemical processes, and the expression of genes, proteins and metabolites have daily ~24-h biological rhythms and thus peak only at specific times of the day. This daily variation can lead to incorrect interpretations, lack of reproducibility across laboratories and challenges in translating preclinical studies to humans. In this Review, we discuss the biological, environmental and experimental factors affecting circadian rhythms in rodents, which can in turn alter their metabolic pathways and the outcomes of experiments. We recommend that these variables be duly considered and suggest best practices for designing, analysing and reporting metabolic experiments in a circadian context.
Collapse
Affiliation(s)
- Shaunak Deota
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Charna Dibner
- Department of Surgery and Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute for Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Deborah M Muoio
- Departments of Medicine and Pharmacology & Cancer Biology, Duke Molecular Physiology Institute, Durham, NC, USA
| | | | - Gökhan S Hotamışlıgil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Katja A Lamia
- Department of Molecular and Cellular Biology and Department of Molecular Medicine, the Scripps Research Institute, La Jolla, CA, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Shingo Kajimura
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- AIRC Institute of Molecular Oncology, Italian Foundation for Cancer Research Institute of Molecular Oncology, Milan, Italy
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity and Metabolism and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
7
|
Miles TK, Odle AK, Byrum SD, Lagasse AN, Haney AC, Ortega VG, Herdman AK, MacNicol MC, MacNicol AM, Childs GV. Ablation of Leptin Receptor Signaling Alters Somatotrope Transcriptome Maturation in Female Mice. Endocrinology 2025; 166:bqaf036. [PMID: 39964842 PMCID: PMC11919818 DOI: 10.1210/endocr/bqaf036] [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: 10/08/2024] [Revised: 01/13/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
Anterior pituitary somatotropes respond to metabolic signals from the adipokine leptin to optimize functional responses to the body's nutritional state via growth hormone (GH) secretion. Molecular targets of leptin in pituitary somatotropes include GH, the GH-releasing hormone receptor (GHRHR), and, in females, the transcription factor POU1F1, all of which are dependent on leptin stimulation for expression. To identify the trophic mechanisms underlying leptin action upon somatotropes, we analyzed single-cell gene transcriptomes comparing pituitaries from a female mouse model bearing somatotropes lacking leptin receptors (LEPR-null mutants) and control pituitaries. Computational clustering of results identified all common pituitary cell types and differentially expressed genes. Mutant female somatotrope clusters showed decreased levels of Gh and Htatsf1 mRNA, which was also reduced in mutant pituitaries lacking Prop1 or POU1F1. Mutant somatotropes also showed increased expression of markers for pituitary stem and progenitor cells (eg, Sox9) and increased (1.73-6.7 fold) expression of nonsomatotrope hormones, Pomc, Lhb, Tshb, Cga, and Prl. Conversely, the mutant female Sox2-positive stem cell cluster showed decreased expression of markers for stem cells and increased expression of pituitary hormone genes. The data support a model in which the female pituitary somatotrope cell population's development and/or maintenance requires leptin trophic signals and also suggests that, in the absence of normal somatotrope maturation, pituitary stem cells are driven towards premature differentiation.
Collapse
Affiliation(s)
- Tiffany K Miles
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angela K Odle
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alex N Lagasse
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Anessa C Haney
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Victoria G Ortega
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ashley K Herdman
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melanie C MacNicol
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angus M MacNicol
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gwen V Childs
- Department of Neuroscience and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
8
|
Patel TP, Jun JY, Seo AY, Levi NJ, Elizondo DM, Chen J, Wong AM, Tugarinov N, Altman EK, Gehle DB, Jung SM, Patel P, Ericson M, Haskell-Luevano C, Demby TC, Cougnoux A, Wolska A, Yanovski JA. Melanocortin 3 receptor regulates hepatic autophagy and systemic adiposity. Nat Commun 2025; 16:1690. [PMID: 39956805 PMCID: PMC11830824 DOI: 10.1038/s41467-025-56936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/05/2025] [Indexed: 02/18/2025] Open
Abstract
Systemic lipid homeostasis requires hepatic autophagy, a major cellular program for intracellular fat recycling. Here, we find melanocortin 3 receptor (MC3R) regulates hepatic autophagy in addition to its previously established CNS role in systemic energy partitioning and puberty. Mice with Mc3r deficiency develop obesity with hepatic triglyceride accumulation and disrupted hepatocellular autophagosome turnover. Mice with partially inactive human MC3R due to obesogenic variants demonstrate similar hepatic autophagic dysfunction. In vitro and in vivo activation of hepatic MC3R upregulates autophagy through LC3II activation, TFEB cytoplasmic-to-nuclear translocation, and subsequent downstream gene activation. MC3R-deficient hepatocytes had blunted autophagosome-lysosome docking and lipid droplet clearance. Finally, the liver-specific rescue of Mc3r was sufficient to restore hepatocellular autophagy, improve hepatocyte mitochondrial function and systemic energy expenditures, reduce adipose tissue lipid accumulation, and partially restore body weight in both male and female mice. We thus report a role for MC3R in regulating hepatic autophagy and systemic adiposity.
Collapse
Affiliation(s)
- Tushar P Patel
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Joo Yun Jun
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Arnold Y Seo
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Noah J Levi
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Diana M Elizondo
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Jocelyn Chen
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Adrian M Wong
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Nicol Tugarinov
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Elizabeth K Altman
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Daniel B Gehle
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Sun Min Jung
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Pooja Patel
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Mark Ericson
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Tamar C Demby
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Antony Cougnoux
- Section on Molecular Dysmorphology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Jack A Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA.
| |
Collapse
|
9
|
Adahman Z, Ooyama R, Gashi DB, Medik ZZ, Hollosi HK, Sahoo B, Akowuah ND, Riceberg JS, Carcea I. Hypothalamic Vasopressin Neurons Enable Maternal Thermoregulatory Behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634569. [PMID: 40196592 PMCID: PMC11974691 DOI: 10.1101/2025.01.23.634569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Newborns of many mammalian species are partial poikilotherms and require adult thermoregulatory care for survival. In mice, pup survival in cold and cool ambient temperature depends on the ability of adult caregivers to huddle pups and bring them into a high-quality nest. It is therefore essential that adult mice adjust parental care as a function of changes in ambient temperature. Here, we investigated how mouse maternal care adapts to a range of temperatures, from cold to warm. We show that changes in ambient temperature affect several individual and co-parenting maternal behaviors in both dams and virgin female mice, and modulate activity of vasopressin neurons. Furthermore, we establish that the effects of ambient temperature on both maternal care and the activity of vasopressin neurons depend in part on thermosensation, specifically on the TRPM8 sensor. Using trans-synaptic anterograde tracing and whole-brain activity mapping, we find that vasopressin neurons from the paraventricular hypothalamic nucleus connect synaptically with temperature-responsive brain structures implicated in maternal care. We then show that optogenetic activation of vasopressin projections to the central amygdala, a structure activated by cold ambient temperature, recapitulates the effects of cold on co-parenting behaviors. Our data provide a biological mechanism for maternal thermoregulatory behavior in mice with translational relevance to the reported association between ecosystem temperature fluctuations and variations in human child neglect cases.
Collapse
Affiliation(s)
- Zahra Adahman
- Rutgers, The State University of New Jersey, New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ, USA
- Rutgers, The State University of New Jersey, School of Graduate Studies, Health Sciences Campus, Newark, NJ, USA
- Rutgers, The State University of New Jersey, Brain Health Institute, Piscataway, NJ, USA
| | - Rumi Ooyama
- Rutgers, The State University of New Jersey, New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ, USA
- Rutgers, The State University of New Jersey, School of Graduate Studies, Health Sciences Campus, Newark, NJ, USA
- Rutgers, The State University of New Jersey, Brain Health Institute, Piscataway, NJ, USA
| | - Dinore B. Gashi
- Rutgers, The State University of New Jersey, School of Graduate Studies, Health Sciences Campus, Newark, NJ, USA
| | - Zeyneb Z. Medik
- Rutgers, The State University of New Jersey, New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ, USA
- Bezmialem Vakıf University, Department of Medicine, Instanbul, Turkey
| | - Hannah K. Hollosi
- Rutgers, The State University of New Jersey, New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ, USA
| | - Biswaranjan Sahoo
- Rutgers, The State University of New Jersey, New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ, USA
- Rutgers, The State University of New Jersey, Brain Health Institute, Piscataway, NJ, USA
| | - Nana D. Akowuah
- Rutgers, The State University of New Jersey, New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ, USA
- Rutgers, The State University of New Jersey, School of Graduate Studies, Health Sciences Campus, Newark, NJ, USA
- Rutgers, The State University of New Jersey, Brain Health Institute, Piscataway, NJ, USA
| | | | - Ioana Carcea
- Rutgers, The State University of New Jersey, New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ, USA
- Rutgers, The State University of New Jersey, Brain Health Institute, Piscataway, NJ, USA
| |
Collapse
|
10
|
Cypess AM, Cannon B, Nedergaard J, Kazak L, Chang DC, Krakoff J, Tseng YH, Schéele C, Boucher J, Petrovic N, Blondin DP, Carpentier AC, Virtanen KA, Kooijman S, Rensen PCN, Cero C, Kajimura S. Emerging debates and resolutions in brown adipose tissue research. Cell Metab 2025; 37:12-33. [PMID: 39644896 PMCID: PMC11710994 DOI: 10.1016/j.cmet.2024.11.002] [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: 03/26/2024] [Revised: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
Until two decades ago, brown adipose tissue (BAT) was studied primarily as a thermogenic organ of small rodents in the context of cold adaptation. The discovery of functional human BAT has opened new opportunities to understand its physiological role in energy balance and therapeutic applications for metabolic disorders. Significantly, the role of BAT extends far beyond thermogenesis, including glucose and lipid homeostasis, by releasing mediators that communicate with other cells and organs. The field has made major advances by using new model systems, ranging from subcellular studies to clinical trials, which have also led to debates. In this perspective, we identify six fundamental issues that are currently controversial and comprise dichotomous models. Each side presents supporting evidence and, critically, the necessary methods and falsifiable experiments that would resolve the dispute. With this collaborative approach, the field will continue to productively advance the understanding of BAT physiology, appreciate the importance of thermogenic adipocytes as a central area of ongoing research, and realize the therapeutic potential.
Collapse
Affiliation(s)
- Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Camilla Schéele
- Novo Nordisk Foundation Center for Basic Metabolic Research, The Center of Inflammation and Metabolism and the Center for Physical Activity Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Cheryl Cero
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
11
|
Kingren MS, Hall JS, Ross TJ, Barre MC, Barlow A, Morales M, Treas LD, Maxson RT, Teo E, Porter C. HOUSING TEMPERATURE ALTERS BURN-INDUCED HYPERMETABOLISM IN MICE. Shock 2025; 63:118-131. [PMID: 39450911 DOI: 10.1097/shk.0000000000002476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
ABSTRACT Mice used in biomedical research are typically housed at ambient temperatures (22°C-24°C) below thermoneutrality (26°C-31°C). This chronic cold stress triggers a hypermetabolic response that may limit the utility of mice in modeling hypermetabolism in response to burns. To evaluate the effect of housing temperature on burn-induced hypermetabolism, mice were randomly assigned to receive sham, small, or large scald burns. Mice recovered for 21 days in metabolic phenotyping cages at 24°C or 30°C. Regardless of sex or sham/burn treatment, mice housed at 24°C had greater total energy expenditure ( P < 0.001), which was largely attributable to greater basal energy expenditure when compared to mice housed at 30°C ( P < 0.001). Thermoneutral housing (30°C) altered adipose tissue mass in a sex-dependent manner. Compared to sham and small burn groups, large burns resulted in greater water vapor loss, regardless of housing temperature ( P < 0.01). Compared to sham, large burns resulted in greater basal energy expenditure and total energy expenditure in mice housed at 24°C; however, this hypermetabolic response to large burns was blunted in female mice housed at 30°C, and absent in male mice housed at 30°C. Locomotion was significantly reduced in mice with large burns compared to sham and small burn groups, irrespective of sex or housing temperature ( P < 0.05). Housing at 30°C revealed sexual dimorphism in terms of the impact of burns on body mass and composition, where males with large burns displayed marked cachexia, whereas females did not. Collectively, this study demonstrates a sex-dependent role for housing temperature in influencing energetics and body composition in a rodent model of burn trauma.
Collapse
Affiliation(s)
| | | | | | | | - Abigail Barlow
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Martin Morales
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | | | | | | | | |
Collapse
|
12
|
Sylvester E, Deng C, McIntosh R, Iskra S, Frankland J, McKenzie R, Croft RJ. Characterising core body temperature response of free-moving C57BL/6 mice to 1.95 GHz whole-body radiofrequency-electromagnetic fields. Bioelectromagnetics 2024; 45:387-398. [PMID: 39402826 DOI: 10.1002/bem.22527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/22/2024] [Accepted: 09/23/2024] [Indexed: 11/14/2024]
Abstract
The present study investigated the core body temperature (CBT) response of free-moving adult male and female C57BL/6 mice, during and following a 2-h exposure to 1.95 GHz RF-EMF within custom-built reverberation chambers, using temperature capsules implanted within the intraperitoneal cavity and data continuously logged and transmitted via radiotelemetry postexposure. Comparing RF-EMF exposures (WBA-SAR of 1.25, 2.5, 3.75, and 5 W/kg) to the sham-exposed condition, we identified a peak in CBT within the first 16 min of RF-EMF exposure (+0.15, +0.31, +0.24, +0.37°C at 1.25, 2.5, 3.75, and 5 W/kg respectively; statistically significant at WBA-SAR ≥ 2.5 W/kg only), which largely dissipated for the remainder of the exposure period. Immediately before the end of exposure, only the CBT of the 5 W/kg condition was statistically differentiable from sham. Based on our findings, it is apparent that mice are able to effectively compensate for the increased thermal load at RF-EMF strengths up to 5 W/kg. In addition, the elevated CBT at the end of the exposure period in the 5 W/kg condition was statistically significantly reduced compared to the sham condition immediately after RF-EMF exposure ceased. This would indicate that measures of CBT following the end of an RF-EMF exposure period may not reflect the actual change in the CBT of mice caused by RF-EMF exposure in mice.
Collapse
Affiliation(s)
- Emma Sylvester
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, Australia
- Wollongong Bioelectromagnetics Laboratory, Wollongong, Australia
| | - Chao Deng
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, Australia
- Wollongong Bioelectromagnetics Laboratory, Wollongong, Australia
| | - Robert McIntosh
- 6G Research and Innovation Lab, Swinburne University of Technology, Hawthorn, Australia
- Telstra Limited, Melbourne, Australia
| | - Steve Iskra
- 6G Research and Innovation Lab, Swinburne University of Technology, Hawthorn, Australia
| | - John Frankland
- 6G Research and Innovation Lab, Swinburne University of Technology, Hawthorn, Australia
| | - Raymond McKenzie
- 6G Research and Innovation Lab, Swinburne University of Technology, Hawthorn, Australia
- School of Psychology, University of Wollongong, Wollongong, Australia
| | - Rodney J Croft
- Wollongong Bioelectromagnetics Laboratory, Wollongong, Australia
- School of Psychology, University of Wollongong, Wollongong, Australia
- Australian Centre for Electromagnetic Bioeffects Research, Wollongong, New South Wales, Australia
| |
Collapse
|
13
|
Paim MP, Strelow DN, Krüger LD, Magalhães LS, Hall TK, Brüning CA, Bortolatto CF. Intermittent access to sugary drinks associated with fasting induces overeating and depressive-like behavior in female C57BL/6J mice. Neuroscience 2024; 560:20-35. [PMID: 39293731 DOI: 10.1016/j.neuroscience.2024.09.028] [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: 09/28/2023] [Revised: 08/31/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Binge eating disorder is the most prevalent eating disorder, affecting both sexes but more commonly found in women. Given the frequent co-occurrence of psychiatric disorders, this study aimed to establish a standardized experimental intermittent protocol to investigate overeating associated with depression. A 10-day protocol induced uncontrolled eating behavior in C57BL/6J female mice. The first experiment included the following groups: naive group (chow ad libitum), control group (chow and sucrose solution ad libitum), and fasting groups (16 and 20 h) exposed to an intermittent sucrose solution (10 %) and chow regimen. Subsequently, the feeding test, open field test, elevated plus maze test, tail suspension test, and light/dark conflict test were conducted. Furthermore, monoamine oxidase (MAO) A and B activities in brain structures and plasma corticosterone levels were assessed. Food overconsumption and depressive-like behavior were observed in both sucrose fasting groups, while risk-taking behaviors were specifically observed in the 20-hour fasting sucrose group. While both fasting sucrose groups caused reduced hippocampal MAO-A activity, only the F20 sucrose group inhibited MAO-B in the cortex and hypothalamus. Moreover, both fasting sucrose groups exhibited elevated corticosterone levels. In a separate design (Experiment 2), groups with 16 and 20 h of fasting alone (without sucrose) did not show the same behavioral results as the intermittent fasting sucrose groups, thus avoiding fasting bias. Based on these results, the 20-hour sucrose fasting group was chosen as the ideal protocol for mimicking overeating behavior associated with depression to investigate future therapeutic approaches for this comorbidity.
Collapse
Affiliation(s)
- Mariana Parron Paim
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Dianer Nornberg Strelow
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Letícia Devantier Krüger
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Larissa Sander Magalhães
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - Tácia Katiane Hall
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil
| | - César Augusto Brüning
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil.
| | - Cristiani Folharini Bortolatto
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), CEP 96010-900 Pelotas, RS, Brazil.
| |
Collapse
|
14
|
Prabhat A, Sami D, Ehlman A, Stumpf I, Seward T, Su W, Gong MC, Schroder EA, Delisle BP. Dim light at night unmasks sex-specific differences in circadian and autonomic regulation of cardiovascular physiology. Commun Biol 2024; 7:1191. [PMID: 39333678 PMCID: PMC11437115 DOI: 10.1038/s42003-024-06861-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/06/2024] [Indexed: 09/29/2024] Open
Abstract
Shift work and artificial light at night disrupt the entrainment of endogenous circadian rhythms in physiology and behavior to the day-night cycle. We hypothesized that exposure to dim light at night (dLAN) disrupts feeding rhythms, leading to sex-specific changes in autonomic signaling and day-night heart rate and blood pressure rhythms. Compared to mice housed in 12-hour light/12-hour dark cycles, mice exposed to dLAN showed reduced amplitudes in day-night feeding, heart rate, and blood pressure rhythms. In female mice, dLAN reduced the amplitude of day-night cardiovascular rhythms by decreasing the relative sympathetic regulation at night, while in male mice, it did so by increasing the relative sympathetic regulation during the daytime. Time-restricted feeding to the dim light cycle reversed these autonomic changes in both sexes. We conclude that dLAN induces sex-specific changes in autonomic regulation of heart rate and blood pressure, and time-restricted feeding may represent a chronotherapeutic strategy to mitigate the cardiovascular impact of light at night.
Collapse
Affiliation(s)
- Abhilash Prabhat
- Department of Physiology, University of Kentucky, Lexington, USA.
| | - Dema Sami
- Department of Physiology, University of Kentucky, Lexington, USA
| | - Allison Ehlman
- Department of Physiology, University of Kentucky, Lexington, USA
| | - Isabel Stumpf
- Department of Physiology, University of Kentucky, Lexington, USA
| | - Tanya Seward
- Department of Physiology, University of Kentucky, Lexington, USA
| | - Wen Su
- Department of Physiology, University of Kentucky, Lexington, USA
| | - Ming C Gong
- Department of Physiology, University of Kentucky, Lexington, USA
| | - Elizabeth A Schroder
- Department of Physiology, University of Kentucky, Lexington, USA
- Department of Internal Medicine, University of Kentucky, Lexington, USA
| | - Brian P Delisle
- Department of Physiology, University of Kentucky, Lexington, USA.
| |
Collapse
|
15
|
Sadler DG, Landes RD, Treas L, Sikes J, Porter C. Protonophore treatment augments energy expenditure in mice housed at thermoneutrality. Front Physiol 2024; 15:1452986. [PMID: 39381330 PMCID: PMC11458463 DOI: 10.3389/fphys.2024.1452986] [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: 06/21/2024] [Accepted: 08/26/2024] [Indexed: 10/10/2024] Open
Abstract
Background Sub-thermoneutral housing increases facultative thermogenesis in mice, which may mask the pre-clinical efficacy of anti-obesity strategies that target energy expenditure (EE). Here, we quantified the impact of protonophore treatment on whole-body energetics in mice housed at 30°C. Methods C57BL/6J mice (n = 48, 24M/24F) were housed at 24°C for 2 weeks; 32 (16M/16F) were then transitioned to 30°C for a further 4 weeks. Following 2 weeks acclimation at 30°C, mice (n = 16 per group, 8M/8F) received either normal (0 mg/L; Control) or supplemented (400 mg/L; 2,4-Dinitrophenol [DNP]) drinking water. Mice were singly housed in metabolic cages to determine total EE (TEE) and its components via respiratory gas exchange. Mitochondrial respiratory function of permeabilized liver tissue was determined by high-resolution respirometry. Results Transitioning mice from 24°C to 30°C reduced TEE and basal EE (BEE) by 16% and 41%, respectively (both P < 0.001). Compared to 30°C controls, TEE was 2.6 kcal/day greater in DNP-treated mice (95% CI: 1.6-3.6 kcal/day, P < 0.001), which was partly due to a 1.2 kcal/day higher BEE in DNP-treated mice (95% CI: 0.6-1.7 kcal/day, P < 0.001). The absolute TEE of 30°C DNP-treated mice was lower than that of mice housed at 24°C in the absence of DNP (DNP: 9.4 ± 0.7 kcal/day vs. 24°C control: 10.4 ± 1.5 kcal/day). DNP treatment reduced overall body fat of females by 2.9 percentage points versus sex-matched controls (95% CI: 1.3%-4.5%, P < 0.001), which was at least partly due to a reduction in inguinal white fat mass. Conclusion Protonophore treatment markedly increases EE in mice housed at 30°C. The magnitude of change in TEE of mice receiving protonophore treatment at 30°C was smaller than that brought about by transitioning mice from 24°C to 30°C, emphasizing that housing temperature must be considered when assessing anti-obesity strategies that target EE in mice.
Collapse
Affiliation(s)
- Daniel G. Sadler
- Arkansas Children’s Nutrition Center, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Department of Pediatrics, Univesrity of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Reid D. Landes
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Departments of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Lillie Treas
- Arkansas Children’s Nutrition Center, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - James Sikes
- Arkansas Children’s Nutrition Center, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Craig Porter
- Arkansas Children’s Nutrition Center, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Department of Pediatrics, Univesrity of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
16
|
Landen JG, Vandendoren M, Killmer S, Bedford NL, Nelson AC. Huddling substates in mice facilitate dynamic changes in body temperature and are modulated by Shank3b and Trpm8 mutation. Commun Biol 2024; 7:1186. [PMID: 39304735 PMCID: PMC11415358 DOI: 10.1038/s42003-024-06781-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Social thermoregulation is a means of maintaining homeostatic body temperature. While adult mice are a model organism for studying both social behavior and energy regulation, the relationship between huddling and core body temperature (Tb) is poorly understood. Here, we develop a behavioral paradigm and computational tools to identify active-huddling and quiescent-huddling as distinct thermal substates. We find that huddling is an effective thermoregulatory strategy in female but not male groups. At 23 °C (room temperature), but not 30 °C (near thermoneutrality), huddling facilitates large reductions in Tb and Tb-variance. Notably, active-huddling is associated with bidirectional changes in Tb, depending on its proximity to bouts of quiescent-huddling. Further, group-housed animals lacking the synaptic scaffolding gene Shank3b have hyperthermic Tb and spend less time huddling. In contrast, individuals lacking the cold-sensing gene Trpm8 have hypothermic Tb - a deficit that is rescued by increased huddling time. These results reveal how huddling behavior facilitates acute adjustments of Tb in a state-dependent manner.
Collapse
Affiliation(s)
- Jason G Landen
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
- University of Wyoming Sensory Biology Center, Laramie, WY, USA
| | - Morgane Vandendoren
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
- University of Wyoming Sensory Biology Center, Laramie, WY, USA
| | - Samantha Killmer
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
- University of Wyoming Sensory Biology Center, Laramie, WY, USA
| | - Nicole L Bedford
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
| | - Adam C Nelson
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA.
- University of Wyoming Sensory Biology Center, Laramie, WY, USA.
| |
Collapse
|
17
|
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
|
18
|
Jacobsen JM, Petersen N, Torz L, Gerstenberg MK, Pedersen K, Østergaard S, Wulff BS, Andersen B, Raun K, Christoffersen BØ, John LM, Reitman ML, Kuhre RE. Housing mice near vs. below thermoneutrality affects drug-induced weight loss but does not improve prediction of efficacy in humans. Cell Rep 2024; 43:114501. [PMID: 39067024 PMCID: PMC11380917 DOI: 10.1016/j.celrep.2024.114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Evaluation of weight loss drugs is usually performed in diet-induced obese mice housed at ∼22°C. This is a cold stress that increases energy expenditure by ∼35% compared to thermoneutrality (∼30°C), which may overestimate drug-induced weight loss. We investigated five anti-obesity mechanisms that have been in clinical development, comparing weight loss in mice housed at 22°C vs. 30°C. Glucagon-like peptide-1 (GLP-1), human fibroblast growth factor 21 (hFGF21), and melanocortin-4 receptor (MC4R) agonist induced similar weight losses. Peptide YY elicited greater vehicle-subtracted weight loss at 30°C (7.2% vs. 1.4%), whereas growth differentiation factor 15 (GDF15) was more effective at 22°C (13% vs. 6%). Independent of ambient temperature, GLP-1 and hFGF21 prevented the reduction in metabolic rate caused by weight loss. There was no simple rule for a better prediction of human drug efficacy based on ambient temperature, but since humans live at thermoneutrality, drug testing using mice should include experiments near thermoneutrality.
Collapse
Affiliation(s)
- Julie M Jacobsen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Natalia Petersen
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Lola Torz
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Kent Pedersen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Søren Østergaard
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte S Wulff
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte Andersen
- Diabetes, Obesity and NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Kirsten Raun
- Lead Portfolio Projects, Research and Early Development, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Linu M John
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Rune E Kuhre
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark; Department of Biomedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
19
|
Mountain RV, Peters RL, Langlais AL, Stohn JP, Lary CW, Motyl KJ. Thermoneutral Housing has Limited Effects on Social Isolation-Induced Bone Loss in Male C57BL/6J Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607315. [PMID: 39149234 PMCID: PMC11326229 DOI: 10.1101/2024.08.09.607315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Social isolation stress has numerous known negative health effects, including increased risk for cardiovascular disease, dementia, as well as overall mortality. The impacts of social isolation on skeletal health, however, have not been thoroughly investigated. We previously found that four weeks of social isolation through single housing led to a significant reduction in trabecular and cortical bone in male, but not female, mice. One possible explanation for these changes in male mice is thermal stress due to sub-thermoneutral housing. Single housing at room temperature (~20-25°C)-below the thermoneutral range of mice (~26-34°C)-may lead to cold stress, which has known negative effects on bone. Therefore, the aim of this study was to test the hypothesis that housing mice near thermoneutrality, thereby ameliorating cold-stress, will prevent social isolation-induced bone loss in male C57BL/6J mice. 16-week-old mice were randomized into social isolation (1 mouse/cage) or grouped housing (4 mice/cage) at either room temperature (~23°C) or in a warm temperature incubator (~28°C) for four weeks (N=8/group). As seen in our previous studies, isolated mice at room temperature had significantly reduced bone parameters, including femoral bone volume fraction (BV/TV), bone mineral density (BMD), and cortical thickness. Contrary to our hypothesis, these negative effects on bone were not ameliorated by thermoneutral housing. Social isolation increased glucocorticoid-related gene expression in bone and Ucp1 and Pdk4 expression in BAT across temperatures, while thermoneutral housing increased percent lipid area and decreased Ucp1 and Pdk4 expression in BAT across housing conditions. Overall, our data suggest social isolation-induced bone loss is not a result of thermal stress from single housing and provides a key insight into the mechanism mediating the effects of isolation on skeletal health.
Collapse
Affiliation(s)
- Rebecca V. Mountain
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Rebecca L. Peters
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Audrie L. Langlais
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - J. Patrizia Stohn
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Christine W. Lary
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- Roux Institute, Northeastern University, Portland, ME, USA
| | - Katherine J. Motyl
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- Tufts University School of Medicine, Tufts University, Boston, MA, USA
| |
Collapse
|
20
|
Farooqi IS, Xu Y. Translational potential of mouse models of human metabolic disease. Cell 2024; 187:4129-4143. [PMID: 39067442 DOI: 10.1016/j.cell.2024.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Obesity causes significant morbidity and mortality globally. Research in the last three decades has delivered a step-change in our understanding of the fundamental mechanisms that regulate energy homeostasis, building on foundational discoveries in mouse models of metabolic disease. However, not all findings made in rodents have translated to humans, hampering drug discovery in this field. Here, we review how studies in mice and humans have informed our current framework for understanding energy homeostasis, discuss their challenges and limitations, and offer a perspective on how human studies may play an increasingly important role in the discovery of disease mechanisms and identification of therapeutic targets in the future.
Collapse
Affiliation(s)
- I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
21
|
Chau PK, Ryan E, Dalen KT, Haugen F. Timing of acute cold exposure determines UCP1 and FGF21 expression - Possible interactions between the thermal environment, thermoregulatory responses, and peripheral clocks. J Therm Biol 2024; 124:103938. [PMID: 39142264 DOI: 10.1016/j.jtherbio.2024.103938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024]
Abstract
Thermoregulation is synchronized across the circadian cycle to uphold thermal homeostasis. To test if time-of-day matters for the response to environmental cold exposure, mice were acclimated to thermoneutrality (27 °C) for 2 months were subjected acutely (8 h) to cold ambient conditions (15 °C), whereas controls were maintained at thermoneutral conditions. The thermal exposure was tested in separate groups (N = 8) at three distinct time-of-day periods: in the LIGHT phase (L); the DARK phase (D); and a mix of the two (D + L). The magnitude of UCP1 protein and mRNA induction in brown adipose tissue (BAT) in response to acute cold exposure was time-of-day sensitive, peaking in LIGHT, whereas lower induction levels were observed in D + L, and DARK. Plasma levels of FGF21 were induced 3-fold by acute cold exposure at LIGHT and D + L, compared to the time-matched thermoneutral controls, whereas cold in DARK did not cause a significant increase of FGF21 plasma levels. Cold exposure affected, in BAT, the temporal mRNA expression patterns of core circadian clock components: Bmal1, Clock, Per1, Per3, Cry1, Cry2 Nr1d1, and Nr1d2, but in the liver, none of the transcripts were modified. Behavioral assessment using the Thermal Gradient Test (TGT) showed that acute cold exposure reduced cold sensitivity in D + L, but not in DARK. RNA-seq analyses of somatosensory neurons in DRG highlighted the role of the core circadian components in these cells, as well as transcriptional changes due to acute cold exposure. This elucidates the sensory system as a gauge and potential regulator of thermoregulatory responses based on circadian physiology. In conclusion, acute cold exposure elicits time-of-day specific effects on thermoregulatory pathways, which may involve underlying changes in thermal perception. These results have implications for efforts aimed at reducing risks associated with the organization of shift work in cold environments.
Collapse
Affiliation(s)
- Phong Kt Chau
- Division of Work Psychology and Physiology, National Institute of Occupational Health (STAMI), Oslo, Norway
| | - Elin Ryan
- Division of Work Psychology and Physiology, National Institute of Occupational Health (STAMI), Oslo, Norway
| | - Knut Tomas Dalen
- Department of Nutrition and Norwegian Transgenic Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Fred Haugen
- Division of Work Psychology and Physiology, National Institute of Occupational Health (STAMI), Oslo, Norway.
| |
Collapse
|
22
|
Mattar P, Reginato A, Lavados C, Das D, Kalyani M, Martinez-Lopez N, Sharma M, Skovbjerg G, Skytte JL, Roostalu U, Subbarayan R, Picarda E, Zang X, Zhang J, Guha C, Schwartz G, Rajbhandari P, Singh R. Insulin and leptin oscillations license food-entrained browning and metabolic flexibility. Cell Rep 2024; 43:114390. [PMID: 38900636 PMCID: PMC11562929 DOI: 10.1016/j.celrep.2024.114390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/23/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
Timed feeding drives adipose browning, although the integrative mechanisms for the same remain unclear. Here, we show that twice-a-night (TAN) feeding generates biphasic oscillations of circulating insulin and leptin, representing their entrainment by timed feeding. Insulin and leptin surges lead to marked cellular, functional, and metabolic remodeling of subcutaneous white adipose tissue (sWAT), resulting in increased energy expenditure. Single-cell RNA-sequencing (scRNA-seq) analyses and flow cytometry demonstrate a role for insulin and leptin surges in innate lymphoid type 2 (ILC2) cell recruitment and sWAT browning, since sWAT depot denervation or loss of leptin or insulin receptor signaling or ILC2 recruitment each dampens TAN feeding-induced sWAT remodeling and energy expenditure. Consistently, recreating insulin and leptin oscillations via once-a-day timed co-injections is sufficient to favorably remodel innervated sWAT. Innervation is necessary for sWAT remodeling, since denervation of sWAT, but not brown adipose tissue (BAT), blocks TAN-induced sWAT remodeling and resolution of inflammation. In sum, reorganization of nutrient-sensitive pathways remodels sWAT and drives the metabolic benefits of timed feeding.
Collapse
Affiliation(s)
- Pamela Mattar
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA
| | - Andressa Reginato
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Christian Lavados
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Debajyoti Das
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA
| | - Manu Kalyani
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nuria Martinez-Lopez
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA; Comprehensive Liver Research Center at UCLA, University of Los Angeles, Los Angeles, CA, USA
| | - Mridul Sharma
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | - Elodie Picarda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chandan Guha
- Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gary Schwartz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Prashant Rajbhandari
- Department of Medicine, Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rajat Singh
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Comprehensive Liver Research Center at UCLA, University of Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Wang L, Sun Y, Yang L, Wang S, Liu C, Wang Y, Niu Y, Huang Z, Zhang J, Wang C, Dong L. Engineering an energy-dissipating hybrid tissue in vivo for obesity treatment. Cell Rep 2024; 43:114425. [PMID: 38970789 DOI: 10.1016/j.celrep.2024.114425] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 07/08/2024] Open
Abstract
Obesity is a global health challenge with limited therapeutic solutions. Here, we demonstrate the engineering of an energy-dissipating hybrid tissue (EDHT) in the body for weight control. EDHT is constructed by implanting a synthetic gel matrix comprising immunomodulatory signals and functional cells into the recipient mouse. The immunomodulatory signals induce the host stromal cells to create an immunosuppressive niche that protects the functional cells, which are overexpressing the uncoupling protein 1 (UCP1), from immune rejection. Consequently, these endogenous and exogenous cells co-develop a hybrid tissue that sustainedly produces UCP1 to accelerate the host's energy expenditure. Systematic experiments in high-fat diet (HFD) and transgenic (ob/ob) mice show that EDHT efficiently reduces body weight and relieves obesity-associated pathological conditions. Importantly, an 18-month observation for safety assessment excludes cell leakage from EDHT and reports no adverse physiological responses. Overall, EDHT demonstrates convincing efficacy and safety in controlling body weight.
Collapse
Affiliation(s)
- Lintao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China; School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Yajie Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China
| | - Lifang Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China
| | - Shaocong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China
| | - Chunyan Liu
- Medical School, Nanjing University, Nanjing 210093, China
| | - Yulian Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China; School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Medical School, Nanjing University, Nanjing 210093, China.
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China; Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Taipa, Macau SAR, China.
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210093, China; Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210093, China; National Resource Center for Mutant Mice, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
24
|
Ono M, Burgess DE, Johnson SR, Elayi CS, Esser KA, Seward TS, Boychuk CR, Carreño AP, Stalcup RA, Prabhat A, Schroder EA, Delisle BP. Feeding behavior modifies the circadian variation in RR and QT intervals by distinct mechanisms in mice. Am J Physiol Regul Integr Comp Physiol 2024; 327:R109-R121. [PMID: 38766772 PMCID: PMC11380991 DOI: 10.1152/ajpregu.00025.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Rhythmic feeding behavior is critical for regulating phase and amplitude in the ≈24-h variation of heart rate (RR intervals), ventricular repolarization (QT intervals), and core body temperature in mice. We hypothesized changes in cardiac electrophysiology associated with feeding behavior were secondary to changes in core body temperature. Telemetry was used to record electrocardiograms and core body temperature in mice during ad libitum-fed conditions and after inverting normal feeding behavior by restricting food access to the light cycle. Light cycle-restricted feeding modified the phase and amplitude of 24-h rhythms in RR and QT intervals, and core body temperature to realign with the new feeding time. Changes in core body temperature alone could not account for changes in phase and amplitude in the ≈24-h variation of the RR intervals. Heart rate variability analysis and inhibiting β-adrenergic and muscarinic receptors suggested that changes in the phase and amplitude of 24-h rhythms in RR intervals were secondary to changes in autonomic signaling. In contrast, changes in QT intervals closely mirrored changes in core body temperature. Studies at thermoneutrality confirmed that the daily variation in QT interval, but not RR interval, primarily reflected daily changes in core body temperature (even in ad libitum-fed conditions). Correcting the QT interval for differences in core body temperature helped unmask QT interval prolongation after starting light cycle-restricted feeding and in a mouse model of long QT syndrome. We conclude feeding behavior alters autonomic signaling and core body temperature to regulate phase and amplitude in RR and QT intervals, respectively.NEW & NOTEWORTHY We used time-restricted feeding and thermoneutrality to demonstrate that different mechanisms regulate the 24-h rhythms in heart rate and ventricular repolarization. The daily rhythm in heart rate reflects changes in autonomic input, whereas daily rhythms in ventricular repolarization reflect changes in core body temperature. This novel finding has major implications for understanding 24-h rhythms in mouse cardiac electrophysiology, arrhythmia susceptibility in transgenic mouse models, and interpretability of cardiac electrophysiological data acquired in thermoneutrality.
Collapse
Affiliation(s)
- Makoto Ono
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Don E Burgess
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Sidney R Johnson
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Claude S Elayi
- CHI Saint Joseph Hospital, Lexington, Kentucky, United States
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Tanya S Seward
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Carie R Boychuk
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Andrés P Carreño
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Rebecca A Stalcup
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Abhilash Prabhat
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Elizabeth A Schroder
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
- Department of Internal Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Brian P Delisle
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
25
|
Landen JG, Vandendoren M, Killmer S, Bedford NL, Nelson AC. Huddling substates in mice facilitate dynamic changes in body temperature and are modulated by Shank3b and Trpm8 mutation. RESEARCH SQUARE 2024:rs.3.rs-3904829. [PMID: 38978581 PMCID: PMC11230468 DOI: 10.21203/rs.3.rs-3904829/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Social thermoregulation is a means of maintaining homeostatic body temperature. While adult mice are a model organism for studying both social behavior and energy regulation, the relationship between huddling and core body temperature (Tb) is poorly understood. Here, we develop a behavioral paradigm and computational tools to identify active-huddling and quiescent-huddling as distinct thermal substates. We find that huddling is an effective thermoregulatory strategy in female but not male groups. At 23°C (room temperature), but not 30°C (near thermoneutrality), huddling facilitates large reductions in Tb and Tb-variance. Notably, active-huddling is associated with bidirectional changes in Tb, depending on its proximity to bouts of quiescent-huddling. Further, group-housed animals lacking the synaptic scaffolding gene Shank3b have hyperthermic Tb and spend less time huddling. In contrast, individuals lacking the cold-sensing gene Trpm8 have hypothermic Tb - a deficit that is rescued by increased huddling time. These results reveal how huddling behavior facilitates acute adjustments of Tb in a state-dependent manner.
Collapse
Affiliation(s)
- Jason G. Landen
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
- University of Wyoming Sensory Biology Center, Laramie, WY, USA
| | - Morgane Vandendoren
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
- University of Wyoming Sensory Biology Center, Laramie, WY, USA
| | - Samantha Killmer
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
- University of Wyoming Sensory Biology Center, Laramie, WY, USA
| | - Nicole L. Bedford
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
| | - Adam C. Nelson
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
- University of Wyoming Sensory Biology Center, Laramie, WY, USA
| |
Collapse
|
26
|
Luu DD, Ramesh N, Kazan IC, Shah KH, Lahiri G, Mana MD, Ozkan SB, Van Horn WD. Evidence that the cold- and menthol-sensing functions of the human TRPM8 channel evolved separately. SCIENCE ADVANCES 2024; 10:eadm9228. [PMID: 38905339 PMCID: PMC11192081 DOI: 10.1126/sciadv.adm9228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Transient receptor potential melastatin 8 (TRPM8) is a temperature- and menthol-sensitive ion channel that contributes to diverse physiological roles, including cold sensing and pain perception. Clinical trials targeting TRPM8 have faced repeated setbacks predominantly due to the knowledge gap in unraveling the molecular underpinnings governing polymodal activation. A better understanding of the molecular foundations between the TRPM8 activation modes may aid the development of mode-specific, thermal-neutral therapies. Ancestral sequence reconstruction was used to explore the origins of TRPM8 activation modes. By resurrecting key TRPM8 nodes along the human evolutionary trajectory, we gained valuable insights into the trafficking, stability, and function of these ancestral forms. Notably, this approach unveiled the differential emergence of cold and menthol sensitivity over evolutionary time, providing a fresh perspective on complex polymodal behavior. These studies provide a paradigm for understanding polymodal behavior in TRPM8 and other proteins with the potential to enhance our understanding of sensory receptor biology and pave the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Dustin D. Luu
- School of Molecular Sciences and The Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Nikhil Ramesh
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, AZ, USA
| | - I. Can Kazan
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, AZ, USA
| | - Karan H. Shah
- School of Molecular Sciences and The Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Gourab Lahiri
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Miyeko D. Mana
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - S. Banu Ozkan
- Department of Physics and Center for Biological Physics, Arizona State University, Tempe, AZ, USA
| | - Wade D. Van Horn
- School of Molecular Sciences and The Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
27
|
Wang J, Huffman D, Ajwad A, McLouth CJ, Bachstetter A, Kohler K, Murphy MP, O'Hara BF, Duncan MJ, Sunderam S. Thermoneutral temperature exposure enhances slow-wave sleep with a correlated improvement in amyloid pathology in a triple-transgenic mouse model of Alzheimer's disease. Sleep 2024; 47:zsae078. [PMID: 38512801 DOI: 10.1093/sleep/zsae078] [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: 08/02/2023] [Revised: 01/14/2024] [Indexed: 03/23/2024] Open
Abstract
Accumulation of amyloid-β (Aβ) plays an important role in Alzheimer's disease (AD) pathology. There is growing evidence that disordered sleep may accelerate AD pathology by impeding the physiological clearance of Aβ from the brain that occurs in normal sleep. Therapeutic strategies for improving sleep quality may therefore help slow disease progression. It is well documented that the composition and dynamics of sleep are sensitive to ambient temperature. We therefore compared Aβ pathology and sleep metrics derived from polysomnography in 12-month-old female 3xTg-AD mice (n = 8) exposed to thermoneutral temperatures during the light period over 4 weeks to those of age- and sex-matched controls (n = 8) that remained at normal housing temperature (22°C) during the same period. The treated group experienced greater proportions of slow wave sleep (SWS)-i.e. epochs of elevated 0.5-2 Hz EEG slow wave activity during non-rapid eye movement (NREM) sleep-compared to controls. Assays performed on mouse brain tissue harvested at the end of the experiment showed that exposure to thermoneutral temperatures significantly reduced levels of DEA-soluble (but not RIPA- or formic acid-soluble) Aβ40 and Aβ42 in the hippocampus, though not in the cortex. With both groups pooled together and without regard to treatment condition, NREM sleep continuity and any measure of SWS within NREM at the end of the treatment period were inversely correlated with DEA-soluble Aβ40 and Aβ42 levels, again in the hippocampus but not in the cortex. These findings suggest that experimental manipulation of SWS could offer useful clues into the mechanisms and treatment of AD.
Collapse
Affiliation(s)
- Jun Wang
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| | - Dillon Huffman
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| | - Asma'a Ajwad
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Diyala College of Medicine, Diyala, Iraq
| | | | - Adam Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Katarina Kohler
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - M Paul Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - Marilyn J Duncan
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Sridhar Sunderam
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
28
|
Han A, Hudson-Paz C, Robinson BG, Becker L, Jacobson A, Kaltschmidt JA, Garrison JL, Bhatt AS, Monack DM. Temperature-dependent differences in mouse gut motility are mediated by stress. Lab Anim (NY) 2024; 53:148-159. [PMID: 38806681 PMCID: PMC11147774 DOI: 10.1038/s41684-024-01376-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 04/19/2024] [Indexed: 05/30/2024]
Abstract
Researchers have advocated elevating mouse housing temperatures from the conventional ~22 °C to the mouse thermoneutral point of 30 °C to enhance translational research. However, the impact of environmental temperature on mouse gastrointestinal physiology remains largely unexplored. Here we show that mice raised at 22 °C exhibit whole gut transit speed nearly twice as fast as those raised at 30 °C, primarily driven by a threefold increase in colon transit speed. Furthermore, gut microbiota composition differs between the two temperatures but does not dictate temperature-dependent differences in gut motility. Notably, increased stress signals from the hypothalamic-pituitary-adrenal axis at 22 °C have a pivotal role in mediating temperature-dependent differences in gut motility. Pharmacological and genetic depletion of the stress hormone corticotropin-releasing hormone slows gut motility in stressed 22 °C mice but has no comparable effect in relatively unstressed 30 °C mice. In conclusion, our findings highlight that colder mouse facility temperatures significantly increase gut motility through hormonal stress pathways.
Collapse
Affiliation(s)
- Alvin Han
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | | | - Beatriz G Robinson
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Laren Becker
- Department of Medicine (Gastroenterology and Hepatology), Stanford University, Stanford, CA, USA
| | - Amanda Jacobson
- Genentech Inc., Research and Early Development, Immunology Discovery, South San Francisco, CA, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer L Garrison
- Buck Institute for Research on Aging, Novato, CA, USA
- Global Consortium for Reproductive Longevity & Equality, Novato, CA, USA
| | - Ami S Bhatt
- Department of Medicine (Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Denise M Monack
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
29
|
Škop V, Liu N, Xiao C, Stinson E, Chen KY, Hall KD, Piaggi P, Gavrilova O, Reitman ML. Beyond day and night: The importance of ultradian rhythms in mouse physiology. Mol Metab 2024; 84:101946. [PMID: 38657735 PMCID: PMC11070603 DOI: 10.1016/j.molmet.2024.101946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024] Open
Abstract
Our circadian world shapes much of metabolic physiology. In mice ∼40% of the light and ∼80% of the dark phase time is characterized by bouts of increased energy expenditure (EE). These ultradian bouts have a higher body temperature (Tb) and thermal conductance and contain virtually all of the physical activity and awake time. Bout status is a better classifier of mouse physiology than photoperiod, with ultradian bouts superimposed on top of the circadian light/dark cycle. We suggest that the primary driver of ultradian bouts is a brain-initiated transition to a higher defended Tb of the active/awake state. Increased energy expenditure from brown adipose tissue, physical activity, and cardiac work combine to raise Tb from the lower defended Tb of the resting/sleeping state. Thus, unlike humans, much of mouse metabolic physiology is episodic with large ultradian increases in EE and Tb that correlate with the active/awake state and are poorly aligned with circadian cycling.
Collapse
Affiliation(s)
- Vojtěch Škop
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA; Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czech Republic.
| | - 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
| | - Emma Stinson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ 85016, USA
| | - Kong Y Chen
- 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
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ 85016, USA; Department of Information Engineering, University of Pisa, Pisa 56122, Italy
| | - 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
|
30
|
Rogers JF, Vandendoren M, Prather JF, Landen JG, Bedford NL, Nelson AC. Neural cell-types and circuits linking thermoregulation and social behavior. Neurosci Biobehav Rev 2024; 161:105667. [PMID: 38599356 PMCID: PMC11163828 DOI: 10.1016/j.neubiorev.2024.105667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Understanding how social and affective behavioral states are controlled by neural circuits is a fundamental challenge in neurobiology. Despite increasing understanding of central circuits governing prosocial and agonistic interactions, how bodily autonomic processes regulate these behaviors is less resolved. Thermoregulation is vital for maintaining homeostasis, but also associated with cognitive, physical, affective, and behavioral states. Here, we posit that adjusting body temperature may be integral to the appropriate expression of social behavior and argue that understanding neural links between behavior and thermoregulation is timely. First, changes in behavioral states-including social interaction-often accompany changes in body temperature. Second, recent work has uncovered neural populations controlling both thermoregulatory and social behavioral pathways. We identify additional neural populations that, in separate studies, control social behavior and thermoregulation, and highlight their relevance to human and animal studies. Third, dysregulation of body temperature is linked to human neuropsychiatric disorders. Although body temperature is a "hidden state" in many neurobiological studies, it likely plays an underappreciated role in regulating social and affective states.
Collapse
Affiliation(s)
- Joseph F Rogers
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Morgane Vandendoren
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Jonathan F Prather
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Jason G Landen
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Nicole L Bedford
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Adam C Nelson
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA.
| |
Collapse
|
31
|
Meng Z, Liu C, Xu M, Tao Y, Li H, Wang X, Liao J, Wang M. Adipose transplantation improves metabolism and atherosclerosis but not perivascular adipose tissue abnormality or vascular dysfunction in lipodystrophic Seipin/Apoe null mice. Am J Physiol Cell Physiol 2024; 326:C1410-C1422. [PMID: 38525541 PMCID: PMC11371364 DOI: 10.1152/ajpcell.00698.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Adipose dysfunction in lipodystrophic SEIPIN deficiency is associated with multiple metabolic disorders and increased risks of developing cardiovascular diseases, such as atherosclerosis, cardiac hypertrophy, and heart failure. Recently, adipose transplantation has been found to correct adipose dysfunction and metabolic disorders in lipodystrophic Seipin knockout mice; however, whether adipose transplantation could improve lipodystrophy-associated cardiovascular consequences is still unclear. Here, we aimed to explore the effects of adipose transplantation on lipodystrophy-associated metabolic cardiovascular diseases in Seipin knockout mice crossed into atherosclerosis-prone apolipoprotein E (Apoe) knockout background. At 2 months of age, lipodystrophic Seipin/Apoe double knockout mice and nonlipodystrophic Apoe knockout controls were subjected to adipose transplantation or sham operation. Seven months later, mice were euthanized. Our data showed that although adipose transplantation had no significant impact on endogenous adipose atrophy or gene expression, it remarkably increased plasma leptin but not adiponectin concentration in Seipin/Apoe double knockout mice. This led to significantly reduced hyperlipidemia, hepatic steatosis, and insulin resistance in Seipin/Apoe double knockout mice. Consequently, atherosclerosis burden, intraplaque macrophage infiltration, and aortic inflammatory gene expression were all attenuated in Seipin/Apoe double knockout mice with adipose transplantation. However, adipocyte morphology, macrophage infiltration, or fibrosis of the perivascular adipose tissue was not altered in Seipin/Apoe double knockout mice with adipose transplantation, followed by no significant improvement of vasoconstriction or relaxation. In conclusion, we demonstrate that adipose transplantation could alleviate lipodystrophy-associated metabolic disorders and atherosclerosis but has an almost null impact on perivascular adipose abnormality or vascular dysfunction in lipodystrophic Seipin/Apoe double knockout mice.NEW & NOTEWORTHY Adipose transplantation (AT) reverses multiply metabolic derangements in lipodystrophy, but whether it could improve lipodystrophy-related cardiovascular consequences is unknown. Here, using Seipin/Apoe double knockout mice as a lipodystrophy disease model, we showed that AT partially restored adipose functionality, which translated into significantly reduced atherosclerosis. However, AT was incapable of reversing perivascular adipose abnormality or vascular dysfunction. The current study provides preliminary experimental evidence on the therapeutic potential of AT on lipodystrophy-related metabolic cardiovascular diseases.
Collapse
Affiliation(s)
- Zhe Meng
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chuangxing Liu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengke Xu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yongqiang Tao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyu Li
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xijia Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mengyu Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
32
|
Engström Ruud L, Font-Gironès F, Zajdel J, Kern L, Teixidor-Deulofeu J, Mannerås-Holm L, Carreras A, Becattini B, Björefeldt A, Hanse E, Fenselau H, Solinas G, Brüning JC, Wunderlich TF, Bäckhed F, Ruud J. Activation of GFRAL + neurons induces hypothermia and glucoregulatory responses associated with nausea and torpor. Cell Rep 2024; 43:113960. [PMID: 38507407 DOI: 10.1016/j.celrep.2024.113960] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/11/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
GFRAL-expressing neurons actuate aversion and nausea, are targets for obesity treatment, and may mediate metformin effects by long-term GDF15-GFRAL agonism. Whether GFRAL+ neurons acutely regulate glucose and energy homeostasis is, however, underexplored. Here, we report that cell-specific activation of GFRAL+ neurons using a variety of techniques causes a torpor-like state, including hypothermia, the release of stress hormones, a shift from glucose to lipid oxidation, and impaired insulin sensitivity, glucose tolerance, and skeletal muscle glucose uptake but augmented glucose uptake in visceral fat. Metabolomic analysis of blood and transcriptomics of muscle and fat indicate alterations in ketogenesis, insulin signaling, adipose tissue differentiation and mitogenesis, and energy fluxes. Our findings indicate that acute GFRAL+ neuron activation induces endocrine and gluco- and thermoregulatory responses associated with nausea and torpor. While chronic activation of GFRAL signaling promotes weight loss in obesity, these results show that acute activation of GFRAL+ neurons causes hypothermia and hyperglycemia.
Collapse
Affiliation(s)
- Linda Engström Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ferran Font-Gironès
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joanna Zajdel
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lara Kern
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Júlia Teixidor-Deulofeu
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Mannerås-Holm
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alba Carreras
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbara Becattini
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Björefeldt
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eric Hanse
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Giovanni Solinas
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | | | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, The Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
33
|
Benevento M, Alpár A, Gundacker A, Afjehi L, Balueva K, Hevesi Z, Hanics J, Rehman S, Pollak DD, Lubec G, Wulff P, Prevot V, Horvath TL, Harkany T. A brainstem-hypothalamus neuronal circuit reduces feeding upon heat exposure. Nature 2024; 628:826-834. [PMID: 38538787 PMCID: PMC11041654 DOI: 10.1038/s41586-024-07232-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 02/22/2024] [Indexed: 04/06/2024]
Abstract
Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Leila Afjehi
- Programme Proteomics, Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Kira Balueva
- Institute of Physiology, Christian Albrechts University, Kiel, Germany
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - János Hanics
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Sabah Rehman
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Programme Proteomics, Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Peer Wulff
- Institute of Physiology, Christian Albrechts University, Kiel, Germany
| | - Vincent Prevot
- University of Lille, INSERM, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR S1172, EGID, Lille, France
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
34
|
Wang H, Ülgen M, Trajkovski M. Importance of temperature on immuno-metabolic regulation and cancer progression. FEBS J 2024; 291:832-845. [PMID: 36152006 DOI: 10.1111/febs.16632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/01/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022]
Abstract
Cancer immunotherapies emerge as promising strategies for restricting tumour growth. The tumour microenvironment (TME) has a major impact on the anti-tumour immune response and on the efficacy of the immunotherapies. Recent studies have linked changes in the ambient temperature with particular immuno-metabolic reprogramming and anti-cancer immune response in laboratory animals. Here, we describe the energetic balance of the organism during change in temperature, and link this to the immune alterations that could be of relevance for cancer, as well as for other human diseases. We highlight the contribution of the gut microbiota in modifying this interaction. We describe the overall metabolic response and underlying mechanisms of tumourigenesis in mouse models at varying ambient temperatures and shed light on their potential importance in developing therapeutics against cancer.
Collapse
Affiliation(s)
- Haiping Wang
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Medical Universitaire (CMU), University of Geneva, Geneva, Switzerland
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
| | - Melis Ülgen
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Medical Universitaire (CMU), University of Geneva, Geneva, Switzerland
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
| | - Mirko Trajkovski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Medical Universitaire (CMU), University of Geneva, Geneva, Switzerland
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
| |
Collapse
|
35
|
Miles TK, Odle AK, Byrum SD, Lagasse A, Haney A, Ortega VG, Bolen CR, Banik J, Reddick MM, Herdman A, MacNicol MC, MacNicol AM, Childs GV. Anterior Pituitary Transcriptomics Following a High-Fat Diet: Impact of Oxidative Stress on Cell Metabolism. Endocrinology 2023; 165:bqad191. [PMID: 38103263 PMCID: PMC10771268 DOI: 10.1210/endocr/bqad191] [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: 09/29/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/18/2023]
Abstract
Anterior pituitary cell function requires a high level of protein synthesis and secretion which depend heavily on mitochondrial adenosine triphosphate production and functional endoplasmic reticula. Obesity adds stress to tissues, requiring them to adapt to inflammation and oxidative stress, and adding to their allostatic load. We hypothesized that pituitary function is vulnerable to the stress of obesity. Here, we utilized a 10- to 15-week high-fat diet (HFD, 60%) in a thermoneutral environment to promote obesity, testing both male and female FVB.129P mice. We quantified serum hormones and cytokines, characterized the metabolic phenotype, and defined changes in the pituitary transcriptome using single-cell RNA-sequencing analysis. Weight gain was significant by 3 weeks in HFD mice, and by 10 weeks all HFD groups had gained 20 g. HFD females (15 weeks) had increased energy expenditure and decreased activity. All HFD groups showed increases in serum leptin and decreases in adiponectin. HFD caused increased inflammatory markers: interleukin-6, resistin, monocyte chemoattractant protein-1, and tumor necrosis factorα. HFD males and females also had increased insulin and increased TSH, and HFD females had decreased serum prolactin and growth hormone pulse amplitude. Pituitary single-cell transcriptomics revealed modest or no changes in pituitary cell gene expression from HFD males after 10 or 15 weeks or from HFD females after 10 weeks. However, HFD females (15 weeks) showed significant numbers of differentially expressed genes in lactotropes and pituitary stem cells. Collectively, these studies reveal that pituitary cells from males appear to be more resilient to the oxidative stress of obesity than females and identify the most vulnerable pituitary cell populations in females.
Collapse
Affiliation(s)
- Tiffany K Miles
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Arkansas Children's Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Victoria G Ortega
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Cole R Bolen
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Milla M Reddick
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ashley Herdman
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
36
|
Wong CP, Iwaniec UT, Turner RT. Brown adipose tissue but not tibia exhibits a dramatic response to acute reduction in environmental temperature in growing male mice. Bone Rep 2023; 19:101706. [PMID: 37637756 PMCID: PMC10448410 DOI: 10.1016/j.bonr.2023.101706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/29/2023] Open
Abstract
Mice are typically housed at room temperature (∼22 °C), which is well below their thermoneutral zone and results in cold stress. Chronic cold stress leads to increased adaptive thermogenesis and reductions in cancellous bone volume and bone marrow adipose tissue mass in long bones of growing mice. There is strong evidence that increased neuronal activity initiates the metabolic response of intrascapular brown adipose tissue (BAT) to cold stress, but it is less clear whether bone is regulated through a similar mechanism. Therefore, we compared the short-term response of BAT and whole tibia to a reduction in environmental temperature. To accomplish this, we transferred a group of 6-week-old male mice from 32 °C to 22 °C housing and sacrificed the mice 24 h later. Age-matched controls were maintained at 32 °C. We then evaluated expression levels of a panel of genes related to adipocyte differentiation and fat metabolism in BAT and tibia, and a panel of genes related to bone metabolism in tibia. The decrease in housing temperature resulted in changes in expression levels for 47/86 genes related to adipocyte differentiation and fat metabolism in BAT, including 9-fold and 17-fold increases in Ucp1 and Dio2, respectively. In contrast, only 1/86 genes related to adipocyte differentiation and fat metabolism and 4/84 genes related to bone metabolism were differentially expressed in tibia. These findings suggest that bone, although innervated with sensory and sympathetic neurons, does not respond as rapidly as BAT to changes in environmental temperature.
Collapse
Affiliation(s)
- Carmen P. Wong
- 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
| | - 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
| |
Collapse
|
37
|
Appana B, Queen NJ, Cao L. Protocol to minimize the confounding effect of cold stress on socially isolated mice using thermoneutral housing. STAR Protoc 2023; 4:102533. [PMID: 37660300 PMCID: PMC10491848 DOI: 10.1016/j.xpro.2023.102533] [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: 05/05/2023] [Revised: 06/26/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Social isolation, a risk factor for mortality and various disease states, in mice remains poorly understood, due in part to under-consideration of housing temperature and the murine thermoneutral zone. Here, we present a housing protocol to minimize the confounding effect of chronic cold stress on socially isolated mice that are unable to socially thermoregulate. We describe steps for allocating mice to group housing or social isolation conditions, housing mice in thermoneutral cabinets, feeding mice with high-fat diet, and measuring body weight, food intake, and metabolic indicators. For complete details on the use and execution of this protocol, please refer to Queen et al..1.
Collapse
Affiliation(s)
- Bhavya Appana
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nicholas J Queen
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
38
|
Tabh JKR, Nord A. Temperature-dependent Developmental Plasticity and Its Effects on Allen's and Bergmann's Rules in Endotherms. Integr Comp Biol 2023; 63:758-771. [PMID: 37160342 PMCID: PMC10503470 DOI: 10.1093/icb/icad026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/11/2023] Open
Abstract
Ecogeographical rules, describing common trends in animal form across space and time, have provided key insights into the primary factors driving species diversity on our planet. Among the most well-known ecogeographical rules are Bergmann's rule and Allen's rule, with each correlating ambient temperature to the size and shape of endotherms within a species. In recent years, these two rules have attracted renewed research attention, largely with the goal of understanding how they emerge (e.g., via natural selection or phenotypic plasticity) and, thus, whether they may emerge quickly enough to aid adaptations to a warming world. Yet despite this attention, the precise proximate and ultimate drivers of Bergmann's and Allen's rules remain unresolved. In this conceptual paper, we articulate novel and classic hypotheses for understanding whether and how plastic responses to developmental temperatures might contributed to each rule. Next, we compare over a century of empirical literature surrounding Bergmann's and Allen's rules against our hypotheses to uncover likely avenues by which developmental plasticity might drive temperature-phenotype correlations. Across birds and mammals, studies strongly support developmental plasticity as a driver of Bergmann's and Allen's rules, particularly with regards to Allen's rule. However, plastic contributions toward each rule appear largely non-linear and dependent upon: (1) efficiency of energy use (Bergmann's rule) and (2) thermal advantages (Allen's rule) at given ambient temperatures. These findings suggest that, among endotherms, rapid changes in body shape and size will continue to co-occur with our changing climate, but generalizing the direction of responses across populations is likely naive.
Collapse
Affiliation(s)
- Joshua K R Tabh
- Lund University, Department of Biology, Section for Evolutionary Ecology, Sölvegatan 37, SE-223 62, Sweden
| | - Andreas Nord
- Lund University, Department of Biology, Section for Evolutionary Ecology, Sölvegatan 37, SE-223 62, Sweden
| |
Collapse
|
39
|
Wittek L, Touma C, Nitezki T, Laeger T, Krämer S, Raila J. Reduction in Cold Stress in an Innovative Metabolic Cage Housing System Increases Animal Welfare in Laboratory Mice. Animals (Basel) 2023; 13:2866. [PMID: 37760266 PMCID: PMC10525209 DOI: 10.3390/ani13182866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Housing in metabolic cages can induce a pronounced stress response. Metabolic cage systems imply housing mice on metal wire mesh for the collection of urine and feces in addition to monitoring food and water intake. Moreover, mice are single-housed, and no nesting, bedding, or enrichment material is provided, which is often argued to have a not negligible impact on animal welfare due to cold stress. We therefore attempted to reduce stress during metabolic cage housing for mice by comparing an innovative metabolic cage (IMC) with a commercially available metabolic cage from Tecniplast GmbH (TMC) and a control cage. Substantial refinement measures were incorporated into the IMC cage design. In the frame of a multifactorial approach for severity assessment, parameters such as body weight, body composition, food intake, cage and body surface temperature (thermal imaging), mRNA expression of uncoupling protein 1 (Ucp1) in brown adipose tissue (BAT), fur score, and fecal corticosterone metabolites (CMs) were included. Female and male C57BL/6J mice were single-housed for 24 h in either conventional Macrolon cages (control), IMC, or TMC for two sessions. Body weight decreased less in the IMC (females-1st restraint: -6.94%; 2nd restraint: -6.89%; males-1st restraint: -8.08%; 2nd restraint: -5.82%) compared to the TMC (females-1st restraint: -13.2%; 2nd restraint: -15.0%; males-1st restraint: -13.1%; 2nd restraint: -14.9%) and the IMC possessed a higher cage temperature (females-1st restraint: 23.7 °C; 2nd restraint: 23.5 °C; males-1st restraint: 23.3 °C; 2nd restraint: 23.5 °C) compared with the TMC (females-1st restraint: 22.4 °C; 2nd restraint: 22.5 °C; males-1st restraint: 22.6 °C; 2nd restraint: 22.4 °C). The concentration of fecal corticosterone metabolites in the TMC (females-1st restraint: 1376 ng/g dry weight (DW); 2nd restraint: 2098 ng/g DW; males-1st restraint: 1030 ng/g DW; 2nd restraint: 1163 ng/g DW) was higher compared to control cage housing (females-1st restraint: 640 ng/g DW; 2nd restraint: 941 ng/g DW; males-1st restraint: 504 ng/g DW; 2nd restraint: 537 ng/g DW). Our results show the stress potential induced by metabolic cage restraint that is markedly influenced by the lower housing temperature. The IMC represents a first attempt to target cold stress reduction during metabolic cage application thereby producing more animal welfare friendlydata.
Collapse
Affiliation(s)
- Laura Wittek
- Department of Physiology and Pathophysiology of Nutrition, Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany (T.L.); (J.R.)
| | - Chadi Touma
- Department of Behavioural Biology, Osnabruck University, 49076 Osnabruck, Germany;
| | - Tina Nitezki
- Department of Physiology and Pathophysiology of Nutrition, Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany (T.L.); (J.R.)
| | - Thomas Laeger
- Department of Physiology and Pathophysiology of Nutrition, Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany (T.L.); (J.R.)
| | - Stephanie Krämer
- Interdisciplinary Center of 3Rs in Animal Research (ICAR3R), Clinic of Veterinary Medicine, Justus Liebig University of Giessen, 35392 Giessen, Germany;
| | - Jens Raila
- Department of Physiology and Pathophysiology of Nutrition, Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany (T.L.); (J.R.)
| |
Collapse
|
40
|
Alitalo O, González-Hernández G, Rosenholm M, Kohtala P, Matsui N, Müller HK, Theilmann W, Klein A, Kärkkäinen O, Rozov S, Rantamäki T, Kohtala S. Linking Hypothermia and Altered Metabolism with TrkB Activation. ACS Chem Neurosci 2023; 14:3212-3225. [PMID: 37551888 PMCID: PMC10485900 DOI: 10.1021/acschemneuro.3c00350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/13/2023] [Indexed: 08/09/2023] Open
Abstract
Many mechanisms have been proposed to explain acute antidepressant drug-induced activation of TrkB neurotrophin receptors, but several questions remain. In a series of pharmacological experiments, we observed that TrkB activation induced by antidepressants and several other drugs correlated with sedation, and most importantly, coinciding hypothermia. Untargeted metabolomics of pharmacologically dissimilar TrkB activating treatments revealed effects on shared bioenergetic targets involved in adenosine triphosphate (ATP) breakdown and synthesis, demonstrating a common perturbation in metabolic activity. Both activation of TrkB signaling and hypothermia were recapitulated by administration of inhibitors of glucose and lipid metabolism, supporting a close relationship between metabolic inhibition and neurotrophic signaling. Drug-induced TrkB phosphorylation was independent of electroencephalography slow-wave activity and remained unaltered in knock-in mice with the brain-derived neurotrophic factor (BDNF) Val66Met allele, which have impaired activity-dependent BDNF release, alluding to an activation mechanism independent from BDNF and neuronal activity. Instead, we demonstrated that the active maintenance of body temperature prevents activation of TrkB and other targets associated with antidepressants, including p70S6 kinase downstream of the mammalian target of rapamycin (mTOR) and glycogen synthase kinase 3β (GSK3β). Increased TrkB, GSK3β, and p70S6K phosphorylation was also observed during recovery sleep following sleep deprivation, when a physiological temperature drop is known to occur. Our results suggest that the changes in bioenergetics and thermoregulation are causally connected to TrkB activation and may act as physiological regulators of signaling processes involved in neuronal plasticity.
Collapse
Affiliation(s)
- Okko Alitalo
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Gemma González-Hernández
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Marko Rosenholm
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Center
for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Piia Kohtala
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Department
of Psychiatry, Weill Cornell Medicine, New York, New York 10021, United States
| | - Nobuaki Matsui
- Faculty
of Pharmacy, Gifu University of Medical
Science, 4-3-3 Nijigaoka,
Kani, Gifu 509-0293, Japan
| | - Heidi Kaastrup Müller
- Translational
Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus N 8200, Denmark
| | - Wiebke Theilmann
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Anders Klein
- Novo
Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen DK-2200, Denmark
- Department
of Drug Design & Pharmacology, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Olli Kärkkäinen
- School
of Pharmacy, University of Eastern Finland, Kuopio 70210, Finland
- Afekta
Technologies Ltd., Kuopio 70210, Finland
| | - Stanislav Rozov
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Tomi Rantamäki
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Samuel Kohtala
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Department
of Psychiatry, Weill Cornell Medicine, New York, New York 10021, United States
| |
Collapse
|
41
|
Grobe CC, Reho JJ, Brown-Williams D, Ziegler AA, Mathieu NM, Lawton SB, Fekete EM, Brozoski DT, Wackman KK, Burnett CM, Nakagawa P, Sigmund CD, Segar JL, Grobe JL. Cardiometabolic Effects of DOCA-Salt in Mice Depend on Ambient Temperature. Hypertension 2023; 80:1871-1880. [PMID: 37470185 PMCID: PMC10528934 DOI: 10.1161/hypertensionaha.122.20415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 07/10/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Mice prefer warmer environments than humans. For this reason, behavioral and physiological thermoregulatory responses are engaged by mice in response to a standard room temperature of 22 to 24 °C. Autonomic mechanisms mediating thermoregulatory responses overlap with mechanisms activated in hypertension, and, therefore, we hypothesized that housing at thermoneutral temperatures (TNs; 30 °C) would modify the cardiometabolic effects of deoxycorticosterone acetate (DOCA)-salt in mice. METHODS The effects of DOCA-salt treatment upon ingestive behaviors, energy expenditure, blood pressure, heart rate (HR), and core temperature were assessed in C57BL/6J mice housed at room temperature or TN. RESULTS Housing at TN reduced food intake, energy expenditure, blood pressure, and HR and attenuated HR responses to acute autonomic blockade by chlorisondamine. At room temperature, DOCA-salt caused expected increases in fluid intake, sodium retention in osmotically inactive pools, blood pressure, core temperature, and also caused expected decreases in fat-free mass, total body water, and HR. At TN, the effects of DOCA-salt upon fluid intake, fat gains, hydration, and core temperature were exaggerated, but effects on energy expenditure and HR were blunted. Effects of DOCA-salt upon blood pressure were similar for 3 weeks and exaggerated by TN housing in the fourth week. CONCLUSIONS Ambient temperature robustly influences behavioral and physiological functions in mice, including metabolic and cardiovascular phenotype development in response to DOCA-salt treatment. Studying cardiometabolic responses of mice at optimal ambient temperatures promises to improve the translational relevance of rodent models.
Collapse
Affiliation(s)
- Connie C. Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - John J. Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
| | | | - Alisha A. Ziegler
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Natalia M. Mathieu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Samuel B.R. Lawton
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Eva M. Fekete
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Daniel T. Brozoski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Kelsey K. Wackman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Colin M.L. Burnett
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jeffrey L. Segar
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
42
|
Dragan M, Chen Z, Li Y, Le J, Sun P, Haensel D, Sureshchandra S, Pham A, Lu E, Pham KT, Verlande A, Vu R, Gutierrez G, Li W, Jang C, Masri S, Dai X. Ovol1/2 loss-induced epidermal defects elicit skin immune activation and alter global metabolism. EMBO Rep 2023; 24:e56214. [PMID: 37249012 PMCID: PMC10328084 DOI: 10.15252/embr.202256214] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Skin epidermis constitutes the outer permeability barrier that protects the body from dehydration, heat loss, and myriad external assaults. Mechanisms that maintain barrier integrity in constantly challenged adult skin and how epidermal dysregulation shapes the local immune microenvironment and whole-body metabolism remain poorly understood. Here, we demonstrate that inducible and simultaneous ablation of transcription factor-encoding Ovol1 and Ovol2 in adult epidermis results in barrier dysregulation through impacting epithelial-mesenchymal plasticity and inflammatory gene expression. We find that aberrant skin immune activation then ensues, featuring Langerhans cell mobilization and T cell responses, and leading to elevated levels of secreted inflammatory factors in circulation. Finally, we identify failure to gain body weight and accumulate body fat as long-term consequences of epidermal-specific Ovol1/2 loss and show that these global metabolic changes along with the skin barrier/immune defects are partially rescued by immunosuppressant dexamethasone. Collectively, our study reveals key regulators of adult barrier maintenance and suggests a causal connection between epidermal dysregulation and whole-body metabolism that is in part mediated through aberrant immune activation.
Collapse
Affiliation(s)
- Morgan Dragan
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Zeyu Chen
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Department of Dermatology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Present address:
Institute of PsoriasisTongji University School of MedicineShanghaiChina
| | - Yumei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Johnny Le
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Peng Sun
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Daniel Haensel
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Program in Epithelial BiologyStanford University School of MedicineStanfordCAUSA
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Anh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Eddie Lu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Katherine Thanh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Amandine Verlande
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Remy Vu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Guadalupe Gutierrez
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Wei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Cholsoon Jang
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Selma Masri
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Xing Dai
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
- Department of Dermatology, School of MedicineUniversity of CaliforniaIrvineCAUSA
| |
Collapse
|
43
|
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
|
44
|
Kasza I, Cuncannan C, Michaud J, Nelson D, Yen CLE, Jain R, Simcox J, MacDougald OA, Parks BW, Alexander CM. "Humanizing" mouse environments: Humidity, diurnal cycles and thermoneutrality. Biochimie 2023; 210:82-98. [PMID: 36372307 PMCID: PMC10172392 DOI: 10.1016/j.biochi.2022.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022]
Abstract
Thermoneutral housing has been shown to promote more accurate and robust development of several pathologies in mice. Raising animal housing temperatures a few degrees may create a relatively straightforward opportunity to improve translatability of mouse models. In this commentary, we discuss the changes of physiology induced in mice housed at thermoneutrality, and review techniques for measuring systemic thermogenesis, specifically those affecting storage and mobilization of lipids in adipose depots. Environmental cues are a component of the information integrated by the brain to calculate food consumption and calorie deposition. We show that relative humidity is one of those cues, inducing a rapid sensory response that is converted to a more chronic susceptibility to obesity. Given high inter-institutional variability in the regulation of relative humidity, study reproducibility may be improved by consideration of this factor. We evaluate a "humanized" environmental cycling protocol, where mice sleep in warm temperature housing, and are cool during the wake cycle. We show that this protocol suppresses adaptation to cool exposure, with consequence for adipose-associated lipid storage. To evaluate systemic cues in mice housed at thermoneutral temperatures, we characterized the circulating lipidome, and show that sera are highly depleted in some HDL-associated phospholipids, specifically phospholipids containing the essential fatty acid, 18:2 linoleic acid, and its derivative, arachidonic acid (20:4) and related ether-phospholipids. Given the role of these fatty acids in inflammatory responses, we propose they may underlie the differences in disease progression observed at thermoneutrality.
Collapse
Affiliation(s)
- Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, United States
| | - Colleen Cuncannan
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, United States
| | - Julian Michaud
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, United States
| | - Dave Nelson
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States
| | - Chi-Liang E Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States
| | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, United States
| | - Judi Simcox
- Department of Biochemistry, University of Wisconsin-Madison, United States
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan, United States
| | - Brian W Parks
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States
| | - Caroline M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, United States.
| |
Collapse
|
45
|
Amin A, Badenes M, Tüshaus J, de Carvalho É, Burbridge E, Faísca P, Trávníčková K, Barros A, Carobbio S, Domingos PM, Vidal-Puig A, Moita LF, Maguire S, Stříšovský K, Ortega FJ, Fernández-Real JM, Lichtenthaler SF, Adrain C. Semaphorin 4B is an ADAM17-cleaved adipokine that inhibits adipocyte differentiation and thermogenesis. Mol Metab 2023; 73:101731. [PMID: 37121509 PMCID: PMC10197113 DOI: 10.1016/j.molmet.2023.101731] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023] Open
Abstract
OBJECTIVE The metalloprotease ADAM17 (also called TACE) plays fundamental roles in homeostasis by shedding key signaling molecules from the cell surface. Although its importance for the immune system and epithelial tissues is well-documented, little is known about the role of ADAM17 in metabolic homeostasis. The purpose of this study was to determine the impact of ADAM17 expression, specifically in adipose tissues, on metabolic homeostasis. METHODS We used histopathology, molecular, proteomic, transcriptomic, in vivo integrative physiological and ex vivo biochemical approaches to determine the impact of adipose tissue-specific deletion of ADAM17 upon adipocyte and whole organism metabolic physiology. RESULTS ADAM17adipoq-creΔ/Δ mice exhibited a hypermetabolic phenotype characterized by elevated energy consumption and increased levels of adipocyte thermogenic gene expression. On a high fat diet, these mice were more thermogenic, while exhibiting elevated expression levels of genes associated with lipid oxidation and lipolysis. This hypermetabolic phenotype protected mutant mice from obesogenic challenge, limiting weight gain, hepatosteatosis and insulin resistance. Activation of beta-adrenoceptors by the neurotransmitter norepinephrine, a key regulator of adipocyte physiology, triggered the shedding of ADAM17 substrates, and regulated ADAM17 expression at the mRNA and protein levels, hence identifying a functional connection between thermogenic licensing and the regulation of ADAM17. Proteomic studies identified Semaphorin 4B (SEMA4B), as a novel ADAM17-shed adipokine, whose expression is regulated by physiological thermogenic cues, that acts to inhibit adipocyte differentiation and dampen thermogenic responses in adipocytes. Transcriptomic data showed that cleaved SEMA4B acts in an autocrine manner in brown adipocytes to repress the expression of genes involved in adipogenesis, thermogenesis, and lipid uptake, storage and catabolism. CONCLUSIONS Our findings identify a novel ADAM17-dependent axis, regulated by beta-adrenoceptors and mediated by the ADAM17-cleaved form of SEMA4B, that modulates energy balance in adipocytes by inhibiting adipocyte differentiation, thermogenesis and lipid catabolism.
Collapse
Affiliation(s)
- Abdulbasit Amin
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | - Marina Badenes
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Faculty of Veterinary Medicine, Lusofona University, Lisbon, Portugal; Faculty of Veterinary Nursing, Polytechnic Institute of Lusofonia, Lisbon, Portugal
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Érika de Carvalho
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Instituto de Tecnologia Química da Universidade Nova de Lisboa (ITQB-Nova), Oeiras, Portugal
| | - Emma Burbridge
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland
| | - Pedro Faísca
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Květa Trávníčková
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - André Barros
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Stefania Carobbio
- Centro de Investigacíon Principe Felipe (CIPF), Valencia, Spain; Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, UK
| | - Pedro M Domingos
- Instituto de Tecnologia Química da Universidade Nova de Lisboa (ITQB-Nova), Oeiras, Portugal
| | - Antonio Vidal-Puig
- Centro de Investigacíon Principe Felipe (CIPF), Valencia, Spain; Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, UK
| | - Luís F Moita
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Sarah Maguire
- Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland
| | - Kvido Stříšovský
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Francisco J Ortega
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Department of Medical Sciences, University of Girona, Girona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), and Institute of Salud Carlos III (ISCIII), Madrid, Spain
| | - José Manuel Fernández-Real
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Department of Medical Sciences, University of Girona, Girona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), and Institute of Salud Carlos III (ISCIII), Madrid, Spain
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Colin Adrain
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland.
| |
Collapse
|
46
|
Motyl KJ, MacDougald OA. Bring the heat: Thermal stress, metabolic plasticity, and considerations for biomedical research. Biochimie 2023; 210:1-2. [PMID: 37331726 DOI: 10.1016/j.biochi.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Affiliation(s)
- Katherine J Motyl
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, United States; Tufts University School of Medicine, Tufts University, United States.
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan, United States.
| |
Collapse
|
47
|
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: 9] [Impact Index Per Article: 4.5] [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
|
48
|
Langlais AL, Mountain RV, Kunst RF, Barlow D, Houseknecht KL, Motyl KJ. Thermoneutral housing does not rescue olanzapine-induced trabecular bone loss in C57BL/6J female mice. Biochimie 2023; 210:50-60. [PMID: 37236340 PMCID: PMC10357956 DOI: 10.1016/j.biochi.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023]
Abstract
Antipsychotic drugs are prescribed to a wide range of individuals to treat mental health conditions including schizophrenia. However, antipsychotic drugs cause bone loss and increase fracture risk. We previously found that the atypical antipsychotic (AA) drug risperidone causes bone loss through multiple pharmacological mechanisms, including activation of the sympathetic nervous system in mice treated with clinically relevant doses. However, bone loss was dependent upon housing temperature, which modulates sympathetic activity. Another AA drug, olanzapine, has substantial metabolic side effects, including weight gain and insulin resistance, but it is unknown whether bone and metabolic outcomes of olanzapine are also dependent upon housing temperature in mice. We therefore treated eight week-old female mice with vehicle or olanzapine for four weeks, housed at either room temperature (23 °C) or thermoneutrality (28-30 °C), which has previously been shown to be positive for bone. Olanzapine caused significant trabecular bone loss (-13% BV/TV), likely through increased RANKL-dependent osteoclast resorption, which was not suppressed by thermoneutral housing. Additionally, olanzapine inhibited cortical bone expansion at thermoneutrality, but did not alter cortical bone expansion at room temperature. Olanzapine also increased markers of thermogenesis within brown and inguinal adipose depots independent of housing temperature. Overall, olanzapine causes trabecular bone loss and inhibits the positive effect of thermoneutral housing on bone. Understanding how housing temperature modulates the impact of AA drugs on bone is important for future pre-clinical studies, as well as for the prescription of AA drugs, particularly to older adults and adolescents who are most vulnerable to the effects on bone.
Collapse
Affiliation(s)
- Audrie L Langlais
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Rebecca V Mountain
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Roni F Kunst
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Deborah Barlow
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA
| | - Karen L Houseknecht
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA
| | - Katherine J Motyl
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA; Tufts University School of Medicine, Tufts University, Boston, MA, USA.
| |
Collapse
|
49
|
Sattgast LH, Wong CP, Branscum AJ, Olson DA, Aguirre-Burk AM, Iwaniec UT, Turner RT. Small changes in thermoregulation influence cancellous bone turnover balance in distal femur metaphysis in growing female mice. Bone Rep 2023; 18:101675. [PMID: 37007217 PMCID: PMC10063413 DOI: 10.1016/j.bonr.2023.101675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Mice are typically housed at temperatures well below their thermoneutral zone. When individually housed at room temperature (~22 °C) mice experience cold stress which results in cancellous bone loss and has the potential to alter the skeletal response to treatment. It is not clear if there is a threshold temperature for cold stress-induced bone loss. It is also not clear if alternative strategies for attenuating cold stress, such as group housing, influence bone accrual and turnover. This study aimed to determine how small differences in temperature (4 °C) or heat loss (individual versus group housing with nestlets) influence bone in growing female C57BL/6 J mice. Five-week-old mice were randomized by weight to 1 of 4 treatment groups (N = 10/group): 1) baseline, 2) single housed at 22 °C, 3) single housed at 26 °C, or 4) group housed (n = 5/cage) with nestlets at 22 °C. Mice in the baseline group were sacrificed 1 week later, at 6 weeks of age. The other 3 groups of mice were maintained at their respective temperatures and housing conditions for 13 weeks until 18 weeks of age. Compared to baseline, mice single housed at room temperature had increased body weight and femur size, but dramatically decreased cancellous bone volume fraction in distal femur metaphysis. The cancellous bone loss was attenuated but not prevented in mice individually housed at 26 °C or group housed at 22 °C. In conclusion, by impacting thermogenesis or heat loss, modest differences in housing conditions could influence experimental results.
Collapse
Affiliation(s)
- Lara H. Sattgast
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Carmen P. Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Adam J. Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Dawn A. Olson
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Allan M. Aguirre-Burk
- 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
| | - 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
- Corresponding author at: Skeletal Biology Laboratory, School of Biological and Population Health Sciences, 127 Milam Hall, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
50
|
Osborne-Lawrence S, Lawrence C, Metzger NP, Klavon J, Baig HR, Richard C, Varshney S, Gupta D, Singh O, Ogden SB, Shankar K, Paul S, Butler RK, Zigman JM. Effects of thermoneutrality on food intake, body weight, and body composition in a Prader-Willi syndrome mouse model. Obesity (Silver Spring) 2023; 31:1644-1654. [PMID: 37161883 PMCID: PMC12002079 DOI: 10.1002/oby.23766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/28/2023] [Accepted: 02/28/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE Prader-Willi syndrome (PWS) is a multisystem genetic disorder. Unfortunately, none of several mouse models carrying PWS mutations emulates the entirety of the human PWS phenotype, including hyperphagia plus obesity. METHODS To determine whether housing at thermoneutrality (TN, 30 °C) permits the development of hyperphagia and obesity in the Snord116del PWS mouse model, the effects of housing three different ages of Snord116del and wild-type (WT) littermates at TN versus room temperature (RT, 22-24 °C) for 8 weeks were compared. RESULTS Snord116del mice born and maintained at TN exhibited lower body weight curves, lower percentage fat mass, and lower food intake than WT mice at RT. In 4- to 6-month-old high-fat diet-fed female mice, TN raised the Snord116del body weight curve closer to that of RT-housed WT mice although the TN-housed Snord116del mice did not gain more adiposity or exhibit greater food intake. In 6- to 8-month-old high-fat diet-fed male mice, body weight, adiposity, and food intake of TN-housed Snord116del mice remained far below levels in RT-housed WT mice. TN elicited hypotonia in Snord116del adults and exacerbated mortality of Snord116del newborns. CONCLUSIONS In none of three tested TN protocols were greater food intake, body weight, or adiposity induced in Snord116del mice compared with RT-housed WT mice.
Collapse
Affiliation(s)
- Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Connor Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Nathan P. Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Julia Klavon
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Hassan R. Baig
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Corine Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sean B. Ogden
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Subhojit Paul
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ryan K. Butler
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA
- O’Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas, USA
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA
- O’Donnell Brain Institute, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|