1
|
Mota CMD, Madden CJ. Neural circuits of long-term thermoregulatory adaptations to cold temperatures and metabolic demands. Nat Rev Neurosci 2024; 25:143-158. [PMID: 38316956 DOI: 10.1038/s41583-023-00785-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 02/07/2024]
Abstract
The mammalian brain controls heat generation and heat loss mechanisms that regulate body temperature and energy metabolism. Thermoeffectors include brown adipose tissue, cutaneous blood flow and skeletal muscle, and metabolic energy sources include white adipose tissue. Neural and metabolic pathways modulating the activity and functional plasticity of these mechanisms contribute not only to the optimization of function during acute challenges, such as ambient temperature changes, infection and stress, but also to longitudinal adaptations to environmental and internal changes. Exposure of humans to repeated and seasonal cold ambient conditions leads to adaptations in thermoeffectors such as habituation of cutaneous vasoconstriction and shivering. In animals that undergo hibernation and torpor, neurally regulated metabolic and thermoregulatory adaptations enable survival during periods of significant reduction in metabolic rate. In addition, changes in diet can activate accessory neural pathways that alter thermoeffector activity. This knowledge may be harnessed for therapeutic purposes, including treatments for obesity and improved means of therapeutic hypothermia.
Collapse
Affiliation(s)
- Clarissa M D Mota
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Christopher J Madden
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
2
|
Yamaguchi H, Murphy KR, Fukatsu N, Sato K, Yamanaka A, de Lecea L. Dorsomedial and preoptic hypothalamic circuits control torpor. Curr Biol 2023; 33:5381-5389.e4. [PMID: 37992720 DOI: 10.1016/j.cub.2023.10.076] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/25/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023]
Abstract
Endotherms can survive low temperatures and food shortage by actively entering a hypometabolic state known as torpor. Although the decrease in metabolic rate and body temperature (Tb) during torpor is controlled by the brain, the specific neural circuits underlying these processes have not been comprehensively elucidated. In this study, we identify the neural circuits involved in torpor regulation by combining whole-brain mapping of torpor-activated neurons, cell-type-specific manipulation of neural activity, and viral tracing-based circuit mapping. We find that Trpm2-positive neurons in the preoptic area and Vgat-positive neurons in the dorsal medial hypothalamus are activated during torpor. Genetic silencing shows that the activity of either cell type is necessary to enter the torpor state. Finally, we show that these cells receive projections from the arcuate and suprachiasmatic nucleus and send projections to brain regions involved in thermoregulation. Our results demonstrate an essential role of hypothalamic neurons in the regulation of Tb and metabolic rate during torpor and identify critical nodes of the torpor regulatory network.
Collapse
Affiliation(s)
- Hiroshi Yamaguchi
- Department of Neural Regulation, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan; PRESTO, Japan Science and Technology Agency (JST), Tokyo, Japan.
| | - Keith R Murphy
- Department of Psychiatry and Behavioral Sciences, Stanford University, 1201 Welch Road, Stanford, CA 94305, USA
| | - Noriaki Fukatsu
- Department of System Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 464-8601, Japan; Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 464-8601, Japan
| | - Kazuhide Sato
- Institute for Advanced Research, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | | | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, 1201 Welch Road, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Mousavi S, Qiu H, Andrews MT, Checco JW. Peptidomic Analysis Reveals Seasonal Neuropeptide and Peptide Hormone Changes in the Hypothalamus and Pituitary of a Hibernating Mammal. ACS Chem Neurosci 2023; 14:2569-2581. [PMID: 37395621 PMCID: PMC10529138 DOI: 10.1021/acschemneuro.3c00268] [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] [Indexed: 07/04/2023] Open
Abstract
During the winter, hibernating mammals undergo extreme changes in physiology, which allow them to survive several months without access to food. These animals enter a state of torpor, which is characterized by decreased metabolism, near-freezing body temperatures, and a dramatically reduced heart rate. The neurochemical basis of this regulation is largely unknown. Based on prior evidence suggesting that the peptide-rich hypothalamus plays critical roles in hibernation, we hypothesized that changes in specific cell-cell signaling peptides (neuropeptides and peptide hormones) underlie physiological changes during torpor/arousal cycles. To test this hypothesis, we used a mass spectrometry-based peptidomics approach to examine seasonal changes of endogenous peptides that occur in the hypothalamus and pituitary of a model hibernating mammal, the thirteen-lined ground squirrel (Ictidomys tridecemlineatus). In the pituitary, we observed changes in several distinct peptide hormones as animals prepared for torpor in October, exited torpor in March, and progressed from spring (March) to fall (August). In the hypothalamus, we observed an overall increase in neuropeptides in October (pre-torpor), a decrease as the animal entered torpor, and an increase in a subset of neuropeptides during normothermic interbout arousals. Notable changes were observed for feeding regulatory peptides, opioid peptides, and several peptides without well-established functions. Overall, our study provides critical insight into changes in endogenous peptides in the hypothalamus and pituitary during mammalian hibernation that were not available from transcriptomic measurements. Understanding the molecular basis of the hibernation phenotype may pave the way for future efforts to employ hibernation-like strategies for organ preservation, combating obesity, and treatment of stroke.
Collapse
Affiliation(s)
- Somayeh Mousavi
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Haowen Qiu
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| | - Matthew T. Andrews
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - James W. Checco
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, NE 68588, United States
| |
Collapse
|
4
|
Ambler M, Hitrec T, Pickering A. Turn it off and on again: characteristics and control of torpor. Wellcome Open Res 2022; 6:313. [PMID: 35087956 PMCID: PMC8764563 DOI: 10.12688/wellcomeopenres.17379.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 11/20/2022] Open
Abstract
Torpor is a hypothermic, hypoactive, hypometabolic state entered into by a wide range of animals in response to environmental challenge. This review summarises the current understanding of torpor. We start by describing the characteristics of the wide-ranging physiological adaptations associated with torpor. Next follows a discussion of thermoregulation, control of food intake and energy expenditure, and the interactions of sleep and thermoregulation, with particular emphasis on how those processes pertain to torpor. We move on to review the evidence for the systems that control torpor entry, including both the efferent circulating factors that signal the need for torpor, and the central processes that orchestrate it. Finally, we consider how the putative circuits responsible for torpor induction integrate with the established understanding of thermoregulation under non-torpid conditions and highlight important areas of uncertainty for future studies.
Collapse
Affiliation(s)
- Michael Ambler
- School of Physiology, Pharmacology, & Neuroscience, University of Bristol, Bristol, Bristol, BS8 1TD, UK
| | - Timna Hitrec
- School of Physiology, Pharmacology, & Neuroscience, University of Bristol, Bristol, Bristol, BS8 1TD, UK
| | - Anthony Pickering
- School of Physiology, Pharmacology, & Neuroscience, University of Bristol, Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
5
|
Junkins MS, Bagriantsev SN, Gracheva EO. Towards understanding the neural origins of hibernation. J Exp Biol 2022; 225:273864. [PMID: 34982152 DOI: 10.1242/jeb.229542] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hibernators thrive under harsh environmental conditions instead of initiating canonical behavioral and physiological responses to promote survival. Although the physiological changes that occur during hibernation have been comprehensively researched, the role of the nervous system in this process remains relatively underexplored. In this Review, we adopt the perspective that the nervous system plays an active, essential role in facilitating and supporting hibernation. Accumulating evidence strongly suggests that the hypothalamus enters a quiescent state in which powerful drives to thermoregulate, eat and drink are suppressed. Similarly, cardiovascular and pulmonary reflexes originating in the brainstem are altered to permit the profoundly slow heart and breathing rates observed during torpor. The mechanisms underlying these changes to the hypothalamus and brainstem are not currently known, but several neuromodulatory systems have been implicated in the induction and maintenance of hibernation. The intersection of these findings with modern neuroscience approaches, such as optogenetics and in vivo calcium imaging, has opened several exciting avenues for hibernation research.
Collapse
Affiliation(s)
- Madeleine S Junkins
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.,Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.,Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| |
Collapse
|
6
|
Marine Neurotoxins' Effects on Environmental and Human Health: An OMICS Overview. Mar Drugs 2021; 20:md20010018. [PMID: 35049872 PMCID: PMC8778346 DOI: 10.3390/md20010018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
Harmful algal blooms (HAB), and the consequent release of toxic metabolites, can be responsible for seafood poisoning outbreaks. Marine wildlife can accumulate these toxins throughout the food chain, which presents a threat to consumers’ health. Some of these toxins, such as saxitoxin (STX), domoic acid (DA), ciguatoxin (CTX), brevetoxin (BTX), tetrodotoxin (TTX), and β-N-methylamino-L-alanine (BMAA), cause severe neurological symptoms in humans. Considerable information is missing, however, notably the consequences of toxin exposures on changes in gene expression, protein profile, and metabolic pathways. This information could lead to understanding the consequence of marine neurotoxin exposure in aquatic organisms and humans. Nevertheless, recent contributions to the knowledge of neurotoxins arise from OMICS-based research, such as genomics, transcriptomics, proteomics, and metabolomics. This review presents a comprehensive overview of the most recent research and of the available solutions to explore OMICS datasets in order to identify new features in terms of ecotoxicology, food safety, and human health. In addition, future perspectives in OMICS studies are discussed.
Collapse
|
7
|
Ambler M, Hitrec T, Pickering A. Turn it off and on again: characteristics and control of torpor. Wellcome Open Res 2021; 6:313. [DOI: 10.12688/wellcomeopenres.17379.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 11/20/2022] Open
Abstract
Torpor is a hypothermic, hypoactive, hypometabolic state entered into by a wide range of animals in response to environmental challenge. This review summarises the current understanding of torpor. We start by describing the characteristics of the wide-ranging physiological adaptations associated with torpor. Next follows a discussion of thermoregulation, control of food intake and energy expenditure, and the interactions of sleep and thermoregulation, with particular emphasis on how those processes pertain to torpor. We move on to take a critical view of the evidence for the systems that control torpor entry, including both the efferent circulating factors that signal the need for torpor, and the central processes that orchestrate it. Finally, we consider how the putative circuits responsible for torpor induction integrate with the established understanding of thermoregulation under non-torpid conditions and highlight important areas of uncertainty for future studies.
Collapse
|
8
|
Jaschke N, Pählig S, Pan YX, Hofbauer LC, Göbel A, Rachner TD. From Pharmacology to Physiology: Endocrine Functions of μ-Opioid Receptor Networks. Trends Endocrinol Metab 2021; 32:306-319. [PMID: 33676828 PMCID: PMC8035298 DOI: 10.1016/j.tem.2021.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 01/04/2023]
Abstract
The steady rise in opioid users and abusers has uncovered multiple detrimental health consequences of perturbed opioid receptor signaling, thereby creating the need to better understand the biology of these systems. Among endogenous opioid networks, μ-receptors have received special attention due to their unprecedented biological complexity and broad implications in homeostatic functions. Here, we review the origin, molecular biology, and physiology of endogenous opioids with a special focus on μ-opioid receptor networks within the endocrine system. Moreover, we summarize the current evidence supporting an involvement of the latter in regulating distinct endocrine functions. Finally, we combine these insights to present an integrated perspective on μ-opioid receptor biology and provide an outlook on future studies and unresolved questions in this field.
Collapse
Affiliation(s)
- Nikolai Jaschke
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| | - Sophie Pählig
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ying-Xian Pan
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Andy Göbel
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Ouchi Y, Yamato M, Chowdhury VS, Bungo T. Adenosine 5'-monophosphate induces hypothermia and alters gene expressions in the brain and liver of chicks. Brain Res Bull 2021; 172:14-21. [PMID: 33862124 DOI: 10.1016/j.brainresbull.2021.04.008] [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: 01/19/2021] [Revised: 04/05/2021] [Accepted: 04/10/2021] [Indexed: 10/21/2022]
Abstract
The adenosine A1 receptor is important for body temperature regulation in mammals; however, little is known about its function in avian species. In this study, we investigated the effects of the adenosine A1 receptor agonist and antagonist (adenosine 5'-monophosphate [5'-AMP] and 8 p-sulfophenyl theophylline [8-SPT], respectively) on thermoregulation in chickens. Male chicks were used in this study. After administration of 5'-AMP and 8-SPT, the rectal temperature, plasma metabolites, and gene expressions in the hypothalamus and liver were measured. The rectal temperature was reduced by peripheral administration of 5'-AMP, and the hypothermic effect of 5'-AMP was attenuated by central injection of 8-SPT in chicks. In the hypothalamus, the mRNA level of the agouti-related protein (AgRP) was increased by 5'-AMP administration, whereas it was suppressed by 8-SPT. The plasma levels of free fatty acid were elevated in 5'-AMP-treated chicks and that elevation was suppressed by the 8-SPT treatment. The gene expression of proopiomelanocortin in the hypothalamus was affected by 8-SPT. Nevertheless, the gene expressions of the thermoregulation-related genes, such as the thyrotropin-releasing hormone, were not affected by 5'-AMP and 8-SPT. Hepatic gene expressions related to lipid intake and metabolism were suppressed by 5'-AMP. However, the gene expression of the uncoupling protein was upregulated by 5'-AMP. Based on these results, birds, like mammals, will undergo adenosine A1 receptor-induced hypothermia. In conclusion, it is suggested that 5'-AMP-mediated hypothermia via the adenosine A1 receptor may affect the central melanocortin system and suppress hepatic lipid metabolism in chickens.
Collapse
Affiliation(s)
- Yoshimitsu Ouchi
- Laboratory of Animal Behavior and Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi, Hiroshima, 739-8528, Japan
| | - Miko Yamato
- Faculty of Applied Biological Science, Hiroshima University, Higashi, Hiroshima, 739-8528, Japan
| | | | - Takashi Bungo
- Laboratory of Animal Behavior and Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi, Hiroshima, 739-8528, Japan.
| |
Collapse
|
10
|
Shimaoka H, Shiina T, Suzuki H, Horii Y, Horii K, Shimizu Y. Successful induction of deep hypothermia by isoflurane anesthesia and cooling in a non-hibernator, the rat. J Physiol Sci 2021; 71:10. [PMID: 33784982 PMCID: PMC10717611 DOI: 10.1186/s12576-021-00794-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 03/03/2021] [Indexed: 11/10/2022]
Abstract
The aim of the present study was to establish a novel method for inducing deep hypothermia in rats. Cooling rats anesthetized with isoflurane caused a time-dependent decrease in rectal temperature, but cardiac arrest occurred before their body temperature reached 20 °C when isoflurane inhalation was continued during the cooling process. Stopping inhalation of isoflurane when the rectal temperature reached 22.5 °C successfully induced deep hypothermia, although stopping the inhalation at 27.5 °C resulted in spontaneous recovery of rectal temperature. The hypothermic condition was able to be maintained for up to 6 h. A large number of c-Fos-positive cells were detected in the hypothalamus during hypothermia. Both the maintenance of and recovery from hypothermia caused organ injury, but the damage was transient and recovered within 1 week. These findings indicate that the established procedure is appropriate for inducing deep hypothermia without accompanying serious organ injury in rats.
Collapse
Affiliation(s)
- Hiroki Shimaoka
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Takahiko Shiina
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, 1-1 Yanagido, Gifu, 501-1193, Japan.
| | - Hayato Suzuki
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yuuki Horii
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kazuhiro Horii
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yasutake Shimizu
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, 1-1 Yanagido, Gifu, 501-1193, Japan
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| |
Collapse
|
11
|
Frare C, Williams CT, Drew KL. Thermoregulation in hibernating mammals: The role of the "thyroid hormones system". Mol Cell Endocrinol 2021; 519:111054. [PMID: 33035626 PMCID: PMC8091518 DOI: 10.1016/j.mce.2020.111054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 07/15/2020] [Accepted: 10/04/2020] [Indexed: 12/19/2022]
Abstract
Hibernation is a unique evolutionary adaptation to conserve energy. During the pre-hibernation (i.e. fall) season, a progressive decline in core body temperature and further decrease in metabolism underlie a seasonal modulation in thermoregulation. The onset of hibernation requires marked changes in thermoregulatory attributes including adjustment in body temperature and tissue specific increases in thermogenic capacity. The hibernation season is characterized by a regulated suppression in thermogenesis allowing the onset of torpor interrupted by periodic activation of thermogenesis to sustain interbout arousals. Thyroid hormones are known to regulate both body temperature and metabolism, and for this reason, the hypothalamic-pituitary-thyroid axis and thyroid hormones have been investigated as modulators of thermogenesis in the phenomenon of hibernation, but the mechanisms remain poorly understood. In this review, we present an overview of what is known about the thermogenic roles of thyroid hormones in hibernating species across seasons and within the hibernating season (torpor-interbout arousal cycle). Overall, the hypothalamic-pituitary-thyroid axis and thyroid hormones play a role in the pre-hibernation season to enhance thermogenic capacity. During hibernation, thermogenesis is attenuated at the level of sympathetic premotor neurons within the raphe pallidus and by deiodinase expression in the hypothalamus. Further, as recent work highlights the direct effect of thyroid hormones within the central nervous system in activating thermogenesis, we speculate how similar mechanisms may occur in hibernating species to modulate thermogenesis across seasons and to sustain interbout arousals. However, further experiments are needed to elucidate the role of thyroid hormones in hibernation, moving towards the understanding that thyroid hormones metabolism, transport and availability within tissues may be the most telling indicator of thyroid status.
Collapse
Affiliation(s)
- C Frare
- Department of Chemistry and Biochemistry University of Alaska Fairbanks, Fairbanks, AK, 99775, USA; Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA
| | - Cory T Williams
- Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA; Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, 99775, USA
| | - Kelly L Drew
- Department of Chemistry and Biochemistry University of Alaska Fairbanks, Fairbanks, AK, 99775, USA; Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA.
| |
Collapse
|
12
|
Temperature-Dependent Alternative Splicing of Precursor mRNAs and Its Biological Significance: A Review Focused on Post-Transcriptional Regulation of a Cold Shock Protein Gene in Hibernating Mammals. Int J Mol Sci 2020; 21:ijms21207599. [PMID: 33066638 PMCID: PMC7590145 DOI: 10.3390/ijms21207599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 01/18/2023] Open
Abstract
Multiple mRNA isoforms are often generated during processing such as alternative splicing of precursor mRNAs (pre-mRNA), resulting in a diversity of generated proteins. Alternative splicing is an essential mechanism for the functional complexity of eukaryotes. Temperature, which is involved in all life activities at various levels, is one of regulatory factors for controlling patterns of alternative splicing. Temperature-dependent alternative splicing is associated with various phenotypes such as flowering and circadian clock in plants and sex determination in poikilothermic animals. In some specific situations, temperature-dependent alternative splicing can be evoked even in homothermal animals. For example, the splicing pattern of mRNA for a cold shock protein, cold-inducible RNA-binding protein (CIRP or CIRBP), is changed in response to a marked drop in body temperature during hibernation of hamsters. In this review, we describe the current knowledge about mechanisms and functions of temperature-dependent alternative splicing in plants and animals. Then we discuss the physiological significance of hypothermia-induced alternative splicing of a cold shock protein gene in hibernating and non-hibernating animals.
Collapse
|
13
|
Frare C, Jenkins ME, McClure KM, Drew KL. Seasonal decrease in thermogenesis and increase in vasoconstriction explain seasonal response to N 6 -cyclohexyladenosine-induced hibernation in the Arctic ground squirrel (Urocitellus parryii). J Neurochem 2019; 151:316-335. [PMID: 31273780 PMCID: PMC6819227 DOI: 10.1111/jnc.14814] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/12/2019] [Accepted: 07/01/2019] [Indexed: 01/07/2023]
Abstract
Hibernation is a seasonal phenomenon characterized by a drop in metabolic rate and body temperature. Adenosine A1 receptor agonists promote hibernation in different mammalian species, and the understanding of the mechanism inducing hibernation will inform clinical strategies to manipulate metabolic demand that are fundamental to conditions such as obesity, metabolic syndrome, and therapeutic hypothermia. Adenosine A1 receptor agonist-induced hibernation in Arctic ground squirrels is regulated by an endogenous circannual (seasonal) rhythm. This study aims to identify the neuronal mechanism underlying the seasonal difference in response to the adenosine A1 receptor agonist. Arctic ground squirrels were implanted with body temperature transmitters and housed at constant ambient temperature (2°C) and light cycle (4L:20D). We administered CHA (N6 -cyclohexyladenosine), an adenosine A1 receptor agonist in euthermic-summer phenotype and euthermic-winter phenotype and used cFos and phenotypic immunoreactivity to identify cell groups affected by season and treatment. We observed lower core and subcutaneous temperature in winter animals and CHA produced a hibernation-like response in winter, but not in summer. cFos-ir was greater in the median preoptic nucleus and the raphe pallidus in summer after CHA. CHA administration also resulted in enhanced cFos-ir in the nucleus tractus solitarius and decreased cFos-ir in the tuberomammillary nucleus in both seasons. In winter, cFos-ir was greater in the supraoptic nucleus and lower in the raphe pallidus than in summer. The seasonal decrease in the thermogenic response to CHA and the seasonal increase in vasoconstriction, assessed by subcutaneous temperature, reflect the endogenous seasonal modulation of the thermoregulatory systems necessary for CHA-induced hibernation. Cover Image for this issue: doi: 10.1111/jnc.14528.
Collapse
Affiliation(s)
- Carla Frare
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, Alaska, USA
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - Mackenzie E Jenkins
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - Kelsey M McClure
- Department of Veterinary Medicine, Colorado State University, Fort Collins, Colorado, USA
| | - Kelly L Drew
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, Alaska, USA
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| |
Collapse
|
14
|
Horii Y, Shimaoka H, Horii K, Shiina T, Shimizu Y. Mild hypothermia causes a shift in the alternative splicing of cold-inducible RNA-binding protein transcripts in Syrian hamsters. Am J Physiol Regul Integr Comp Physiol 2019; 317:R240-R247. [PMID: 31188649 DOI: 10.1152/ajpregu.00012.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cold-shock proteins are thought to participate in the cold-tolerant nature of hibernating animals. We previously demonstrated that an alternative splicing may allow rapid induction of functional cold-inducible RNA-binding protein (CIRBP) in the hamster heart. The purpose of the present study was to determine the major cause of the alternative splicing in Syrian hamsters. RT-PCR analysis revealed that CIRBP mRNA is constitutively expressed in the heart, brain, lung, liver, and kidney of nonhibernating euthermic hamsters with several alternative splicing variants. In contrast, the short variant containing an open-reading frame for functional CIRBP was dominantly found in the hibernating animals. Keeping the animals in a cold and dark environment did not cause a shift in the alternative splicing. Induction of hypothermia by central administration of an adenosine A1-receptor agonist reproduced the shift in the splicing pattern. However, the agonist failed to shift the pattern when body temperature was kept at 37°C, suggesting that central adenosine A1 receptors are not directly linked to the shift of the alternative splicing. Rapid reduction of body temperature to 10°C by isoflurane anesthesia combined with cooling did not alter the splicing pattern, but maintenance of mild hypothermia (~28°C) for 2 h elicited the shift in the pattern. The results suggest that animals need to be maintained at mild hypothermia for an adequate duration to induce the shift in the alternative splicing. This is applicable to natural hibernation because hamsters entering hibernation show a gradual decrease in body temperature, being maintained at mild hypothermia for several hours.
Collapse
Affiliation(s)
- Yuuki Horii
- Laboratory of Physiology, Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University , Gifu , Japan
| | - Hiroki Shimaoka
- Laboratory of Physiology, Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University , Gifu , Japan
| | - Kazuhiro Horii
- Laboratory of Physiology, Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University , Gifu , Japan
| | - Takahiko Shiina
- Laboratory of Physiology, Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University , Gifu , Japan
| | - Yasutake Shimizu
- Laboratory of Physiology, Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University , Gifu , Japan.,Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University , Gifu , Japan
| |
Collapse
|
15
|
Chayama Y, Ando L, Sato Y, Shigenobu S, Anegawa D, Fujimoto T, Taii H, Tamura Y, Miura M, Yamaguchi Y. Molecular Basis of White Adipose Tissue Remodeling That Precedes and Coincides With Hibernation in the Syrian Hamster, a Food-Storing Hibernator. Front Physiol 2019; 9:1973. [PMID: 30745884 PMCID: PMC6360343 DOI: 10.3389/fphys.2018.01973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/31/2018] [Indexed: 12/31/2022] Open
Abstract
Mammalian hibernators store fat extensively in white adipose tissues (WATs) during pre-hibernation period (Pre-HIB) to prepare for hibernation. However, the molecular mechanisms underlying the pre-hibernation remodeling of WAT have not been fully elucidated. Syrian hamsters, a food-storing hibernator, can hibernate when exposed to a winter-like short day photoperiod and cold ambient temperature (SD-Cold). Animals subjected to prolonged SD-Cold had smaller white adipocytes and beige-like cells within subcutaneous inguinal WAT (iWAT). Time-course analysis of gene expression with RNA-sequencing and quantitative PCR demonstrated that the mRNA expression of not only genes involved in lipid catabolism (lipolysis and beta-oxidation) but also lipid anabolism (lipogenesis and lipid desaturation) was simultaneously up-regulated prior to hibernation onset in the animals. The enhanced capacity of both lipid catabolism and lipid anabolism during hibernation period (HIB) is striking contrast to previous observations in fat-storing hibernators that only enhance catabolism during HIB. The mRNA expression of mTORC1 and PPAR signaling molecules increased, and pharmacological activation of PPARs indeed up-regulated lipid metabolism genes in iWAT explants from Syrian hamsters. These results suggest that the Syrian hamster rewires lipid metabolisms while preparing for hibernation to effectively utilize body fat and synthesize it from food intake during HIB.
Collapse
Affiliation(s)
- Yuichi Chayama
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Lisa Ando
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuya Sato
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shuji Shigenobu
- Functional Genomics Facility, National Institute for Basic Biology, Okazaki, Japan
| | - Daisuke Anegawa
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takayuki Fujimoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroki Taii
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yutaka Tamura
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshifumi Yamaguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Hibernation Metabolism, Physiology and Development Group, Institute of Low Temperature Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
16
|
Oliver SR, Anderson KJ, Hunstiger MM, Andrews MT. Turning down the heat: Down-regulation of sarcolipin in a hibernating mammal. Neurosci Lett 2018; 696:13-19. [PMID: 30528880 DOI: 10.1016/j.neulet.2018.11.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 02/01/2023]
Abstract
Hibernation in mammals is a whole-body phenotype that involves profound reductions in oxygen consumption, metabolic reactions, core body temperature, neural activity and heart rate. An important aspect of mammalian hibernation is the ability to reverse this state of hypothermic torpor by rewarming and subsequent arousal. Brown adipose tissue (BAT) and skeletal muscle shivering have been characterized as the predominant driving forces for thermogenesis during arousal. Conversely, the thermogenic contribution of these organs needs to be minimized as hibernating mammals enter torpor. Because skeletal muscle accounts for approximately 40% of the dry mass of the typical mammalian body, we aim to broaden the spotlight to include the importance of down-regulating skeletal muscle non-shivering thermogenesis during hibernation to allow for whole-body cooling and long-term maintenance of a depressed core body temperature when the animal is in torpor. This minireview will briefly describe the current understanding of thermoregulation in hibernating mammals and present new preliminary data on the importance of skeletal muscle and the micro-peptide sarcolipin as a major thermogenic target.
Collapse
Affiliation(s)
- S Ryan Oliver
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, AK 99775, USA.
| | - Kyle J Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN 55812, USA.
| | - Moriah M Hunstiger
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, AK 99775, USA.
| | - Matthew T Andrews
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
17
|
Shimaoka H, Kawaguchi T, Morikawa K, Sano Y, Naitou K, Nakamori H, Shiina T, Shimizu Y. Induction of hibernation-like hypothermia by central activation of the A1 adenosine receptor in a non-hibernator, the rat. J Physiol Sci 2018; 68:425-430. [PMID: 28508339 PMCID: PMC10717028 DOI: 10.1007/s12576-017-0543-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 05/10/2017] [Indexed: 11/30/2022]
Abstract
Central adenosine A1-receptor (A1AR)-mediated signals play a role in the induction of hibernation. We determined whether activation of the central A1AR enables rats to maintain normal sinus rhythm even after their body temperature has decreased to less than 20 °C. Intracerebroventricular injection of an adenosine A1 agonist, N6-cyclohexyladenosine (CHA), followed by cooling decreased the body temperature of rats to less than 20 °C. Normal sinus rhythm was fundamentally maintained during the extreme hypothermia. In contrast, forced induction of hypothermia by cooling anesthetized rats caused cardiac arrest. Additional administration of pentobarbital to rats in which hypothermia was induced by CHA also caused cardiac arrest, suggesting that the operation of some beneficial mechanisms that are not activated under anesthesia may be essential to keep heart beat under the hypothermia. These results suggest that central A1AR-mediated signals in the absence of anesthetics would provide an appropriate condition for maintaining normal sinus rhythm during extreme hypothermia.
Collapse
Affiliation(s)
- Hiroki Shimaoka
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Takayuki Kawaguchi
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kahori Morikawa
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yuuki Sano
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kiyotada Naitou
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Hiroyuki Nakamori
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Takahiko Shiina
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yasutake Shimizu
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
- Laboratory of Veterinary Physiology, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
| |
Collapse
|
18
|
Transcriptomic Analysis of Ciguatoxin-Induced Changes in Gene Expression in Primary Cultures of Mice Cortical Neurons. Toxins (Basel) 2018; 10:toxins10050192. [PMID: 29748486 PMCID: PMC5983248 DOI: 10.3390/toxins10050192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/02/2018] [Accepted: 05/07/2018] [Indexed: 11/17/2022] Open
Abstract
Ciguatoxins are polyether marine toxins that act as sodium channel activators. These toxins cause ciguatera, one of the most widespread nonbacterial forms of food poisoning, which presents several symptoms in humans including long-term neurological alterations. Earlier work has shown that both acute and chronic exposure of primary cortical neurons to synthetic ciguatoxin CTX3C have profound impacts on neuronal function. Thus, the present work aimed to identify relevant neuronal genes and metabolic pathways that could be altered by ciguatoxin exposure. To study the effect of ciguatoxins in primary neurons in culture, we performed a transcriptomic analysis using whole mouse genome microarrays, for primary cortical neurons exposed during 6, 24, or 72 h in culture to CTX3C. Here, we have shown that the effects of the toxin on gene expression differ with the exposure time. The results presented here have identified several relevant genes and pathways related to the effect of ciguatoxins on neurons and may assist in future research or even treatment of ciguatera. Moreover, we demonstrated that the effects of the toxin on gene expression were exclusively consequential of its action as a voltage-gated sodium channel activator, since all the effects of CTX3C were avoided by preincubation of the neurons with the sodium channel blocker tetrodotoxin.
Collapse
|
19
|
Horii Y, Shiina T, Shimizu Y. The Mechanism Enabling Hibernation in Mammals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1081:45-60. [PMID: 30288703 DOI: 10.1007/978-981-13-1244-1_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Some rodents including squirrels and hamsters undergo hibernation. During hibernation, body temperature drops to only a few degrees above ambient temperature. The suppression of whole-body energy expenditure is associated with regulated, but not passive, reduction of cellular metabolism. The heart retains the ability to beat constantly, although body temperature drops to less than 10 °C during hibernation. Cardiac myocytes of hibernating mammals are characterized by reduced Ca2+ entry into the cell membrane and a concomitant enhancement of Ca2+ release from and reuptake by the sarcoplasmic reticulum. These adaptive changes would help in preventing excessive Ca2+ entry and its overload and in maintaining the resting levels of intracellular Ca2+. Adaptive changes in gene expression in the heart prior to hibernation may be indispensable for acquiring cold resistance. In addition, protective effects of cold-shock proteins are thought to have an important role. We recently reported the unique expression pattern of cold-inducible RNA-binding protein (CIRP) in the hearts of hibernating hamsters. The CIRP mRNA is constitutively expressed in the heart of a nonhibernating euthermic hamster with several different forms probably due to alternative splicing. The short product contained the complete open reading frame for full-length CIRP, while the long product had inserted sequences containing a stop codon, suggesting production of a C-terminal deletion isoform of CIRP. In contrast to nonhibernating hamsters, only the short product was found in hibernating animals. Thus, these results indicate that CIRP expression in the hamster heart is regulated at the level of alternative splicing, which would permit a rapid increment of functional CIRP when entering hibernation. We will summarize the current understanding of the cold-resistant property of the heart in hibernating animals.
Collapse
Affiliation(s)
- Yuuki Horii
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Takahiko Shiina
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Yasutake Shimizu
- Department of Basic Veterinary Science, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan.
| |
Collapse
|
20
|
Swoap SJ. Central adenosine and daily torpor in mice. Temperature (Austin) 2017; 4:350-352. [DOI: 10.1080/23328940.2017.1345713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Steven J. Swoap
- Department of Biology, Williams College, 59 Lab Campus Drive, Williamstown, MA 01267, USA
| |
Collapse
|
21
|
Central activation of the A 1 adenosine receptor in fed mice recapitulates only some of the attributes of daily torpor. J Comp Physiol B 2017; 187:835-845. [PMID: 28378088 DOI: 10.1007/s00360-017-1084-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/02/2016] [Accepted: 03/07/2017] [Indexed: 01/23/2023]
Abstract
Mice enter bouts of daily torpor, drastically reducing metabolic rate, core body temperature (T b), and heart rate (HR), in response to reduced caloric intake. Because central adenosine activation has been shown to induce a torpor-like state in the arctic ground squirrel, and blocking the adenosine-1 (A1) receptor prevents daily torpor, we hypothesized that central activation of the A1 adenosine receptors would induce a bout of natural torpor in mice. To test the hypothesis, mice were subjected to four different hypothermia bouts: natural torpor, forced hypothermia (FH), isoflurane-anesthesia, and an intracerebroventricular injection of the selective A1 receptor agonist N6-cyclohexyladenosine (CHA). All conditions induced profound hypothermia. T b fell more rapidly in the FH, isoflurane-anesthesia, and CHA conditions compared to torpor, while mice treated with CHA recovered at half the rate of torpid mice. FH, isoflurane-anesthesia, and CHA-treated mice exhibited a diminished drop in HR during entry into hypothermia as compared to torpor. Mice in all conditions except CHA shivered while recovering from hypothermia, and only FH mice shivered substantially while entering hypothermia. Circulating lactate during the hypothermic bouts was not significantly different between the CHA and torpor conditions, both of which had lower than baseline lactate levels. Arrhythmias were largely absent in the FH and isoflurane-anesthesia conditions, while skipped beats were observed in natural torpor and periodic extended (>1 s) HR pauses in the CHA condition. Lastly, the hypothermic bouts showed distinct patterns of gene expression, with torpor characterized by elevated hepatic and cardiac Txnip expression and all other hypothermic states characterized by elevated c-Fos and Egr-1 expression. We conclude that CHA-induced hypothermia and natural torpor are largely different physiological states.
Collapse
|
22
|
Tupone D, Cano G, Morrison SF. Thermoregulatory inversion: a novel thermoregulatory paradigm. Am J Physiol Regul Integr Comp Physiol 2017; 312:R779-R786. [PMID: 28330964 DOI: 10.1152/ajpregu.00022.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/01/2017] [Accepted: 03/15/2017] [Indexed: 01/06/2023]
Abstract
To maintain core body temperature in mammals, the normal central nervous system (CNS) thermoregulatory reflex networks produce an increase in brown adipose tissue (BAT) thermogenesis in response to skin cooling and an inhibition of the sympathetic outflow to BAT during skin rewarming. In contrast, these normal thermoregulatory reflexes appear to be inverted in hibernation/torpor; thermogenesis is inhibited during exposure to a cold environment, allowing dramatic reductions in core temperature and metabolism, and thermogenesis is activated during skin rewarming, contributing to a return of normal body temperature. Here, we describe two unrelated experimental paradigms in which rats, a nonhibernating/torpid species, exhibit a "thermoregulatory inversion," which is characterized by an inhibition of BAT thermogenesis in response to skin cooling, and a switch in the gain of the skin cooling reflex transfer function from negative to positive values. Either transection of the neuraxis immediately rostral to the dorsomedial hypothalamus in anesthetized rats or activation of A1 adenosine receptors within the CNS of free-behaving rats produces a state of thermoregulatory inversion in which skin cooling inhibits BAT thermogenesis, leading to hypothermia, and skin warming activates BAT, supporting an increase in core temperature. These results reflect the existence of a novel neural circuit that mediates inverted thermoregulatory reflexes and suggests a pharmacological mechanism through which a deeply hypothermic state can be achieved in nonhibernating/torpid mammals, possibly including humans.
Collapse
Affiliation(s)
- Domenico Tupone
- Department of Neurological Surgery, Oregon Health and Science University, Portland, Oregon; and
| | - Georgina Cano
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shaun F Morrison
- Department of Neurological Surgery, Oregon Health and Science University, Portland, Oregon; and
| |
Collapse
|
23
|
Trefna M, Goris M, Thissen CMC, Reitsema VA, Bruintjes JJ, de Vrij EL, Bouma HR, Boerema AS, Henning RH. The influence of sex and diet on the characteristics of hibernation in Syrian hamsters. J Comp Physiol B 2017; 187:725-734. [PMID: 28324158 PMCID: PMC5486544 DOI: 10.1007/s00360-017-1072-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/24/2016] [Accepted: 02/26/2017] [Indexed: 02/07/2023]
Abstract
Research on deep hibernators almost exclusively uses species captured from the wild or from local breeding. An exception is Syrian hamster (Mesocricetus auratus), the only standard laboratory animal showing deep hibernation. In deep hibernators, several factors influence hibernation quality, including body mass, sex and diet. We examined hibernation quality in commercially obtained Syrian hamsters in relation to body mass, sex and a diet enriched in polyunsaturated fatty acids. Animals (M/F:30/30, 12 weeks of age) were obtained from Harlan (IN, USA) and individually housed at 21 °C and L:D 14:10 until 20 weeks of age, followed by L:D 8:16 until 27 weeks. Then conditions were changed to 5 °C and L:D 0:24 for 9 weeks to induce hibernation. Movement was continuously monitored with passive infrared detectors. Hamsters were randomized to control diet or a diet 3× enriched in linoleic acid from 16 weeks of age. Hamsters showed a high rate of premature death (n = 24, 40%), both in animals that did and did not initiate torpor, which was unrelated to body weight, sex and diet. Time to death (31.7 ± 3.1 days, n = 12) or time to first torpor bout (36.6 ± 1.6 days, n = 12) was similar in prematurely deceased hamsters. Timing of induction of hibernation and duration of torpor and arousal was unaffected by body weight, sex or diet. Thus, commercially obtained Syrian hamsters subjected to winter conditions showed poor survival, irrespective of body weight, sex and diet. These factors also did not affect hibernation parameters. Possibly, long-term commercial breeding from a confined genetic background has selected against the hibernation trait.
Collapse
Affiliation(s)
- Marie Trefna
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Maaike Goris
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Cynthia M C Thissen
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Vera A Reitsema
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Jojanneke J Bruintjes
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Edwin L de Vrij
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Hjalmar R Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Ate S Boerema
- Departments of Chronobiology and Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
24
|
Carlin JL, Jain S, Gizewski E, Wan TC, Tosh DK, Xiao C, Auchampach JA, Jacobson KA, Gavrilova O, Reitman ML. Hypothermia in mouse is caused by adenosine A 1 and A 3 receptor agonists and AMP via three distinct mechanisms. Neuropharmacology 2017; 114:101-113. [PMID: 27914963 PMCID: PMC5183552 DOI: 10.1016/j.neuropharm.2016.11.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/02/2016] [Accepted: 11/28/2016] [Indexed: 10/20/2022]
Abstract
Small mammals have the ability to enter torpor, a hypothermic, hypometabolic state, allowing impressive energy conservation. Administration of adenosine or adenosine 5'-monophosphate (AMP) can trigger a hypothermic, torpor-like state. We investigated the mechanisms for hypothermia using telemetric monitoring of body temperature in wild type and receptor knock out (Adora1-/-, Adora3-/-) mice. Confirming prior data, stimulation of the A3 adenosine receptor (AR) induced hypothermia via peripheral mast cell degranulation, histamine release, and activation of central histamine H1 receptors. In contrast, A1AR agonists and AMP both acted centrally to cause hypothermia. Commonly used, selective A1AR agonists, including N6-cyclopentyladenosine (CPA), N6-cyclohexyladenosine (CHA), and MRS5474, caused hypothermia via both A1AR and A3AR when given intraperitoneally. Intracerebroventricular dosing, low peripheral doses of Cl-ENBA [(±)-5'-chloro-5'-deoxy-N6-endo-norbornyladenosine], or using Adora3-/- mice allowed selective stimulation of A1AR. AMP-stimulated hypothermia can occur independently of A1AR, A3AR, and mast cells. A1AR and A3AR agonists and AMP cause regulated hypothermia that was characterized by a drop in total energy expenditure, physical inactivity, and preference for cooler environmental temperatures, indicating a reduced body temperature set point. Neither A1AR nor A3AR was required for fasting-induced torpor. A1AR and A3AR agonists and AMP trigger regulated hypothermia via three distinct mechanisms.
Collapse
Affiliation(s)
- Jesse Lea Carlin
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Shalini Jain
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Elizabeth Gizewski
- Department of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Tina C Wan
- Department of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, 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
| | - John A Auchampach
- Department of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Drew KL, Frare C, Rice SA. Neural Signaling Metabolites May Modulate Energy Use in Hibernation. Neurochem Res 2017; 42:141-150. [PMID: 27878659 PMCID: PMC5284051 DOI: 10.1007/s11064-016-2109-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/05/2016] [Accepted: 11/11/2016] [Indexed: 12/23/2022]
Abstract
Despite an epidemic in obesity and metabolic syndrome limited means exist to effect adiposity or metabolic rate other than life style changes. Here we review evidence that neural signaling metabolites may modulate thermoregulatory pathways and offer novel means to fine tune energy use. We extend prior reviews on mechanisms that regulate thermogenesis and energy use in hibernation by focusing primarily on the neural signaling metabolites adenosine, AMP and glutamate.
Collapse
Affiliation(s)
- Kelly L Drew
- Department of Chemistry and Biochemistry, Institute of Arctic Biology, University of Alaska Fairbanks, 902 N. Koyukuk Drive, Fairbanks, AK, 99775, USA.
| | - Carla Frare
- Department of Chemistry and Biochemistry, Institute of Arctic Biology, University of Alaska Fairbanks, 902 N. Koyukuk Drive, Fairbanks, AK, 99775, USA
| | - Sarah A Rice
- Department of Chemistry and Biochemistry, Institute of Arctic Biology, University of Alaska Fairbanks, 902 N. Koyukuk Drive, Fairbanks, AK, 99775, USA
| |
Collapse
|
26
|
Sunagawa GA, Takahashi M. Hypometabolism during Daily Torpor in Mice is Dominated by Reduction in the Sensitivity of the Thermoregulatory System. Sci Rep 2016; 6:37011. [PMID: 27845399 PMCID: PMC5109469 DOI: 10.1038/srep37011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/21/2016] [Indexed: 01/20/2023] Open
Abstract
Some mammals enter a hypometabolic state either daily torpor (minutes to hours in length) or hibernation (days to weeks), when reducing metabolism would benefit survival. Hibernators demonstrate deep torpor by reducing both the sensitivity (H) and the theoretical set-point temperature (TR) of the thermogenesis system, resulting in extreme hypothermia close to ambient temperature. However, these properties during daily torpor remain poorly understood due to the very short steady state of the hypometabolism and the large variation among species and individuals. To overcome these difficulties in observing and evaluating daily torpor, we developed a novel torpor-detection algorithm based on Bayesian estimation of the basal metabolism of individual mice. Applying this robust method, we evaluated fasting induced torpor in various ambient temperatures (TAs) and found that H decreased 91.5% during daily torpor while TR only decreased 3.79 °C in mice. These results indicate that thermogenesis during daily torpor shares a common property of sensitivity reduction with hibernation while it is distinct from hibernation by not lowering TR. Moreover, our findings support that mice are suitable model animals to investigate the regulation of the heat production during active hypometabolism, thus suggesting further study of mice may provide clues to regulating hypometabolism in mammals.
Collapse
Affiliation(s)
- Genshiro A Sunagawa
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojimaminami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
27
|
Chayama Y, Ando L, Tamura Y, Miura M, Yamaguchi Y. Decreases in body temperature and body mass constitute pre-hibernation remodelling in the Syrian golden hamster, a facultative mammalian hibernator. ROYAL SOCIETY OPEN SCIENCE 2016; 3:160002. [PMID: 27152216 PMCID: PMC4852639 DOI: 10.1098/rsos.160002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 03/08/2016] [Indexed: 06/02/2023]
Abstract
Hibernation is an adaptive strategy for surviving during periods with little or no food availability, by profoundly reducing the metabolic rate and the core body temperature (T b). Obligate hibernators (e.g. bears, ground squirrels, etc.) hibernate every winter under the strict regulation of endogenous circannual rhythms, and they are assumed to undergo adaptive remodelling in autumn, the pre-hibernation period, prior to hibernation. However, little is known about the nature of pre-hibernation remodelling. Syrian hamsters (Mesocricetus auratus) are facultative hibernators that can hibernate irrespective of seasons when exposed to prolonged short photoperiod and cold ambient temperature (SD-Cold) conditions. Their T b set point reduced by the first deep torpor (DT) and then increased gradually after repeated cycles of DT and periodic arousal (PA), and finally recovered to the level observed before the prolonged SD-Cold in the post-hibernation period. We also found that, before the initiation of hibernation, the body mass of animals decreased below a threshold, indicating that hibernation in this species depends on body condition. These observations suggest that Syrian hamsters undergo pre-hibernation remodelling and that T b and body mass can be useful physiological markers to monitor the remodelling process during the pre-hibernation period.
Collapse
Affiliation(s)
- Yuichi Chayama
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Lisa Ando
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yutaka Tamura
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama 729-0292, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Yoshifumi Yamaguchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
28
|
Lossi L, D’Angelo L, De Girolamo P, Merighi A. Anatomical features for an adequate choice of experimental animal model in biomedicine: II. Small laboratory rodents, rabbit, and pig. Ann Anat 2016; 204:11-28. [DOI: 10.1016/j.aanat.2015.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 01/24/2023]
|
29
|
Jinka TR, Combs VM, Drew KL. Translating drug-induced hibernation to therapeutic hypothermia. ACS Chem Neurosci 2015; 6:899-904. [PMID: 25812681 DOI: 10.1021/acschemneuro.5b00056] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Therapeutic hypothermia (TH) improves prognosis after cardiac arrest; however, thermoregulatory responses such as shivering complicate cooling. Hibernators exhibit a profound and safe reversible hypothermia without any cardiovascular side effects by lowering the shivering threshold at low ambient temperatures (Ta). Activation of adenosine A1 receptors (A1ARs) in the central nervous system (CNS) induces hibernation in hibernating species and a hibernation-like state in rats, principally by attenuating thermogenesis. Thus, we tested the hypothesis that targeted activation of the central A1AR combined with a lower Ta would provide a means of managing core body temperature (Tb) below 37 °C for therapeutic purposes. We targeted the A1AR within the CNS by combining systemic delivery of the A1AR agonist (6)N-cyclohexyladenosine (CHA) with 8-(p-sulfophenyl)theophylline (8-SPT), a nonspecific adenosine receptor antagonist that does not readily cross the blood-brain barrier. Results show that CHA (1 mg/kg) and 8-SPT (25 mg/kg), administered intraperitoneally every 4 h for 20 h at a Ta of 16 °C, induce and maintain the Tb between 29 and 31 °C for 24 h in both naïve rats and rats subjected to asphyxial cardiac arrest for 8 min. Faster and more stable hypothermia was achieved by continuous infusion of CHA delivered subcutaneously via minipumps. Animals subjected to cardiac arrest and cooled by CHA survived better and showed less neuronal cell death than normothermic control animals. Central A1AR activation in combination with a thermal gradient shows promise as a novel and effective pharmacological adjunct for inducing safe and reversible targeted temperature management.
Collapse
Affiliation(s)
- Tulasi R. Jinka
- University of Alaska Fairbanks, 902 North Koyukuk Drive, Fairbanks, Alaska 99775-7000, United States
- University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Velva M. Combs
- University of Alaska Fairbanks, 902 North Koyukuk Drive, Fairbanks, Alaska 99775-7000, United States
| | - Kelly L. Drew
- University of Alaska Fairbanks, 902 North Koyukuk Drive, Fairbanks, Alaska 99775-7000, United States
| |
Collapse
|
30
|
Drew KL, Romanovsky AA, Stephen TKL, Tupone D, Williams RH. Future approaches to therapeutic hypothermia: a symposium report. Temperature (Austin) 2015; 2:168-71. [PMID: 27227020 PMCID: PMC4843898 DOI: 10.4161/23328940.2014.976512] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 10/09/2014] [Accepted: 10/09/2014] [Indexed: 12/22/2022] Open
Key Words
- A1AR
- AGS, arctic ground squirrel
- CE, capillary electrophoresis
- EEG,electroencephalogram
- FSCV, Fast scan cyclic voltammetry
- HPLC, high performance liquid chromatography
- ICV, intracerebroventricular
- TRPM8
- Tb, core body temperature
- adenosine
- capillary electrophoresis
- hibernation
- nNOS, neuronal nitric oxide synthase; NTS, nucleus tractus solitarii; TH, therapeutic hypothermia; TRP, transient receptor potential [channel(s)]; TRPM8, TRP melastatin-8
- nNOS/NK1
- targeted temperature management
- therapeutic hypothermia
- torpor
Collapse
Affiliation(s)
- Kelly L Drew
- Institute of Arctic Biology; University of Alaska Fairbanks; Fairbanks, AK, USA
| | - Andrej A Romanovsky
- Systemic Inflammation Laboratory (Fever Lab); Trauma Research, St. Joseph's Hospital and Medical Center; Phoenix, AZ, USA
| | - Terilyn KL Stephen
- Department of Chemistry and Biochemistry; University of Alaska Fairbanks; Fairbanks, AK, USA
| | - Domenico Tupone
- Department of Neurological Surgery; Oregon Health & Science University; Portland, OR, USA
| | | |
Collapse
|
31
|
Shylo AV. Dynamics of the Electrographic Indices in Rats and Hamsters Recovering from Artificial and Natural Hypometabolic States. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9502-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
32
|
Xia T, Zhang Q, Xiao Y, Wang C, Yu J, Liu H, Liu B, Zhang Y, Chen S, Liu Y, Chen Y, Guo F. CREB/TRH pathway in the central nervous system regulates energy expenditure in response to deprivation of an essential amino acid. Int J Obes (Lond) 2014; 39:105-13. [PMID: 24732144 DOI: 10.1038/ijo.2014.65] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/18/2014] [Accepted: 04/06/2014] [Indexed: 01/17/2023]
Abstract
BACKGROUND In the central nervous system (CNS), thyrotropin-releasing hormone (TRH) has an important role in regulating energy balance. We previously showed that dietary deprivation of leucine in mice increases energy expenditure through CNS-dependent regulation. However, the involvement of central TRH in this regulation has not been reported. METHODS Male C57J/B6 mice were maintained on a control or leucine-deficient diet for 7 days. Leucine-deprived mice were either third intracerebroventricular (i.c.v.) injected with a TRH antibody followed by intraperitoneal (i.p.) injection of triiodothyronine (T3) or i.c.v. administrated with an adenovirus of shCREB (cAMP-response element binding protein) followed by i.c.v. injection of TRH. Food intake and body weight were monitored daily. Oxygen consumption, physical activity and rectal temperature were assessed after the treatment. After being killed, the hypothalamus and the brown adipose tissue were collected and the expression of related genes and proteins related was analyzed. In other experiments, control or leucine-deficient medium incubated primary cultured neurons were either infected with adenovirus-mediated short hairpin RNA targeting extracellular signal-regulated kinases 1 and 2 (Ad-shERK1/2) or transfected with plasmid-overexpressing protein phosphatase 1 regulatory subunit 3C (PPP1R3C). RESULTS I.c.v. administration of anti-TRH antibodies significantly reduced leucine deprivation-stimulated energy expenditure. Furthermore, the effects of i.c.v. TRH antibodies were reversed by i.p. injection of T3 during leucine deprivation. Moreover, i.c.v. injection of Ad-shCREB (adenovirus-mediated short hairpin RNA targeting CREB) significantly suppressed leucine deprivation-stimulated energy expenditure via modulation of TRH expression. Lastly, TRH expression was regulated by CREB, which was phosphorylated by ERK1/2 and dephosphorylated by PPP1R3C-containing protein Ser/Thr phosphatase type 1 (PP1) under leucine deprivation in vitro. CONCLUSIONS Our data indicate a novel role for TRH in regulating energy expenditure via T3 during leucine deprivation. Furthermore, our findings reveal that TRH expression is activated by CREB, which is phosphorylated by ERK1/2 and dephosphorylated by PPP1R3C-containing PP1. Collectively, our studies provide novel insights into the regulation of energy homeostasis by the CNS in response to an essential amino-acid deprivation.
Collapse
Affiliation(s)
- T Xia
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Q Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Y Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - C Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - J Yu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - H Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - B Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Y Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - S Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Y Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Y Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - F Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, The Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
33
|
Olson JM, Jinka TR, Larson LK, Danielson JJ, Moore JT, Carpluck J, Drew KL. Circannual rhythm in body temperature, torpor, and sensitivity to A₁ adenosine receptor agonist in arctic ground squirrels. J Biol Rhythms 2013; 28:201-7. [PMID: 23735499 DOI: 10.1177/0748730413490667] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A₁ adenosine receptor (A₁AR) activation within the central nervous system induces torpor, but in obligate hibernators such as the arctic ground squirrel (AGS; Urocitellus parryii), A₁AR stimulation induces torpor only during the hibernation season, suggesting a seasonal increase in sensitivity to A₁AR signaling. The purpose of this research was to investigate the relationship between body temperature (Tb) and sensitivity to an adenosine A1 receptor agonist in AGS. We tested the hypothesis that increased sensitivity in A₁AR signaling would lead to lower Tb in euthermic animals during the hibernation season when compared with the summer season. We further predicted that if a decrease in euthermic Tb reflects increased sensitivity to A₁AR activation, then it should likewise predict spontaneous torpor. We used subcutaneous IPTT-300 transponders to monitor Tb in AGS housed under constant ambient conditions (12:12 L:D, 18 °C) for up to 16 months. These animals displayed an obvious rhythm in euthermic Tb that cycled with a period of approximately 8 months. Synchrony in the Tb rhythm within the group was lost after several months of constant L:D conditions; however, individual rhythms in Tb continued to show clear sine wave-like waxing and waning. AGS displayed spontaneous torpor only during troughs in euthermic Tb. To assess sensitivity to A₁AR activation, AGS were administered the A₁AR agonist N(6)-cyclohexyladenosine (CHA, 0.1 mg/kg, ip), and subcutaneous Tb was monitored. AGS administered CHA during a seasonal minimum in euthermic Tb showed a greater drug-induced decrease in Tb (1.6 ± 0.3 °C) than did AGS administered CHA during a peak in euthermic Tb (0.4 ± 0.3 °C). These results provide evidence for a circannual rhythm in Tb that is associated with increased sensitivity to A₁AR signaling and correlates with the onset of torpor.
Collapse
Affiliation(s)
- Jasmine M Olson
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK 99775-7000, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Nathaniel TI, Otukonyong EE, Okon M, Chaves J, Cochran T, Nathaniel AI. Metabolic regulatory clues from the naked mole rat: toward brain regulatory functions during stroke. Brain Res Bull 2013; 98:44-52. [PMID: 23886571 DOI: 10.1016/j.brainresbull.2013.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/03/2013] [Accepted: 07/15/2013] [Indexed: 12/30/2022]
Abstract
Resistance to tissue hypoxia is a robust fundamental adaptation to low oxygen supply, and represents a novel neuroscience problem with significance to mammalian physiology as well as human health. With the underlying mechanisms strongly conserved in evolution, the ability to resist tissue hypoxia in natural systems has recently emerged as an interesting model in mammalian physiology research to understand mechanisms that can be manipulated for the clinical management of stroke. The extraordinary ability to resist tissue hypoxia by the naked mole rat (NMR) indicates the presence of a unique mechanism that underlies the remarkable healthy life span and exceptional hypoxia resistance. This opens an interesting line of research into understanding the mechanisms employed by the naked mole rat (Heterocephalus glaber) to protect the brain during hypoxia. In a series of studies, we first examined the presence of neuroprotection in the brain cells of naked mole rats (NMRs) subjected to hypoxic insults, and then characterized the expression of such neuroprotection in a wide range of time intervals. We used oxygen nutrient deprivation (OND), an in vitro model of resistance to tissue hypoxia to determine whether there is evidence of neuronal survival in the hippocampal (CA1) slices of NMRs that are subjected to chronic hypoxia. Hippocampus neurons of NMRs that were kept in hypoxic condition consistently tolerated OND right from the onset time of 5h. This tolerance was maintained for 24h. This finding indicates that there is evidence of resistance to tissue hypoxia by CA1 neurons of NMRs. We further examined the effect of hypoxia on metabolic rate in the NMR. Repeated measurement of metabolic rates during exposure of naked mole rats to hypoxia over a constant ambient temperature indicates that hypoxia significantly decreased metabolic rates in the NMR, suggesting that the observed decline in metabolic rate during hypoxia may contribute to the adaptive mechanism used by the NMR to resist tissue hypoxia. This work is aimed to contribute to the understanding of mechanisms of resistance to tissue hypoxia in the NMR as an important life-sustaining process, which can be translated into therapeutic interventions during stroke.
Collapse
Affiliation(s)
- Thomas I Nathaniel
- University of South Carolina School of Medicine, HSEB, 607 Grove Road, Greenville, SC 29605, United States.
| | | | | | | | | | | |
Collapse
|
36
|
Effect of Opioids on Tissue Metabolism in Aestivating and Active Green-Striped Burrowing Frogs,Cyclorana alboguttata. J HERPETOL 2013. [DOI: 10.1670/12-039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Iliff BW, Swoap SJ. Central adenosine receptor signaling is necessary for daily torpor in mice. Am J Physiol Regul Integr Comp Physiol 2012; 303:R477-84. [PMID: 22785425 DOI: 10.1152/ajpregu.00081.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
When calorically restricted at cool ambient temperatures, mice conserve energy by entering torpor, during which metabolic rate (MR), body temperature (T(b)), heart rate (HR), and locomotor activity (LMA) decrease. Treatment with exogenous adenosine produces a similar hypometabolic state. In this study, we conducted a series of experiments using the nonspecific adenosine receptor antagonists aminophylline and 8-sulfophenyltheophylline (8-SPT) to test the hypothesis that adenosine signaling is necessary for torpor in fasted mice. In the first experiment, mice were subcutaneously infused with aminophylline while T(b), HR, and LMA were continuously monitored using implanted radiotelemeters. During a 23-h fast, saline-treated mice were torpid for 518 ± 43 min, whereas aminophylline-treated mice were torpid for significantly less time (54 ± 20 min). In a second experiment, aminophylline was infused subcutaneously into torpid mice to test the role of adenosine in the maintenance of torpor. Aminophylline reversed the hypometabolism, hypothermia, bradycardia, and hypoactivity of torpor, whereas saline did not. In the third and fourth experiments, the polar adenosine antagonist 8-SPT, which does not cross the blood-brain barrier, was infused either subcutaneously or intracerebroventricularly to test the hypothesis that both peripheral and central adenosine receptor signaling are necessary for the maintenance of torpor. Intracerebroventricular, but not subcutaneous, infusion of 8-SPT causes a return to euthermia. These findings support the hypothesis that adenosine is necessary for torpor in mice and further suggest that whereas peripheral adenosine signaling is not necessary for the maintenance of torpor, antagonism of central adenosine is sufficient to disrupt torpor.
Collapse
Affiliation(s)
- Benjamin W Iliff
- Department of Biology, Williams College, Williamstown, MA 01267, USA
| | | |
Collapse
|
38
|
Neuroprotection supports signal processing in the hippocampus of Syrian hamsters, a facultative hibernator. Neurosci Lett 2012; 520:20-5. [DOI: 10.1016/j.neulet.2012.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/10/2012] [Accepted: 05/02/2012] [Indexed: 11/24/2022]
|
39
|
Tamura Y, Shintani M, Inoue H, Monden M, Shiomi H. Regulatory mechanism of body temperature in the central nervous system during the maintenance phase of hibernation in Syrian hamsters: Involvement of β-endorphin. Brain Res 2012; 1448:63-70. [DOI: 10.1016/j.brainres.2012.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 01/19/2012] [Accepted: 02/02/2012] [Indexed: 01/17/2023]
|
40
|
Liu S, Chen JF. Strategies for therapeutic hypometabothermia. JOURNAL OF EXPERIMENTAL STROKE & TRANSLATIONAL MEDICINE 2012; 5:31-42. [PMID: 24179563 PMCID: PMC3811165 DOI: 10.6030/1939-067x-5.1.31] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Although therapeutic hypothermia and metabolic suppression have shown robust neuroprotection in experimental brain ischemia, systemic complications have limited their use in treating acute stroke patients. The core temperature and basic metabolic rate are tightly regulated and maintained in a very stable level in mammals. Simply lowering body temperature or metabolic rate is actually a brutal therapy that may cause more systemic as well as regional problems other than providing protection. These problems are commonly seen in hypothermia and barbiturate coma. The main innovative concept of this review is to propose thermogenically optimal and synergistic reduction of core temperature and metabolic rate in therapeutic hypometabothermia using novel and clinically practical approaches. When metabolism and body temperature are reduced in a systematically synergistic manner, the outcome will be maximal protection and safe recovery, which happen in natural process, such as in hibernation, daily torpor and estivation.
Collapse
Affiliation(s)
- Shimin Liu
- Department of Neurology, Boston University School of Medicine, Boston, USA
| | - Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, USA
| |
Collapse
|
41
|
Season primes the brain in an arctic hibernator to facilitate entrance into torpor mediated by adenosine A(1) receptors. J Neurosci 2011; 31:10752-8. [PMID: 21795527 DOI: 10.1523/jneurosci.1240-11.2011] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Torpor in hibernating mammals defines the nadir in mammalian metabolic demand and body temperature that accommodates seasonal periods of reduced energy availability. The mechanism of metabolic suppression during torpor onset is unknown, although the CNS is a key regulator of torpor. Seasonal hibernators, such as the arctic ground squirrel (AGS), display torpor only during the winter, hibernation season. The seasonal character of hibernation thus provides a clue to its regulation. In the present study, we delivered adenosine receptor agonists and antagonists into the lateral ventricle of AGSs at different times of the year while monitoring the rate of O(2) consumption and core body temperature as indicators of torpor. The A(1) antagonist cyclopentyltheophylline reversed spontaneous entrance into torpor. The adenosine A(1) receptor agonist N(6)-cyclohexyladenosine (CHA) induced torpor in six of six AGSs tested during the mid-hibernation season, two of six AGSs tested early in the hibernation season, and none of the six AGSs tested during the summer, off-season. CHA-induced torpor within the hibernation season was specific to A(1)AR activation; the A(3)AR agonist 2-Cl-IB MECA failed to induce torpor, and the A(2a)R antagonist MSX-3 failed to reverse spontaneous onset of torpor. CHA-induced torpor was similar to spontaneous entrance into torpor. These results show that metabolic suppression during torpor onset is regulated within the CNS via A(1)AR activation and requires a seasonal switch in the sensitivity of purinergic signaling.
Collapse
|
42
|
Darbera L, Chenoune M, Lidouren F, Ghaleh B, Cohen MV, Downey JM, Berdeaux A, Tissier R. Adenosine and Opioid Receptors Do Not Trigger the Cardioprotective Effect of Mild Hypothermia. J Cardiovasc Pharmacol Ther 2011; 17:173-80. [DOI: 10.1177/1074248411412969] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Mild hypothermia (32°C-34°C) exerts a potent cardioprotection in animal models of myocardial infarction. Recently, it has been proposed that this beneficial effect is related to survival signaling. We, therefore, hypothesized that the well-known cardioprotective pathways dependent on adenosine and/or opioid receptors could be the trigger of hypothermia-induced salvage. Open-chest rabbits were accordingly exposed to 30 minutes of coronary artery occlusion (CAO) under normothermic (NT) or hypothermic ([HT] 32°C) conditions. In the latter, hypothermia was induced by total liquid ventilation with temperature-controlled perfluorocarbons in order to effect ultrafast cooling and to accurately control cardiac temperature. After 4 hours of reperfusion, infarct and no-reflow zone sizes were assessed and quantified as a percentage of the risk zone. In animals experiencing HT ischemia, the infarct size was dramatically reduced as compared to NT animals (9% ± 3% vs 55% ± 2% of the risk zone, respectively). Importantly, administration of opioid and adenosine receptor antagonists (naloxone [6 mg/kg iv] and 8-( p-sulfophenyl) theophylline [20 mg/kg iv], respectively) did not alter the infarct size or affect the cardioprotective effect of hypothermia. Doses of these 2 antagonists were appropriately chosen since they blunted infarct size reduction induced by selective opioid or adenosine receptor stimulation with morphine (0.3 mg/kg iv) or N6-cyclopentyladenosine ([CPA] 100 μg/kg iv), respectively. Therefore, the cardioprotective effect of mild hypothermia is not triggered by either opioid or adenosine receptor activation, suggesting the involvement of other cardioprotective pathways.
Collapse
Affiliation(s)
- Lys Darbera
- Université Paris-Est, Laboratoire de Pharmacologie, Faculté de Médecine, Créteil, France
- Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - Mourad Chenoune
- Université Paris-Est, Laboratoire de Pharmacologie, Faculté de Médecine, Créteil, France
- Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - Fanny Lidouren
- Université Paris-Est, Laboratoire de Pharmacologie, Faculté de Médecine, Créteil, France
- Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - Bijan Ghaleh
- Université Paris-Est, Laboratoire de Pharmacologie, Faculté de Médecine, Créteil, France
- Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - Michael V. Cohen
- Department of Physiology, University of South Alabama, College of Medicine, Mobile, AL, USA
- Department of Medicine, University of South Alabama, College of Medicine, Mobile, AL, USA
| | - James M. Downey
- Department of Physiology, University of South Alabama, College of Medicine, Mobile, AL, USA
| | - Alain Berdeaux
- Université Paris-Est, Laboratoire de Pharmacologie, Faculté de Médecine, Créteil, France
- Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - Renaud Tissier
- Université Paris-Est, Laboratoire de Pharmacologie, Faculté de Médecine, Créteil, France
- Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| |
Collapse
|
43
|
Jinka TR, Carlson ZA, Moore JT, Drew KL. Altered thermoregulation via sensitization of A1 adenosine receptors in dietary-restricted rats. Psychopharmacology (Berl) 2010; 209:217-24. [PMID: 20186398 PMCID: PMC2892230 DOI: 10.1007/s00213-010-1778-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 01/04/2010] [Indexed: 11/30/2022]
Abstract
RATIONALE Evidence links longevity to dietary restriction (DR). A decrease in body temperature (T(b)) is thought to contribute to enhanced longevity because lower T(b) reduces oxidative metabolism and oxidative stress. It is as yet unclear how DR decreases T(b). OBJECTIVE Here, we test the hypothesis that prolonged DR decreases T(b) by sensitizing adenosine A(1) receptors (A(1)AR) and adenosine-induced cooling. METHODS AND RESULTS Sprague-Dawley rats were dietary restricted using an every-other-day feeding protocol. Rats were fed every other day for 27 days and then administered the A(1)AR agonist, N(6)-cyclohexyladenosine (CHA; 0.5 mg/kg, i.p.). Respiratory rate (RR) and subcutaneous T(b) measured using IPTT-300 transponders were monitored every day and after drug administration. DR animals displayed lower RR on day 20 and lower T(b) on day 22 compared to animals fed ad libitum and displayed a larger response to CHA. In all cases, RR declined before T(b). Contrary to previous reports, a higher dose of CHA (5 mg/kg, i.p.) was lethal in both dietary groups. We next tested the hypothesis that sensitization to the effects of CHA was due to increased surface expression of A(1)AR within the hypothalamus. We report that the abundance of A(1)AR in the membrane fraction increases in hypothalamus, but not cortex of DR rats. CONCLUSION These results suggest that every-other-day feeding lowers T(b) via sensitization of thermoregulatory effects of endogenous adenosine by increasing surface expression of A(1)AR. DISCUSSION Evidence that diet can modulate purinergic signaling has implications for the treatment of stroke, brain injury, epilepsy, and aging.
Collapse
|
44
|
Otis JP, Ackermann LW, Denning GM, Carey HV. Identification of qRT-PCR reference genes for analysis of opioid gene expression in a hibernator. J Comp Physiol B 2009; 180:619-29. [PMID: 20033416 DOI: 10.1007/s00360-009-0430-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 10/14/2009] [Accepted: 11/23/2009] [Indexed: 12/26/2022]
Abstract
Previous work has suggested that central and peripheral opioid signaling are involved in regulating torpor behavior and tissue protection associated with the hibernation phenotype. We used quantitative real-time PCR (qRT-PCR) to measure mRNA levels of opioid peptide precursors and receptors in the brain and heart of summer ground squirrels (Ictidomys tridecemlineatus) and winter hibernating squirrels in the torpid or interbout arousal states. The use of appropriate reference genes for normalization of qRT-PCR gene expression data can have profound effects on the analysis and interpretation of results. This may be particularly important when experimental subjects, such as hibernating animals, undergo significant morphological and/or functional changes during the study. Therefore, an additional goal of this study was to identify stable reference genes for use in qRT-PCR studies of the 13-lined ground squirrel. Expression levels of 10 potential reference genes were measured in the small intestine, liver, brain, and heart, and the optimal combinations of the most stable reference genes were identified by the GeNorm Excel applet. Based on this analysis, we provide recommendations for reference genes to use in each tissue that would be suitable for comparative studies among different activity states. When appropriate normalization of mRNA levels was used, there were no changes in opioid-related genes in heart among the three activity states; in brain, DOR expression was highest during torpor, lowest in interbout arousal and intermediate in summer. The results support the idea that changes in DOR expression may regulate the level of neuronal activity in brain during the annual hibernation cycle and may contribute to hibernation-associated tissue protection.
Collapse
Affiliation(s)
- Jessica P Otis
- Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Dr., Madison, WI 53706, USA
| | | | | | | |
Collapse
|
45
|
González JA, Horjales-Araujo E, Fugger L, Broberger C, Burdakov D. Stimulation of orexin/hypocretin neurones by thyrotropin-releasing hormone. J Physiol 2009; 587:1179-86. [PMID: 19204048 PMCID: PMC2674990 DOI: 10.1113/jphysiol.2008.167940] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 01/27/2009] [Indexed: 02/01/2023] Open
Abstract
Central orexin/hypocretin neurones are critical for sustaining consciousness: their firing stimulates wakefulness and their destruction causes narcolepsy. We explored whether the activity of orexin cells is modulated by thyrotropin-releasing hormone (TRH), an endogenous stimulant of wakefulness and locomotor activity whose mechanism of action is not fully understood. Living orexin neurones were identified by targeted expression of green fluorescent protein (GFP) in acute brain slices of transgenic mice. Using whole-cell patch-clamp recordings, we found that TRH robustly increased the action potential firing rate of these neurones. TRH-induced excitation persisted under conditions of synaptic isolation, and involved a Na(+)-dependent depolarization and activation of a mixed cation current in the orexin cell membrane. By double-label immunohistochemistry, we found close appositions between TRH-immunoreactive nerve terminals and orexin-A-immunoreactive cell bodies. These results identify a new physiological modulator of orexin cell firing, and suggest that orexin cell excitation may contribute to the arousal-enhancing actions of TRH.
Collapse
Affiliation(s)
- J Antonio González
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | | | | | | | | |
Collapse
|
46
|
Nathaniel TI. Brain-regulated metabolic suppression during hibernation: a neuroprotective mechanism for perinatal hypoxia-ischemia. Int J Stroke 2008; 3:98-104. [PMID: 18706003 DOI: 10.1111/j.1747-4949.2008.00186.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hypoxic-ischemic brain injury in the perinatal period is a major cause of chronic disability and acute mortality in newborns. Despite numerous therapeutic strategies that reduce hypoxia-ischemia-induced damage in different experimental animal models, most of them have failed to translate to clinical therapies. This challenge calls for an urgent need to explore novel approaches to develop effective therapies for the clinical management of perinatal hypoxia-ischemia brain injury. This review focuses on studies that investigate neuroprotective related events during mammalian hibernation, characterized by dramatic reductions in several parameters including body temperature, oxygen consumption and heart rate, such that it is difficult to tell if the hibernating animal is dead or alive. The first part of this article reviews the mechanisms of metabolic suppression related events during hibernation. In the second part, hypoxic-ischemic events in the perinatal brain are discussed, and in turn, contrasted with brains experiencing metabolic suppression during mammalian hibernation. In the last part of this article, the diverse neuroprotective adaptations of hibernators and the mechanisms that might be involved in mammalian hibernation, and how they could in turn, contribute to neurprotection during perinatal hypoxia-ischemia related injuries are discussed. This article appraises the novel idea that knowledge of the central mechanisms involved in the regulatory metabolic suppression, during which; hibernators switch themselves off without dissolving their brains could represent brain neuroprotective strategy for the clinical management of perinatal hypoxia-ischemia brain injuries in newborns.
Collapse
Affiliation(s)
- Thomas I Nathaniel
- Center for Natural and Health Sciences, Marywood University, 2300 Adams Avenue, Scranton, PA 18509, USA.
| |
Collapse
|
47
|
Osborne PG, Hashimoto M. Mammalian cerebral metabolism and amino acid neurotransmission during hibernation. J Neurochem 2008; 106:1888-99. [PMID: 18624914 DOI: 10.1111/j.1471-4159.2008.05543.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This report demonstrates that during the torpor phase of hibernation, hamsters utilize (14)C and (13)C glucose in torpor-specific brain metabolic pathways. Microdialysis of (14)C glucose into the striatum rapidly induced a steady state labeling of extracellular fluid (ECF) lactate and labeling of tissue GABA, glutamate, glutamine, and alanine in ipsilateral and contralateral striata. The same tissue metabolites were labeled in cortex, hypothalamus, and brainstem after microdialysis of (14)C lactate into the lateral ventricle. Serine, aspartate, glycine, taurine, tyrosine, and methionine were not synthesized from glucose or lactate during torpor. ECF levels of amino and organic acids were low and unchanging during torpor and increased late during arousal to cenothermia. Labeled intracellular (14)C GABA and glutamate were not communicated to the striatal ECF or ventricular space during torpor. (13)C NMR demonstrated rapid formation of lactate and functional tricarboxylic acid cycles in GABAergic and glutamatergic neurons, and enrichment of glutamine and alanine after i.v. (13)C glucose. Large changes in tissue levels of amino acids occur prior to or during entrance into torpor but not during torpor. It is proposed that cerebral intracellular dehydration, the enlargement of ECF and the biochemistries associated with brain water homeostasis may have a role in regulating hibernation.
Collapse
|
48
|
Miyazawa S, Shimizu Y, Shiina T, Hirayama H, Morita H, Takewaki T. Central A1-receptor activation associated with onset of torpor protects the heart against low temperature in the Syrian hamster. Am J Physiol Regul Integr Comp Physiol 2008; 295:R991-6. [PMID: 18596109 DOI: 10.1152/ajpregu.00142.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Body temperature drops dramatically during hibernation, but the heart retains the ability to contract and is resistant to induction of arrhythmia. Although adaptive changes in the heart prior to hibernation may be involved in the cold-resistant property, it remains unclear whether these changes are sufficient for maintaining cardiac pulsatility under an extreme hypothermic condition. We forcibly induced hypothermia in Syrian hamsters by pentobarbital anesthesia combined with cooling of the animals. This allows reproduction of a hypothermic condition in the absence of possible hibernation-specific reactions. Unlike hypothermia in natural hibernation, the forced induction of hypothermia caused atrioventricular block. Furthermore, J-waves, which are typically observed during hypothermia in nonhibernators, were recorded on an ECG. The origin of the J-wave seemed to be related to irreversible injury of the myocardium, because J-waves remained after recovery of body temperature. An abnormal ECG was also found when hypothermia was induced in hamsters that were well adapted to a cold and darkened environment or hamsters that had already experienced hibernation. These results suggest that acclimatization prior to hibernation does not have a crucial effect at least on acquisition of cardiac resistance to low temperature. In contrast, an abnormal ECG was not observed in the case of hypothermia induced by central administration of an adenosine A1-receptor agonist and subsequent cooling, confirming the importance of the adenosine system for inducing hibernation. Our results suggest that some specific mechanisms, which may be driven by a central adenosine system, operate for maintaining the proper cardiac pulsatility under extreme hypothermia.
Collapse
Affiliation(s)
- Seiji Miyazawa
- Dept. of Basic Veterinary Sciences, Laboratory of Physiology, The United Graduate School of Veterinary Sciences, Gifu Univ., 1-1 Yanagido, Gifu 501-1193, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Colugnati DB, Arida RM, Cravo SL, Schoorlemmer GHM, de Almeida ACG, Cavalheiro EA, Scorza FA. Hibernating mammals in sudden cardiac death in epilepsy: what do they tell us? Med Hypotheses 2007; 70:929-32. [PMID: 18055132 DOI: 10.1016/j.mehy.2007.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2007] [Accepted: 10/23/2007] [Indexed: 10/22/2022]
Abstract
Epilepsy is the most common neurological disorder; approximately 1% of the population worldwide have epilepsy. Moreover, sudden unexpected death in epilepsy (SUDEP) is the most important direct epilepsy-related cause of death. Information concerning risk factors for SUDEP is conflicting, but potential risk factors include: age, early onset of epilepsy, duration of epilepsy, uncontrolled seizures, seizure frequency and AED number. Additionally, the cause of SUDEP is still unknown; however, the most commonly suggested mechanisms are cardiac abnormalities during and between seizures. Very recently, our research group was the first to annunciate that winter temperatures may lead a cardiac abnormalities and hence sudden death, become a new potential risk factor to SUDEP. Quite interesting, several mammalian species have evolved to develop a physiological phenomenon called hibernation as a strategy for survival under adverse cold conditions. From cardiovascular point of view, it has been established that hibernating mammals inherited a stable cardiovascular function as a result of adaptation to extreme external and internal environments during hibernation. For instance, hibernating mammals show resistance to hypothermia at a cellular level, the membrane potentials and excitability are more stable in the cardiac cells of these animals (action potentials (60 mV) have been recorded in hibernators myocardium at -5 degrees C), the aortic smooth muscle cells from hibernators are able to maintain ionic gradients upon prolonged exposure to low temperatures, and cardiac myocytes from hibernating mammals maintain constant levels of intracellular free calcium and forceful contractility at 10 degrees C or lower. Taken together, in this paper we postulate that hibernators have some cardiovascular particularities that confer heart protection that could positively influence the cardiovascular system of patients with epilepsy.
Collapse
Affiliation(s)
- Diego B Colugnati
- Disciplina de Neurologia Experimental, Universidade Federal de São Paulo/Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
50
|
Popov VI, Medvedev NI, Patrushev IV, Ignat'ev DA, Morenkov ED, Stewart MG. Reversible reduction in dendritic spines in CA1 of rat and ground squirrel subjected to hypothermia-normothermia in vivo: A three-dimensional electron microscope study. Neuroscience 2007; 149:549-60. [PMID: 17919827 DOI: 10.1016/j.neuroscience.2007.07.059] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 07/07/2007] [Accepted: 07/18/2007] [Indexed: 10/22/2022]
Abstract
A study was made at electron microscope level of changes in the three-dimensional (3-D) morphology of dendritic spines and postsynaptic densities (PSDs) in CA1 of the hippocampus in ground squirrels, taken either at low temperature during hibernation (brain temperature 2-4 degrees C), or after warming and recovery to the normothermic state (34 degrees C). In addition, the morphology of PSDs and spines was measured in a non-hibernating mammal, rat, subjected to cooling at 2 degrees C at which time core rectal temperature was 15 degrees C, and then after warming to normothermic conditions. Significant differences were found in the proportion of thin and stubby spines, and shaft synapses in CA1 for rats and ground squirrels for normothermia compared with cooling or hibernation. Hypothermia induced a decrease in the proportion of thin spines, and an increase in stubby and shaft spines, but no change in the proportion of mushroom spines. The changes in redistribution of these three categories of spines in ground squirrel are more prominent than in rat. There were no significant differences in synapse density determined for ground squirrels or rats at normal compared with low temperature. Measurement of spine and PSD volume (for mushroom and thin spines) also showed no significant differences between the two functional states in either rats or ground squirrels, nor were there any differences in distances between neighboring synapses. Spinules on dendritic shafts were notable qualitatively during hibernation, but absent in normothermia. These data show that hypothermia results in morphological changes which are essentially similar in both a hibernating and a non-hibernating animal.
Collapse
Affiliation(s)
- V I Popov
- The Open University, Department of Biological Sciences, Faculty of Sciences, Walton Hall, Milton Keynes MK7 6AA, UK
| | | | | | | | | | | |
Collapse
|