1
|
Hao Z, Han Y, Zhao Q, Zhu M, Liu X, Yang Y, An N, He D, Lefai E, Storey KB, Chang H, Xie M. Involvement of Melatonin, Oxidative Stress, and Inflammation in the Protective Mechanism of the Carotid Artery over the Torpor-Arousal Cycle of Ground Squirrels. Int J Mol Sci 2024; 25:12888. [PMID: 39684599 DOI: 10.3390/ijms252312888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Hibernating mammals experience severe hemodynamic changes over the torpor-arousal cycle, with oxygen consumption reaching peaks during the early stage of torpor to re-enter arousal. Melatonin (MT) can improve mitochondrial function and reduce oxidative stress and inflammation. However, the regulatory mechanisms of MT action on the vascular protective function of hibernators are still unclear. Morphology, hemodynamic, mitochondrial oxidative stress, and inflammatory factors of the carotid artery were assessed in ground squirrels who were sampled during summer active (SA), late torpor (LT), and interbout arousal (IBA) conditions. Changes were assessed by methods including hematoxylin and eosin staining, color Doppler ultrasound, ELISA, Western blots, and qPCR. Changes in arterial blood and serum melatonin were also measured by blood gas analyzer and ELISA, whereas mitochondrial oxidative stress and inflammation factors of primary vascular smooth muscle cells (VSMCs) were assessed by qPCR. (1) Intima-media carotid thickness, peak systolic velocity (PSV), end diastolic blood flow velocity (EDV), maximal blood flow rate (Vmax) and pulsatility index (PI) were significantly decreased in the LT group as compared with the SA group, whereas there were no difference between the SA and IBA groups. (2) PO2, oxygen saturation, hematocrit and PCO2 in the arterial blood were significantly increased, and pH was significantly decreased in the LT group as compared with the SA and IBA groups. (3) The serum melatonin concentration was significantly increased in the LT group as compared with the SA and IBA groups. (4) MT treatment significantly reduced the elevated levels of LONP1, NF-κB, NLRP3 and IL-6 mRNA expression of VSMCs under hypoxic conditions. (5) Protein expression of HSP60 and LONP1 in the carotid artery were significantly reduced in the LT and IBA groups as compared with the SA group. (6) The proinflammatory factors IL-1β, IL-6, and TNF-α were reduced in the carotid artery of the LT group as compared with the SA and IBA groups. The carotid artery experiences no oxidative stress or inflammatory response during the torpor-arousal cycle. In addition, melatonin accumulates during torpor and alleviates oxidative stress and inflammatory responses caused by hypoxia in vitro in VSMCs from ground squirrels.
Collapse
Affiliation(s)
- Ziwei Hao
- Department of Aerospace Physiology, Air Force Medical University, Xi'an 710032, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Yuting Han
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Qi Zhao
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Minghui Zhu
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Xiaoxuan Liu
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Yingyu Yang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Ning An
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Dinglin He
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Etienne Lefai
- INRAE, Unité de Nutrition Humaine, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Kenneth B Storey
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Hui Chang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an 710069, China
| | - Manjiang Xie
- Department of Aerospace Physiology, Air Force Medical University, Xi'an 710032, China
| |
Collapse
|
2
|
Ichinose F, Hindle A. Sulfide catabolism in hibernation and neuroprotection. Nitric Oxide 2024; 146:19-23. [PMID: 38521487 PMCID: PMC11055667 DOI: 10.1016/j.niox.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/27/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
The mammalian brain is exquisitely vulnerable to lack of oxygen. However, the mechanism underlying the brain's sensitivity to hypoxia is incompletely understood. In this narrative review, we present a case for sulfide catabolism as a key defense mechanism of the brain against acute oxygen shortage. We will examine literature on the role of sulfide in hypoxia/ischemia, deep hibernation, and leigh syndrome patients, and present our recent data that support the neuroprotective effects of sulfide catabolism and persulfide production. When oxygen levels become low, hydrogen sulfide (H2S) accumulates in brain cells and impairs the ability of these cells to use the remaining, available oxygen to produce energy. In recent studies, we found that hibernating ground squirrels, which can withstand very low levels of oxygen, have high levels of sulfide:quinone oxidoreductase (SQOR) and the capacity to catabolize hydrogen sulfide in the brain. Silencing SQOR increased the sensitivity of the brain of squirrels and mice to hypoxia, whereas neuron-specific SQOR expression prevented hypoxia-induced sulfide accumulation, bioenergetic failure, and ischemic brain injury in mice. Excluding SQOR from mitochondria increased sensitivity to hypoxia not only in the brain but also in heart and liver. Pharmacological agents that scavenge sulfide and/or increase persulfide maintained mitochondrial respiration in hypoxic neurons and made mice resistant to ischemic injury to the brain or spinal cord. Drugs that oxidize hydrogen sulfide and/or increase persulfide may prove to be an effective approach to the treatment of patients experiencing brain injury caused by oxygen deprivation or mitochondrial dysfunction.
Collapse
Affiliation(s)
- Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Allyson Hindle
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; School of Life Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
3
|
Cogut V, Goris M, Jansma A, van der Staaij M, Henning RH. Hippocampal neuroimmune response in mice undergoing serial daily torpor induced by calorie restriction. Front Neuroanat 2024; 18:1334206. [PMID: 38686173 PMCID: PMC11056553 DOI: 10.3389/fnana.2024.1334206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
Hibernating animals demonstrate a remarkable ability to withstand extreme physiological brain changes without triggering adverse neuroinflammatory responses. While hibernators may offer valuable insights into the neuroprotective mechanisms inherent to hibernation, studies using such species are constrained by the limited availability of molecular tools. Laboratory mice may serve as an alternative, entering states of hypometabolism and hypothermia similar to the torpor observed in hibernation when faced with energy shortage. Notably, prolonged calorie restriction (CR) induces serial daily torpor patterns in mice, comparable to species that utilize daily hibernation. Here, we examined the neuroinflammatory response in the hippocampus of male C57BL/6 mice undergoing serial daily torpor induced by a 30% CR for 4 weeks. During daily torpor episodes, CR mice exhibited transient increases in TNF-α mRNA expression, which normalized upon arousal. Concurrently, the CA1 region of the hippocampus showed persistent morphological changes in microglia, characterized by reduced cell branching, decreased cell complexity and altered shape. Importantly, these morphological changes were not accompanied by evident signs of astrogliosis or oxidative stress, typically associated with detrimental neuroinflammation. Collectively, the adaptive nature of the brain's inflammatory response to CR-induced torpor in mice parallels observations in hibernators, highlighting its value for studying the mechanisms of brain resilience during torpor. Such insights could pave the way for novel therapeutic interventions in stroke and neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- Valeria Cogut
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | | | | | | | | |
Collapse
|
4
|
Bernstock JD, Willis CM, Garcia-Segura ME, Gaude E, Anni D, Lee YJ, Thomas LW, Casey A, Vicario N, Leonardi T, Nicaise AM, Gessler FA, Izzy S, Buffelli MR, Seidlitz J, Srinivasan S, Murphy MP, Ashcroft M, Cambiaghi M, Hallenbeck JM, Peruzzotti-Jametti L. Integrative transcriptomic and metabolic analyses of the mammalian hibernating brain identifies a key role for succinate dehydrogenase in ischemic tolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534718. [PMID: 37205496 PMCID: PMC10187245 DOI: 10.1101/2023.03.29.534718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ischemic stroke results in a loss of tissue homeostasis and integrity, the underlying pathobiology of which stems primarily from the depletion of cellular energy stores and perturbation of available metabolites 1 . Hibernation in thirteen-lined ground squirrels (TLGS), Ictidomys tridecemlineatus , provides a natural model of ischemic tolerance as these mammals undergo prolonged periods of critically low cerebral blood flow without evidence of central nervous system (CNS) damage 2 . Studying the complex interplay of genes and metabolites that unfolds during hibernation may provide novel insights into key regulators of cellular homeostasis during brain ischemia. Herein, we interrogated the molecular profiles of TLGS brains at different time points within the hibernation cycle via RNA sequencing coupled with untargeted metabolomics. We demonstrate that hibernation in TLGS leads to major changes in the expression of genes involved in oxidative phosphorylation and this is correlated with an accumulation of the tricarboxylic acid (TCA) cycle intermediates citrate, cis-aconitate, and α-ketoglutarate-αKG. Integration of the gene expression and metabolomics datasets led to the identification of succinate dehydrogenase (SDH) as the critical enzyme during hibernation, uncovering a break in the TCA cycle at that level. Accordingly, the SDH inhibitor dimethyl malonate (DMM) was able to rescue the effects of hypoxia on human neuronal cells in vitro and in mice subjected to permanent ischemic stroke in vivo . Our findings indicate that studying the regulation of the controlled metabolic depression that occurs in hibernating mammals may lead to novel therapeutic approaches capable of increasing ischemic tolerance in the CNS.
Collapse
|
5
|
Oka K, Yamakawa M, Kawamura Y, Kutsukake N, Miura K. The Naked Mole-Rat as a Model for Healthy Aging. Annu Rev Anim Biosci 2023; 11:207-226. [PMID: 36318672 DOI: 10.1146/annurev-animal-050322-074744] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Naked mole-rats (NMRs, Heterocephalus glaber) are the longest-lived rodents with a maximum life span exceeding 37 years. They exhibit a delayed aging phenotype and resistance to age-related functional decline/diseases. Specifically, they do not display increased mortality with age, maintain several physiological functions until nearly the end of their lifetime, and rarely develop cancer and Alzheimer's disease. NMRs live in a hypoxic environment in underground colonies in East Africa and are highly tolerant of hypoxia. These unique characteristics of NMRs have attracted considerable interest from zoological and biomedical researchers. This review summarizes previous studies of the ecology, hypoxia tolerance, longevity/delayed aging, and cancer resistance of NMRs and discusses possible mechanisms contributing to their healthy aging. In addition, we discuss current issues and future perspectives to fully elucidate the mechanisms underlying delayed aging and resistance to age-related diseases in NMRs.
Collapse
Affiliation(s)
- Kaori Oka
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; , ,
| | - Masanori Yamakawa
- Department of Evolutionary Studies of Biosystems, Sokendai (The Graduate University for Advanced Studies), Kanagawa, Japan; ,
| | - Yoshimi Kawamura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; , ,
| | - Nobuyuki Kutsukake
- Department of Evolutionary Studies of Biosystems, Sokendai (The Graduate University for Advanced Studies), Kanagawa, Japan; , .,Research Center for Integrative Evolutionary Science, Sokendai (The Graduate University for Advanced Studies), Kanagawa, Japan
| | - Kyoko Miura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; , , .,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
6
|
Drew KL, Bhowmick S, Laughlin BW, Goropashnaya AV, Tøien Ø, Sugiura MH, Wong A, Pourrezaei K, Barati Z, Chen CY. Opportunities and barriers to translating the hibernation phenotype for neurocritical care. Front Neurol 2023; 14:1009718. [PMID: 36779060 PMCID: PMC9911456 DOI: 10.3389/fneur.2023.1009718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/11/2023] [Indexed: 01/28/2023] Open
Abstract
Targeted temperature management (TTM) is standard of care for neonatal hypoxic ischemic encephalopathy (HIE). Prevention of fever, not excluding cooling core body temperature to 33°C, is standard of care for brain injury post cardiac arrest. Although TTM is beneficial, HIE and cardiac arrest still carry significant risk of death and severe disability. Mammalian hibernation is a gold standard of neuroprotective metabolic suppression, that if better understood might make TTM more accessible, improve efficacy of TTM and identify adjunctive therapies to protect and regenerate neurons after hypoxic ischemia brain injury. Hibernating species tolerate cerebral ischemia/reperfusion better than humans and better than other models of cerebral ischemia tolerance. Such tolerance limits risk of transitions into and out of hibernation torpor and suggests that a barrier to translate hibernation torpor may be human vulnerability to these transitions. At the same time, understanding how hibernating mammals protect their brains is an opportunity to identify adjunctive therapies for TTM. Here we summarize what is known about the hemodynamics of hibernation and how the hibernating brain resists injury to identify opportunities to translate these mechanisms for neurocritical care.
Collapse
Affiliation(s)
- Kelly L. Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - Saurav Bhowmick
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - Bernard W. Laughlin
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - Anna V. Goropashnaya
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - Øivind Tøien
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - M. Hoshi Sugiura
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
| | - Ardy Wong
- Drexel University School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA, United States
| | - Kambiz Pourrezaei
- Drexel University School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA, United States
| | - Zeinab Barati
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, United States
- Barati Medical LLC, Fairbanks, AK, United States
| | - Chao-Yin Chen
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
7
|
Mu D, Wu X, Feijó A, Wu W, Wen Z, Cheng J, Xia L, Yang Q, Shan W, Ge D. Transcriptome analysis of pika heart tissue reveals mechanisms underlying the adaptation of a keystone species on the roof of the world. Front Genet 2022; 13:1020789. [PMID: 36506315 PMCID: PMC9728954 DOI: 10.3389/fgene.2022.1020789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
High-altitude environments impose intense stresses on living organisms and drive striking phenotypic and genetic adaptations, such as hypoxia resistance, cold tolerance, and increases in metabolic capacity and body mass. As one of the most successful and dominant mammals on the Qinghai-Tibetan Plateau (QHTP), the plateau pika (Ochotona curzoniae) has adapted to the extreme environments of the highest altitudes of this region and exhibits tolerance to cold and hypoxia, in contrast to closely related species that inhabit the peripheral alpine bush or forests. To explore the potential genetic mechanisms underlying the adaptation of O. curzoniae to a high-altitude environment, we sequenced the heart tissue transcriptomes of adult plateau pikas (comparing specimens from sites at two different altitudes) and Gansu pikas (O. cansus). Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were used to identify differentially expressed genes (DEGs) and their primary functions. Key genes and pathways related to high-altitude adaptation were identified. In addition to the biological processes of signal transduction, energy metabolism and material transport, the identified plateau pika genes were mainly enriched in biological pathways such as the negative regulation of smooth muscle cell proliferation, the apoptosis signalling pathway, the cellular response to DNA damage stimulus, and ossification involved in bone maturation and heart development. Our results showed that the plateau pika has adapted to the extreme environments of the QHTP via protection against cardiomyopathy, tissue structure alterations and improvements in the blood circulation system and energy metabolism. These adaptations shed light on how pikas thrive on the roof of the world.
Collapse
Affiliation(s)
- Danping Mu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China,Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinlai Wu
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China,Key Laboratory of Zoological Systematics and Application, School of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei, China
| | - Anderson Feijó
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Wu
- CAS Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization & Ecological Restoration and Biodiversity Conservation Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, China
| | - Zhixin Wen
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jilong Cheng
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lin Xia
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qisen Yang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wenjuan Shan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China,*Correspondence: Wenjuan Shan, ; Deyan Ge,
| | - Deyan Ge
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China,*Correspondence: Wenjuan Shan, ; Deyan Ge,
| |
Collapse
|
8
|
Staples JF, Mathers KE, Duffy BM. Mitochondrial Metabolism in Hibernation: Regulation and Implications. Physiology (Bethesda) 2022; 37:0. [PMID: 35658625 DOI: 10.1152/physiol.00006.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Hibernators rapidly and reversibly suppress mitochondrial respiration and whole animal metabolism. Posttranslational modifications likely regulate these mitochondrial changes, which may help conserve energy in winter. These modifications are affected by reactive oxygen species (ROS), so suppressing mitochondrial ROS production may also be important for hibernators, just as it is important for surviving ischemia-reperfusion injury.
Collapse
Affiliation(s)
- James F Staples
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - Katherine E Mathers
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - Brynne M Duffy
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
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: 1.3] [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
|
10
|
Intracellular Signaling. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
11
|
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
|
12
|
Klichkhanov NK, Nikitina ER, Shihamirova ZM, Astaeva MD, Chalabov SI, Krivchenko AI. Erythrocytes of Little Ground Squirrels Undergo Reversible Oxidative Stress During Arousal From Hibernation. Front Physiol 2021; 12:730657. [PMID: 34690805 PMCID: PMC8529035 DOI: 10.3389/fphys.2021.730657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022] Open
Abstract
The hibernation of small mammals is characterized by long torpor bouts alternating with short periods of arousal. During arousal, due to a significant increase in oxygen consumption, tissue perfusion, and the launch of thermogenesis in cells, a large amount of reactive oxygen species (ROS) and nitrogen (RNS) can be formed, which can trigger oxidative stress in cells. To estimate this possibility, we studied the intensity of free-radical processes in the red blood cells (RBCs) of little ground squirrels (LGS; Spermophilus pygmaeus) in the dynamics of arousal from hibernation. We found that in the torpid state, the degree of generation of ROS and RNS (8.3%, p>0.09; 20.7%, p<0.001, respectively), the degree of oxidative modification of membrane lipids and RBC proteins is at a low level (47%, p<0.001; 82.7%, p<0.001, respectively) compared to the summer control. At the same time, the activity of superoxide dismutase (SOD) and catalase (CAT) in RBC is significantly reduced (32.8%, p<0.001; 22.2%, p<0.001, respectively), but not the level of glutathione (GSH). In the torpid state, SOD is activated by exogenous GSH in concentration-dependent manner, which indicates reversible enzyme inhibition. During the arousal of ground squirrels, when the body temperature reaches 25°C, RBCs are exposed oxidative stress. This is confirmed by the maximum increase in the level of uric acid (25.4%, p<0.001) in plasma, a marker of oxidative modification of lipids [thiobarbituric acid reactive substances (TBARS); 82%, p < 0.001] and proteins (carbonyl groups; 499%, p < 0.001) in RBC membranes, as well as the decrease in the level of GSH (19.7%, p < 0.001) in erythrocytes relative to the torpid state and activity of SOD and CAT in erythrocytes to values at the Tb 20°C. After full recovery of body temperature, the level of GSH increases, the ratio of SOD/CAT is restored, which significantly reduces the degree of oxidative damage of lipids and proteins of RBC membranes. Thus, the oxidative stress detected at Tb 25°C was transient and physiologically regulated.
Collapse
Affiliation(s)
| | - Elena R Nikitina
- Laboratory of Comparative Physiology of Respiration, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Maria D Astaeva
- Department of Biochemistry, Dagestan State University, Makhachkala, Russia
| | - Shamil I Chalabov
- Department of Biochemistry, Dagestan State University, Makhachkala, Russia.,Laboratory of Comparative Physiology of Respiration, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Aleksandr I Krivchenko
- Laboratory of Comparative Physiology of Respiration, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
13
|
Hypoxia Tolerant Species: The Wisdom of Nature Translated into Targets for Stroke Therapy. Int J Mol Sci 2021; 22:ijms222011131. [PMID: 34681788 PMCID: PMC8537001 DOI: 10.3390/ijms222011131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Human neurons rapidly die after ischemia and current therapies for stroke management are limited to restoration of blood flow to prevent further brain damage. Thrombolytics and mechanical thrombectomy are the available reperfusion treatments, but most of the patients remain untreated. Neuroprotective therapies focused on treating the pathogenic cascade of the disease have widely failed. However, many animal species demonstrate that neurons can survive the lack of oxygen for extended periods of time. Here, we reviewed the physiological and molecular pathways inherent to tolerant species that have been described to contribute to hypoxia tolerance. Among them, Foxo3 and Eif5A were reported to mediate anoxic survival in Drosophila and Caenorhabditis elegans, respectively, and those results were confirmed in experimental models of stroke. In humans however, the multiple mechanisms involved in brain cell death after a stroke causes translation difficulties to arise making necessary a timely and coordinated control of the pathological changes. We propose here that, if we were able to plagiarize such natural hypoxia tolerance through drugs combined in a pharmacological cocktail it would open new therapeutic opportunities for stroke and likely, for other hypoxic conditions.
Collapse
|
14
|
Abstract
Hibernation is a powerful response of a number of mammalian species to reduce energy during the cold winter season, when food is scarce. Mammalian hibernators survive winter by spending most of the time in a state of torpor, where basal metabolic rate is strongly suppressed and body temperature comes closer to ambient temperature. These torpor bouts are regularly interrupted by short arousals, where metabolic rate and body temperature spontaneously return to normal levels. The mechanisms underlying these changes, and in particular the strong metabolic suppression of torpor, have long remained elusive. As summarized in this Commentary, increasing evidence points to a potential key role for hydrogen sulfide (H2S) in the suppression of mitochondrial respiration during torpor. The idea that H2S could be involved in hibernation originated in some early studies, where exogenous H2S gas was found to induce a torpor-like state in mice, and despite some controversy, the idea persisted. H2S is a widespread signaling molecule capable of inhibiting mitochondrial respiration in vitro and studies found significant in vivo changes in endogenous H2S metabolites associated with hibernation or torpor. Along with increased expression of H2S-synthesizing enzymes during torpor, H2S degradation catalyzed by the mitochondrial sulfide:quinone oxidoreductase (SQR) appears to have a key role in controlling H2S availability for inhibiting respiration. Specifically, in thirteen-lined squirrels, SQR is highly expressed and inhibited in torpor, possibly by acetylation, thereby limiting H2S oxidation and causing inhibition of respiration. H2S may also control other aspects associated with hibernation, such as synthesis of antioxidant enzymes and of SQR itself.
Collapse
Affiliation(s)
| | - Angela Fago
- Department of Biology, Aarhus University, Aarhus C 8000, Denmark
| |
Collapse
|
15
|
Myrka A, Buck L. Cytoskeletal Arrest: An Anoxia Tolerance Mechanism. Metabolites 2021; 11:metabo11080561. [PMID: 34436502 PMCID: PMC8401981 DOI: 10.3390/metabo11080561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/16/2022] Open
Abstract
Polymerization of actin filaments and microtubules constitutes a ubiquitous demand for cellular adenosine-5′-triphosphate (ATP) and guanosine-5′-triphosphate (GTP). In anoxia-tolerant animals, ATP consumption is minimized during overwintering conditions, but little is known about the role of cell structure in anoxia tolerance. Studies of overwintering mammals have revealed that microtubule stability in neurites is reduced at low temperature, resulting in withdrawal of neurites and reduced abundance of excitatory synapses. Literature for turtles is consistent with a similar downregulation of peripheral cytoskeletal activity in brain and liver during anoxic overwintering. Downregulation of actin dynamics, as well as modification to microtubule organization, may play vital roles in facilitating anoxia tolerance. Mitochondrial calcium release occurs during anoxia in turtle neurons, and subsequent activation of calcium-binding proteins likely regulates cytoskeletal stability. Production of reactive oxygen species (ROS) formation can lead to catastrophic cytoskeletal damage during overwintering and ROS production can be regulated by the dynamics of mitochondrial interconnectivity. Therefore, suppression of ROS formation is likely an important aspect of cytoskeletal arrest. Furthermore, gasotransmitters can regulate ROS levels, as well as cytoskeletal contractility and rearrangement. In this review we will explore the energetic costs of cytoskeletal activity, the cellular mechanisms regulating it, and the potential for cytoskeletal arrest being an important mechanism permitting long-term anoxia survival in anoxia-tolerant species, such as the western painted turtle and goldfish.
Collapse
Affiliation(s)
- Alexander Myrka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
| | - Leslie Buck
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Correspondence: ; Tel.: +1-416-978-3506
| |
Collapse
|
16
|
Hepatic resistance to cold ferroptosis in a mammalian hibernator Syrian hamster depends on effective storage of diet-derived α-tocopherol. Commun Biol 2021; 4:796. [PMID: 34172811 PMCID: PMC8233303 DOI: 10.1038/s42003-021-02297-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Mammalian hibernators endure severe and prolonged hypothermia that is lethal to non-hibernators, including humans and mice. The mechanisms responsible for the cold resistance remain poorly understood. Here, we found that hepatocytes from a mammalian hibernator, the Syrian hamster, exhibited remarkable resistance to prolonged cold culture, whereas murine hepatocytes underwent cold-induced cell death that fulfills the hallmarks of ferroptosis such as necrotic morphology, lipid peroxidation and prevention by an iron chelator. Unexpectedly, hepatocytes from Syrian hamsters exerted resistance to cold- and drug-induced ferroptosis in a diet-dependent manner, with the aid of their superior ability to retain dietary α-tocopherol (αT), a vitamin E analog, in the liver and blood compared with those of mice. The liver phospholipid composition is less susceptible to peroxidation in Syrian hamsters than in mice. Altogether, the cold resistance of the hibernator’s liver is established by the ability to utilize αT effectively to prevent lipid peroxidation and ferroptosis. Daisuke Anegawa et al. investigated the mechanisms responsible for cold resistance in the Syrian hamster’s hepatocytes, which exhibited remarkable resistance to prolonged cold culture. Their results suggest that hepatocytes exhibit diet-dependent resistance to cold, which is linked to the retention of α-tocopherol in the liver.
Collapse
|
17
|
Marutani E, Morita M, Hirai S, Kai S, Grange RMH, Miyazaki Y, Nagashima F, Traeger L, Magliocca A, Ida T, Matsunaga T, Flicker DR, Corman B, Mori N, Yamazaki Y, Batten A, Li R, Tanaka T, Ikeda T, Nakagawa A, Atochin DN, Ihara H, Olenchock BA, Shen X, Nishida M, Hanaoka K, Kevil CG, Xian M, Bloch DB, Akaike T, Hindle AG, Motohashi H, Ichinose F. Sulfide catabolism ameliorates hypoxic brain injury. Nat Commun 2021; 12:3108. [PMID: 34035265 PMCID: PMC8149856 DOI: 10.1038/s41467-021-23363-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 04/27/2021] [Indexed: 01/09/2023] Open
Abstract
The mammalian brain is highly vulnerable to oxygen deprivation, yet the mechanism underlying the brain's sensitivity to hypoxia is incompletely understood. Hypoxia induces accumulation of hydrogen sulfide, a gas that inhibits mitochondrial respiration. Here, we show that, in mice, rats, and naturally hypoxia-tolerant ground squirrels, the sensitivity of the brain to hypoxia is inversely related to the levels of sulfide:quinone oxidoreductase (SQOR) and the capacity to catabolize sulfide. Silencing SQOR increased the sensitivity of the brain to hypoxia, whereas neuron-specific SQOR expression prevented hypoxia-induced sulfide accumulation, bioenergetic failure, and ischemic brain injury. Excluding SQOR from mitochondria increased sensitivity to hypoxia not only in the brain but also in heart and liver. Pharmacological scavenging of sulfide maintained mitochondrial respiration in hypoxic neurons and made mice resistant to hypoxia. These results illuminate the critical role of sulfide catabolism in energy homeostasis during hypoxia and identify a therapeutic target for ischemic brain injury.
Collapse
Affiliation(s)
- Eizo Marutani
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichi Hirai
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Shinichi Kai
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Robert M H Grange
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yusuke Miyazaki
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Fumiaki Nagashima
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lisa Traeger
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aurora Magliocca
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuro Matsunaga
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Daniel R Flicker
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin Corman
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Naohiro Mori
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yumiko Yamazaki
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Annabelle Batten
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca Li
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tomohiro Tanaka
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences & Exploratory Research Center on Life and Living Systems & Center for Novel Science Initiatives, National Institutes of Natural Sciences, Okazaki, Japan
| | - Takamitsu Ikeda
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Akito Nakagawa
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Dmitriy N Atochin
- Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka, Japan
| | - Benjamin A Olenchock
- Harvard Medical School, Boston, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, The Brigham and Women's Hospital, Boston, MA, USA
| | - Xinggui Shen
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences & Exploratory Research Center on Life and Living Systems & Center for Novel Science Initiatives, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Ming Xian
- Department of Chemistry, Brown University, Providence, RI, USA
| | - Donald B Bloch
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Allyson G Hindle
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- School of Life Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Zakharova NM, Tarahovsky YS, Komelina NP, Khrenov MO, Kovtun AL. Pharmacological torpor prolongs rat survival in lethal normobaric hypoxia. J Therm Biol 2021; 98:102906. [PMID: 34016333 DOI: 10.1016/j.jtherbio.2021.102906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022]
Abstract
Resistance to hypoxia is one of the most prominent features of natural hibernation and is expected to be present in the pharmacological torpor (PT) that simulates hibernation. We studied resistance to lethal hypoxia (3.5% oxygen content) in rats under PT. To initiate PT, we used the previously developed pharmacological composition (PC) which, after a single intravenous injection, can induce a daily decrease in Tb by 7 °C-8 °C at the environmental temperature of 22 °C-23 °C. Half-survival (median) time of rats in lethal hypoxia was found to increase from 5 ± 0.8 min in anesthetized control rats to 150 ± 12 min in rats injected with PC, which is a 30-fold increase. Behavioral tests after PT and hypoxia, including the traveling distance, the number of rearing and grooming episodes, revealed that animal responses are significantly restored within a week. It is assumed that the discovered unprecedented resistance of artificially torpid rats to lethal hypoxia may open up broad prospects for the therapeutic use of PT for preconditioning to various damaging factors, treatment of diseases, and extend the so-called "golden hour" for lifesaving interventions.
Collapse
Affiliation(s)
| | - Yury S Tarahovsky
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia; Institute of Theoretical and Experimental Biophysics, RAS, Pushchino, Moscow Region 142290, Russia.
| | - Natalia P Komelina
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia
| | - Maxim O Khrenov
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia
| | | |
Collapse
|
19
|
Dias IB, Bouma HR, Henning RH. Unraveling the Big Sleep: Molecular Aspects of Stem Cell Dormancy and Hibernation. Front Physiol 2021; 12:624950. [PMID: 33867999 PMCID: PMC8047423 DOI: 10.3389/fphys.2021.624950] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
Tissue-resident stem cells may enter a dormant state, also known as quiescence, which allows them to withstand metabolic stress and unfavorable conditions. Similarly, hibernating mammals can also enter a state of dormancy used to evade hostile circumstances, such as food shortage and low ambient temperatures. In hibernation, the dormant state of the individual and its cells is commonly known as torpor, and is characterized by metabolic suppression in individual cells. Given that both conditions represent cell survival strategies, we here compare the molecular aspects of cellular quiescence, particularly of well-studied hematopoietic stem cells, and torpor at the cellular level. Critical processes of dormancy are reviewed, including the suppression of the cell cycle, changes in metabolic characteristics, and cellular mechanisms of dealing with damage. Key factors shared by hematopoietic stem cell quiescence and torpor include a reversible activation of factors inhibiting the cell cycle, a shift in metabolism from glucose to fatty acid oxidation, downregulation of mitochondrial activity, key changes in hypoxia-inducible factor one alpha (HIF-1α), mTOR, reversible protein phosphorylation and autophagy, and increased radiation resistance. This similarity is remarkable in view of the difference in cell populations, as stem cell quiescence regards proliferating cells, while torpor mainly involves terminally differentiated cells. A future perspective is provided how to advance our understanding of the crucial pathways that allow stem cells and hibernating animals to engage in their 'great slumbers.'
Collapse
Affiliation(s)
- Itamar B. Dias
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hjalmar R. Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
20
|
Matsuo T, Isosaka T, Tang L, Soga T, Kobayakawa R, Kobayakawa K. Artificial hibernation/life-protective state induced by thiazoline-related innate fear odors. Commun Biol 2021; 4:101. [PMID: 33483561 PMCID: PMC7822961 DOI: 10.1038/s42003-020-01629-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022] Open
Abstract
Innate fear intimately connects to the life preservation in crises, although this relationships is not fully understood. Here, we report that presentation of a supernormal innate fear inducer 2-methyl-2-thiazoline (2MT), but not learned fear stimuli, induced robust systemic hypothermia/hypometabolism and suppressed aerobic metabolism via phosphorylation of pyruvate dehydrogenase, thereby enabling long-term survival in a lethal hypoxic environment. These responses exerted potent therapeutic effects in cutaneous and cerebral ischemia/reperfusion injury models. In contrast to hibernation, 2MT stimulation accelerated glucose uptake in the brain and suppressed oxygen saturation in the blood. Whole-brain mapping and chemogenetic activation revealed that the sensory representation of 2MT orchestrates physiological responses via brain stem Sp5/NST to midbrain PBN pathway. 2MT, as a supernormal stimulus of innate fear, induced exaggerated, latent life-protective effects in mice. If this system is preserved in humans, it may be utilized to give rise to a new field: "sensory medicine."
Collapse
Affiliation(s)
- Tomohiko Matsuo
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoko Isosaka
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan
| | - Lijun Tang
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
| | - Reiko Kobayakawa
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan.
| | - Ko Kobayakawa
- Institute of Biomedical Science, Kansai Medical University, Osaka, 573-1010, Japan.
| |
Collapse
|
21
|
Shi Z, Qin M, Huang L, Xu T, Chen Y, Hu Q, Peng S, Peng Z, Qu LN, Chen SG, Tuo QH, Liao DF, Wang XP, Wu RR, Yuan TF, Li YH, Liu XM. Human torpor: translating insights from nature into manned deep space expedition. Biol Rev Camb Philos Soc 2020; 96:642-672. [PMID: 33314677 DOI: 10.1111/brv.12671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
During a long-duration manned spaceflight mission, such as flying to Mars and beyond, all crew members will spend a long period in an independent spacecraft with closed-loop bioregenerative life-support systems. Saving resources and reducing medical risks, particularly in mental heath, are key technology gaps hampering human expedition into deep space. In the 1960s, several scientists proposed that an induced state of suppressed metabolism in humans, which mimics 'hibernation', could be an ideal solution to cope with many issues during spaceflight. In recent years, with the introduction of specific methods, it is becoming more feasible to induce an artificial hibernation-like state (synthetic torpor) in non-hibernating species. Natural torpor is a fascinating, yet enigmatic, physiological process in which metabolic rate (MR), body core temperature (Tb ) and behavioural activity are reduced to save energy during harsh seasonal conditions. It employs a complex central neural network to orchestrate a homeostatic state of hypometabolism, hypothermia and hypoactivity in response to environmental challenges. The anatomical and functional connections within the central nervous system (CNS) lie at the heart of controlling synthetic torpor. Although progress has been made, the precise mechanisms underlying the active regulation of the torpor-arousal transition, and their profound influence on neural function and behaviour, which are critical concerns for safe and reversible human torpor, remain poorly understood. In this review, we place particular emphasis on elaborating the central nervous mechanism orchestrating the torpor-arousal transition in both non-flying hibernating mammals and non-hibernating species, and aim to provide translational insights into long-duration manned spaceflight. In addition, identifying difficulties and challenges ahead will underscore important concerns in engineering synthetic torpor in humans. We believe that synthetic torpor may not be the only option for manned long-duration spaceflight, but it is the most achievable solution in the foreseeable future. Translating the available knowledge from natural torpor research will not only benefit manned spaceflight, but also many clinical settings attempting to manipulate energy metabolism and neurobehavioural functions.
Collapse
Affiliation(s)
- Zhe Shi
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.,Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Meng Qin
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lu Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Tao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qin Hu
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, 100024, China
| | - Sha Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhuang Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Li-Na Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shan-Guang Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Qin-Hui Tuo
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Duan-Fang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xiao-Ping Wang
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ren-Rong Wu
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China
| | - Ying-Hui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xin-Min Liu
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| |
Collapse
|
22
|
Morrison ML, Iwata A, Wick ML, VandenEkart E, Insko MA, Henning DJ, Frare C, Rice SA, Drew KL, Maier RV, Roth MB. Iodine Redistribution During Trauma, Sepsis, and Hibernation: An Evolutionarily Conserved Response to Severe Stress. Crit Care Explor 2020; 2:e0215. [PMID: 33063025 PMCID: PMC7531756 DOI: 10.1097/cce.0000000000000215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE We performed these studies to learn how iodine in the form of free iodide behaves during stress. DESIGN Prospective observational trial using samples obtained from human trauma patients and retrospective observational study using remnant samples from human sepsis patients and arctic ground squirrels. Preclinical interventional study using hind-limb ischemia and reperfusion injury in mice. SETTING Level I trauma center emergency room and ICU and animal research laboratories. SUBJECTS Adult human sepsis and trauma patients, wild-caught adult arctic ground squirrels, and sexually mature laboratory mice. INTERVENTIONS Ischemia and reperfusion injury was induced in mice by temporary application of tourniquet to one hind-limb. Iodide was administered IV just prior to reperfusion. MEASUREMENTS AND MAIN RESULTS Free iodide was measured using ion chromatography. Relative to iodide in plasma from normal donors, iodide was increased 17-fold in plasma from trauma patients and 26-fold in plasma from sepsis patients. In arctic ground squirrels, iodide increases over three-fold during hibernation. And during ischemia/reperfusion injury in mice, iodide accumulates in ischemic tissue and reduces both local and systemic tissue damage. CONCLUSIONS Iodide redistributes during stress and improves outcome after injury. Essential functions of iodide may have contributed to its evolutionary selection and be useful as a therapeutic intervention for human patients.
Collapse
Affiliation(s)
- Michael L Morrison
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Akiko Iwata
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Merry L Wick
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Emily VandenEkart
- Laboratory and Clinical Research, Faraday Pharmaceuticals, Seattle, WA
| | - Michael A Insko
- Laboratory and Clinical Research, Faraday Pharmaceuticals, Seattle, WA
| | - Daniel J Henning
- Department of Emergency Medicine, University of Washington, Seattle, WA
| | - Carla Frare
- Department of Chemistry and Biochemistry and Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK
| | - Sarah A Rice
- Department of Chemistry and Biochemistry and Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK
| | - Kelly L Drew
- Department of Chemistry and Biochemistry and Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK
| | - Ronald V Maier
- Department of Surgery, Harborview Medical Center and University of Washington, Seattle, WA
| | - Mark B Roth
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
23
|
Hadj-Moussa H, Wijenayake S, Storey KB. Multi-tissue profile of NFκB pathway regulation during mammalian hibernation. Comp Biochem Physiol B Biochem Mol Biol 2020; 246-247:110460. [PMID: 32445797 DOI: 10.1016/j.cbpb.2020.110460] [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: 02/03/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 11/17/2022]
Abstract
Hibernators have evolved effective mechanisms to overcome the challenges of torpor-arousal cycling. This study focuses on the antioxidant and inflammatory defenses under the control of the redox-sensitive and inflammatory-centered NFκB transcription factor in the thirteen-lined ground squirrel (Ictidomys tridecemlineatus), a well-established model of mammalian hibernation. While hibernators significantly depress oxygen consumption and overall metabolic rate during torpor, arousal brings with it a rapid increase in respiration that is associated with an influx of reactive oxygen species. As such, hibernators employ a variety of antioxidant defenses to combat oxidative damage. Herein, we used Luminex multiplex technology to examine the expression of key proteins in the NFκB transcriptional network, including NFκB, super-repressor IκBα, upstream activators TNFR1 and FADD, and downstream target c-Myc. Transcription factor DNA-binding ELISAs were also used to measure the relative degree of NFκB binding to DNA during hibernation. Analyses were performed across eight different tissues, cerebral cortex, brainstem, white and brown adipose tissue, heart, liver, kidney, and spleen, during euthermic control and late torpor to highlight tissue-specific NFκB mediated cytoprotective responses against oxidative stress experienced during torpor-arousal. Our findings demonstrated brain-specific NFκB activation during torpor, with elevated levels of upstream activators, inactive-phosphorylated IκBα, active-phosphorylated NFκB, and enhanced NFκB-DNA binding. Protein levels of downstream protein, c-Myc, also increased in the brain and adipose tissues during late torpor. The results show that NFκB regulation might serve a critical neuroprotective and cytoprotective role in hibernating brains and selective peripheral tissue.
Collapse
Affiliation(s)
- Hanane Hadj-Moussa
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - Sanoji Wijenayake
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, ON, Canada; Department of Biological Sciences and Center for Environmental Epigenetics and Development, University of Toronto, Toronto, ON, Canada
| | - Kenneth B Storey
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
24
|
Abstract
Despite thousands of neuroprotectants demonstrating promise in preclinical trials, a neuroprotective therapeutic has yet to be approved for the treatment of acute brain injuries such as stroke or traumatic brain injury. Developing a more detailed understanding of models and populations demonstrating "neurological resilience" in spite of brain injury can give us important insights into new translational therapies. Resilience is the process of active adaptation to a stressor. In the context of neuroprotection, models of preconditioning and unique animal models of extreme physiology (such as hibernating species) reliably demonstrate resilience in the laboratory setting. In the clinical setting, resilience is observed in young patients and can be found in those with specific genetic polymorphisms. These important examples of resilience can help transform and extend the current neuroprotective framework from simply countering the injurious cascade into one that anticipates, monitors, and optimizes patients' physiological responses from the time of injury throughout the process of recovery. This review summarizes the underpinnings of key adaptations common to models of resilience and how this understanding can be applied to new neuroprotective approaches.
Collapse
Affiliation(s)
- Neel S Singhal
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA.
| | - Chung-Huan Sun
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Evan M Lee
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Dengke K Ma
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| |
Collapse
|
25
|
Shen-Hui X, Fu WW, Zhang J, Wang HP, Dang K, Chang H, Gao YF. Different fuel regulation in two types of myofiber results in different antioxidant strategies in Daurian ground squirrels (Spermophilus dauricus) during hibernation. J Exp Biol 2020:jeb.231639. [PMID: 34005794 DOI: 10.1242/jeb.231639] [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: 06/20/2020] [Accepted: 12/08/2020] [Indexed: 11/20/2022]
Abstract
We previously showed that different skeletal muscles in Daurian ground squirrels (Spermophilus dauricus) possess different antioxidant strategies during hibernation; however, the reason for these varied strategies remains unclear. To clarify this issue, we studied REDD1, FOXO4, PGC-1α, FOXO1, and atrogin-1 proteins to determine the potential cause of the different antioxidant strategies in Daurian ground squirrels during hibernation, and to clarify whether different strategies affect atrophy-related signals. Results showed that the soleus (SOL) muscle experienced intracellular hypoxia during interbout arousal, but no oxidative stress. This may be due to increased PGC-1α expression enhancing antioxidant capacity in the SOL under hypoxic conditions. Extensor digitorum longus (EDL) muscle showed no change in oxidative stress, hypoxia, or antioxidant capacity during hibernation. The FOXO1 and PGC-1α results strongly suggested differentially regulated fuel metabolism in the SOL and EDL muscles during hibernation, i.e., enhanced lipid oxidation and maintained anaerobic glycolysis, respectively. Atrogin-1 expression did not increase during hibernation in either the SOL or EDL, indicating that protein synthesis was not inhibited by atrogin-1. Thus, our results suggest that different fuel regulation may be one mechanism related to antioxidant defense strategy formation in different kinds of skeletal muscle fibers of Daurian ground squirrels during hibernation.
Collapse
Affiliation(s)
- Xu Shen-Hui
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Wei-Wei Fu
- Shaanxi Key Laboratory for Animal Conservation, Shaanxi Institute of Zoology, Xi'an Shaanxi 710032, China
| | - Jie Zhang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Hui-Ping Wang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Kai Dang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an Shaanxi, China
| | - Hui Chang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Yun-Fang Gao
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| |
Collapse
|
26
|
Postnatal changes in O2 and CO2 sensitivity in rodents. Respir Physiol Neurobiol 2020; 272:103313. [DOI: 10.1016/j.resp.2019.103313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/31/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023]
|
27
|
Wei Y, Zhang J, Yan X, Peng X, Xu S, Chang H, Wang H, Gao Y. Remarkable Protective Effects of Nrf2-Mediated Antioxidant Enzymes and Tissue Specificity in Different Skeletal Muscles of Daurian Ground Squirrels Over the Torpor-Arousal Cycle. Front Physiol 2019; 10:1449. [PMID: 31824343 PMCID: PMC6883408 DOI: 10.3389/fphys.2019.01449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/08/2019] [Indexed: 01/07/2023] Open
Abstract
Hibernating mammals experience conditions of extreme oxidative stress, such as fasting, muscle disuse, and repeated hypoxic ischemia-reperfusion, during the torpor-arousal cycle. Despite this, they experience little oxidative injury and are thus an interesting model of anti-oxidative damage. Thus, in the current study, we explored the levels and underlying mechanism of oxidative stress and antioxidant capacity in three skeletal muscles [slow-twitch soleus (SOL), fast-twitch extensor digitorum longus (EDL), and mixed gastrocnemius (GAS)] of Daurian ground squirrels (Spermophilus dauricus) during hibernation. Results showed that hydrogen peroxide content in the EDL and GAS decreased significantly during pre-hibernation (PRE) and late torpor (LT) compared to levels in the summer active (SA) group. Furthermore, relative to SA levels, malondialdehyde content decreased significantly during interbout arousal (IBA) and early torpor (ET) in all three skeletal muscles and decreased in the EDL and GAS during LT. Compared with the SA group, glutathione peroxidase 1 (GPx1) and catalase (CAT) protein expression in the SOL and superoxide dismutase 1 (SOD1) and SOD2 expression in the GAS increased significantly during the entire hibernation season. Furthermore, SOD1 in the IBA group and CAT and GPx1 in the ET and LT groups increased significantly in the EDL. The activities of most tested antioxidant enzymes were higher in the IBA group than in the LT group, whereas CAT remained highly active throughout the hibernation season in all three muscles. Nrf2 and p-Nrf2 protein levels were significantly elevated in the SOL and EDL during hibernation, and increased during the PRE, IBA, and ET states in the GAS. Thus, activation of the Nrf2/Keap1 antioxidant pathway resulted in the elimination of excess reactive oxygen species (ROS). Specifically, ROS levels were maintained at physiological levels by the up-regulation of antioxidant enzyme expression in skeletal muscles under oxidative stress during hibernation, thus preventing oxidative injury over the torpor-arousal cycle. Different antioxidant patterns and oxidative stress levels were also observed among the different skeletal muscles of hibernating Daurian ground squirrels.
Collapse
Affiliation(s)
- Yanhong Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China.,School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jie Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Xia Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Xin Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Shenhui Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Hui Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Huiping Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Yunfang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| |
Collapse
|
28
|
Gattoni G, Bernocchi G. Calcium-Binding Proteins in the Nervous System during Hibernation: Neuroprotective Strategies in Hypometabolic Conditions? Int J Mol Sci 2019; 20:E2364. [PMID: 31086053 PMCID: PMC6540041 DOI: 10.3390/ijms20092364] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/06/2019] [Accepted: 05/11/2019] [Indexed: 02/07/2023] Open
Abstract
Calcium-binding proteins (CBPs) can influence and react to Ca2+ transients and modulate the activity of proteins involved in both maintaining homeostatic conditions and protecting cells in harsh environmental conditions. Hibernation is a strategy that evolved in vertebrate and invertebrate species to survive in cold environments; it relies on molecular, cellular, and behavioral adaptations guided by the neuroendocrine system that together ensure unmatched tolerance to hypothermia, hypometabolism, and hypoxia. Therefore, hibernation is a useful model to study molecular neuroprotective adaptations to extreme conditions, and can reveal useful applications to human pathological conditions. In this review, we describe the known changes in Ca2+-signaling and the detection and activity of CBPs in the nervous system of vertebrate and invertebrate models during hibernation, focusing on cytosolic Ca2+ buffers and calmodulin. Then, we discuss these findings in the context of the neuroprotective and neural plasticity mechanisms in the central nervous system: in particular, those associated with cytoskeletal proteins. Finally, we compare the expression of CBPs in the hibernating nervous system with two different conditions of neurodegeneration, i.e., platinum-induced neurotoxicity and Alzheimer's disease, to highlight the similarities and differences and demonstrate the potential of hibernation to shed light into part of the molecular mechanisms behind neurodegenerative diseases.
Collapse
Affiliation(s)
- Giacomo Gattoni
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Graziella Bernocchi
- Former Full Professor of Zoology, Neurogenesis and Comparative Neuromorphology, (Residence address) Viale Matteotti 73, I-27100 Pavia, Italy.
| |
Collapse
|
29
|
Zhang J, Wei Y, Qu T, Wang Z, Xu S, Peng X, Yan X, Chang H, Wang H, Gao Y. Prosurvival roles mediated by the PERK signaling pathway effectively prevent excessive endoplasmic reticulum stress-induced skeletal muscle loss during high-stress conditions of hibernation. J Cell Physiol 2019; 234:19728-19739. [PMID: 30941772 DOI: 10.1002/jcp.28572] [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: 11/25/2018] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 01/13/2023]
Abstract
Stress conditions like hypoxia, ischemia, and ischemia/reperfusion can trigger excessive endoplasmic reticulum stress (ERS), which can lead to cell apoptosis-induced skeletal muscle atrophy in non-hibernators. However, although hibernators experience multiple stress conditions during hibernation, their skeletal muscles appear to be well protected. We hypothesize that hibernators effectively avoid cell apoptosis, at least partially, by controlling ERS level. Here, we focused on the potential occurrence of ERS and how hibernators cope with it during different hibernation states. Results indicated that the protein expression levels of glucose-regulated protein 78 (GRP78), phosphorylated PKR-like ER protein kinase, phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α), and activating transcription factor 4 were significantly increased during hibernation, but primarily recovered in posthibernation. In the torpor-arousal cycle, the expression levels of the above indicators were lower during inter-bout arousal (IBA) than that during late torpor (LT). However, there was no change in C/EBP homologous protein expression and no apoptosis in skeletal muscles during the different hibernation states. In conclusion, the upregulation of p-eIF2α and GRP78 were identified as two crucial mechanisms mediated by the PERK signaling pathway to alleviate elevated ERS. The downregulation of ERS during IBA may be a unique countermeasure for hibernating squirrels to prevent excessive ERS. Thus, these special anti-excessive ERS abilities of ground squirrels contribute to the prevention of skeletal muscle cell apoptosis during hibernation.
Collapse
Affiliation(s)
- Jie Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Yanhong Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China.,School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Ting Qu
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Zhe Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Shenhui Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Xin Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Xia Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Hui Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Huiping Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | - Yunfang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an, China.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| |
Collapse
|
30
|
Devaux JBL, Hedges CP, Birch N, Herbert N, Renshaw GMC, Hickey AJR. Acidosis Maintains the Function of Brain Mitochondria in Hypoxia-Tolerant Triplefin Fish: A Strategy to Survive Acute Hypoxic Exposure? Front Physiol 2019; 9:1941. [PMID: 30713504 PMCID: PMC6346031 DOI: 10.3389/fphys.2018.01941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/22/2018] [Indexed: 11/13/2022] Open
Abstract
The vertebrate brain is generally very sensitive to acidosis, so a hypoxia-induced decrease in pH is likely to have an effect on brain mitochondria (mt). Mitochondrial respiration (JO2) is required to generate an electrical gradient (ΔΨm) and a pH gradient to power ATP synthesis, yet the impact of pH modulation on brain mt function remains largely unexplored. As intertidal fishes within rock pools routinely experience hypoxia and reoxygenation, they would most likely experience changes in cellular pH. We hence compared four New Zealand triplefin fish species ranging from intertidal hypoxia-tolerant species (HTS) to subtidal hypoxia-sensitive species (HSS). We predicted that HTS would tolerate acidosis better than HSS in terms of sustaining mt structure and function. Using respirometers coupled to fluorimeters and pH electrodes, we titrated lactic-acid to decrease the pH of the media, and simultaneously recorded JO2, ΔΨm, and H+ buffering capacities within permeabilized brain and swelling of mt isolated from non-permeabilized brains. We then measured ATP synthesis rates in the most HTS (Bellapiscus medius) and the HSS (Forsterygion varium) at pH 7.25 and 6.65. Mitochondria from HTS brain did have greater H+ buffering capacities than HSS mt (∼10 mU pH.mgprotein -1). HTS mt swelled by 40% when exposed to a decrease of 1.5 pH units, and JO2 was depressed by up to 15% in HTS. However, HTS were able to maintain ΔΨm near -120 mV. Estimates of work, in terms of charges moved across the mt inner-membrane, suggested that with acidosis, HTS mt may in part harness extra-mt H+ to maintain ΔΨm, and could therefore support ATP production. This was confirmed with elevated ATP synthesis rates and enhanced P:O ratios at pH 6.65 relative to pH 7.25. In contrast, mt volumes and ΔΨm decreased downward pH 6.9 in HSS mt and paradoxically, JO2 increased (∼25%) but ATP synthesis and P:O ratios were depressed at pH 6.65. This indicates a loss of coupling in the HSS with acidosis. Overall, the mt of these intertidal fish have adaptations that enhance ATP synthesis efficiency under acidic conditions such as those that occur in hypoxic or reoxygenated brain.
Collapse
Affiliation(s)
- Jules B L Devaux
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Christopher P Hedges
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Nigel Birch
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Neill Herbert
- Institute of Marine Science, The University Auckland, Auckland, New Zealand
| | - Gillian M C Renshaw
- School of Allied Health Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Anthony J R Hickey
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Caneo M, Julian V, Byrne AB, Alkema MJ, Calixto A. Diapause induces functional axonal regeneration after necrotic insult in C. elegans. PLoS Genet 2019; 15:e1007863. [PMID: 30640919 PMCID: PMC6347329 DOI: 10.1371/journal.pgen.1007863] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 01/25/2019] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Many neurons are unable to regenerate after damage. The ability to regenerate after an insult depends on life stage, neuronal subtype, intrinsic and extrinsic factors. C. elegans is a powerful model to test the genetic and environmental factors that affect axonal regeneration after damage, since its axons can regenerate after neuronal insult. Here we demonstrate that diapause promotes the complete morphological regeneration of truncated touch receptor neuron (TRN) axons expressing a neurotoxic MEC-4(d) DEG/ENaC channel. Truncated axons of different lengths were repaired during diapause and we observed potent axonal regrowth from somas alone. Complete morphological regeneration depends on DLK-1 but neuronal sprouting and outgrowth is DLK-1 independent. We show that TRN regeneration is fully functional since animals regain their ability to respond to mechanical stimulation. Thus, diapause induced regeneration provides a simple model of complete axonal regeneration which will greatly facilitate the study of environmental and genetic factors affecting the rate at which neurons die.
Collapse
Affiliation(s)
- Mauricio Caneo
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago de Chile, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
| | - Victoria Julian
- Neurobiology Department, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Alexandra B. Byrne
- Neurobiology Department, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Mark J. Alkema
- Neurobiology Department, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Andrea Calixto
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago de Chile, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
- * E-mail: ,
| |
Collapse
|
32
|
Bonis A, Anderson L, Talhouarne G, Schueller E, Unke J, Krus C, Stokka J, Koepke A, Lehrer B, Schuh A, Andersen JJ, Cooper S. Cardiovascular resistance to thrombosis in 13-lined ground squirrels. J Comp Physiol B 2018; 189:167-177. [PMID: 30317383 DOI: 10.1007/s00360-018-1186-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/08/2018] [Accepted: 10/06/2018] [Indexed: 12/24/2022]
Abstract
13-lined ground squirrels (Ictidomys tridecemlineatus) enter hibernation as a survival strategy during extreme environmental conditions. Typical ground squirrel hibernation is characterized by prolonged periods of torpor with significantly reduced heart rate, blood pressure, and blood flow, interrupted every few weeks by brief interbout arousals (IBA) during which blood flow fluctuates dramatically. These physiological conditions should increase the risk of stasis-induced blood clots and myocardial ischemia. However, ground squirrels have adapted to survive repeated bouts of torpor and IBA without forming lethal blood clots or sustaining lethal ischemic myocardial damage. The purpose of this study was to determine if ground squirrels are resistant to thrombosis and myocardial ischemia during hibernation. Blood markers of coagulation, fibrinolysis, thrombosis, and ischemia, as well as histological markers of myocardial ischemia were measured throughout the annual hibernation cycle. Hibernating ground squirrels were also treated with isoprenaline to induce myocardial ischemia. Thrombin-antithrombin complex levels were significantly reduced (p < 0.05) during hibernation, while D-dimer level remained unchanged throughout the annual cycle, both consistent with an antithrombotic state. During torpor, the ground squirrels were in a hyperfibrinolytic state with an elevated ratio of tissue plasminogen activator complexed with plasminogen activator inhibitor to total plasminogen activator inhibitor (p < 0.05). Histological markers of myocardial ischemia were reversibly elevated during hibernation with no increase in markers of myocardial cell death in the blood. These data suggest that ground squirrels do not form major blood clots during hibernation through suppression of coagulation and a hyperfibrinolytic state. These animals also demonstrate myocardial resistance to ischemia.
Collapse
Affiliation(s)
- Alison Bonis
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Leah Anderson
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Gaëlle Talhouarne
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Emily Schueller
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Jenna Unke
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Catherine Krus
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Jordan Stokka
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Anna Koepke
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Brittany Lehrer
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Anthony Schuh
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | | | - Scott Cooper
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA.
| |
Collapse
|
33
|
Wei Y, Zhang J, Xu S, Peng X, Yan X, Li X, Wang H, Chang H, Gao Y. Controllable oxidative stress and tissue specificity in major tissues during the torpor-arousal cycle in hibernating Daurian ground squirrels. Open Biol 2018; 8:rsob.180068. [PMID: 30305429 PMCID: PMC6223210 DOI: 10.1098/rsob.180068] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/31/2018] [Indexed: 01/28/2023] Open
Abstract
Mammalian hibernators experience repeated hypoxic ischaemia and reperfusion during the torpor–arousal cycle. We investigated levels of oxidative stress, antioxidant capacity, and the underlying mechanism in heart, liver, brain and kidney tissue as well as plasma during different periods of hibernation in Daurian ground squirrels (Spermophilus dauricus). Our data showed that the levels of hydrogen peroxide significantly increased in the heart and brain during late torpor (LT) compared with levels during the summer active (SA) state. The content of malondialdehyde (MDA) was significantly lower during interbout arousal (IBA) and early torpor (ET) than that during SA or pre-hibernation (PRE), and MDA levels in the LT brain were significantly higher than the levels in other states. Superoxide dismutase 2 protein levels increased markedly in the heart throughout the entire torpor–arousal cycle. Catalase expression remained at an elevated level in the liver during the hibernation cycle. Superoxide dismutase 1 and glutathione peroxidase 1 (GPx1) expression increased considerably in all tissues during the IBA and ET states. In addition, the activities of the various antioxidant enzymes were higher in all tissues during IBA and ET than during LT; however, GPx activity in plasma decreased significantly during the hibernation season. The expression of p-Nrf2 decreased in all tissue types during IBA, but significantly increased during LT, especially in liver tissue. Interestingly, most changed indicators recovered to SA or PRE levels in post-hibernation (POST). These results suggest that increased reactive oxygen species during LT may activate the Nrf2/Keap1 antioxidant pathway and may contribute to the decreased MDA levels found during the IBA and ET states, thereby protecting organisms from oxidative damage over the torpor-arousal cycle of hibernation. This is the first report on the remarkable controllability of oxidative stress and tissue specificity in major oxidative tissues of a hibernator.
Collapse
Affiliation(s)
- Yanhong Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China.,School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, People's Republic of China
| | - Jie Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| | - Shenhui Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| | - Xin Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| | - Xia Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| | - Xiaoyu Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| | - Huiping Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| | - Hui Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| | - Yunfang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi'an 710069, People's Republic of China
| |
Collapse
|
34
|
Jiang X, Gu T, Liu Y, Gao S, Shi E, Zhang G. Chipmunk Brain Is Resistant to Injury from Deep Hypothermic Circulatory Arrest During Cardiopulmonary Bypass. Ther Hypothermia Temp Manag 2018; 9:118-127. [PMID: 30036167 DOI: 10.1089/ther.2018.0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chipmunk as a food-storing hibernator naturally undergoes hibernation that is linked to great changes in systemic physiology and could protect the central nervous system during drastically reduced cerebral blood flow and low temperature in hibernation. Deep hypothermic circulatory arrest (DHCA) is associated with neurological dysfunction. We aim to test whether the euthermic chipmunk is resistant to injury from DHCA. Sprague-Dawley (SD) rats were used in a positive control. Ten euthermic chipmunks and 10 rats were subjected to 60-minute DHCA. Sham rats and chipmunks received cannulations. The blood samples after surgery were extracted to measure the tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) level. The levels of opioid receptor delta 1 (OPRD1), mature brain-derived neurotrophic factor (m-BDNF), precursor of BDNF (pro-BDNF), TrkB, GRB2, Erk, p-Erk, P38, Bcl-2, P75NTR, TRAF6, JNK, P53, Bax, and Caspase3 of the hippocampus were analyzed at 24 hours after surgery. The brain of chipmunks and rats were fixed for histopathological assessment. In the DHCA rat group, the levels of TNF-α and IL-6 were greater (p < 0.05) compared with DHCA chipmunks. In the DHCA chipmunk group, the levels of OPRD1, mature BDNF/pro-BDNF, TrkB-FL/TrkB-T1, Bcl-2, and p-Erk/Erk of hippocampus were higher than DHCA rats. The levels of GRB2, P75NTR, TRAF6, P53, Bax, and Caspase3 in DHCA chipmunks were lower than DHCA rats. The histopathological assessment showed that the injury in DHCA rat group was more severe than the DHCA chipmunk group. Euthermic chipmunks were greatly tolerant to global cerebral injury during DHCA. Different isoforms of BDNF might be involved in the resistant strategy.
Collapse
Affiliation(s)
- Xuan Jiang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Tianxiang Gu
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yu Liu
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Shilun Gao
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Enyi Shi
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Guangwei Zhang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
35
|
Dugbartey GJ, Bouma HR, Saha MN, Lobb I, Henning RH, Sener A. A Hibernation-Like State for Transplantable Organs: Is Hydrogen Sulfide Therapy the Future of Organ Preservation? Antioxid Redox Signal 2018; 28:1503-1515. [PMID: 28747071 DOI: 10.1089/ars.2017.7127] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SIGNIFICANCE Renal transplantation is the treatment of choice for end-stage renal disease, during which renal grafts from deceased donors are routinely cold stored to suppress metabolic demand and thereby limit ischemic injury. However, prolonged cold storage, followed by reperfusion, induces extensive tissue damage termed cold ischemia/reperfusion injury (IRI) and puts the graft at risk of both early and late rejection. Recent Advances: Deep hibernators constitute a natural model of coping with cold IRI as they regularly alternate between 4°C and 37°C. Recently, endogenous hydrogen sulfide (H2S), a gas with a characteristic rotten egg smell, has been implicated in organ protection in hibernation. CRITICAL ISSUES In renal transplantation, H2S also seems to confer cytoprotection by lowering metabolism, thereby creating a hibernation-like environment, and increasing preservation time while allowing cellular processes of preservation of homeostasis and tissue remodeling to take place, thus increasing renal graft survival. FUTURE DIRECTIONS Although the underlying cellular and molecular mechanisms of organ protection during hibernation have not been fully explored, mammalian hibernation may offer a great clinical promise to safely cold store and reperfuse donor organs. In this review, we first discuss mammalian hibernation as a natural model of cold organ preservation with reference to the kidney and highlight the involvement of H2S during hibernation. Next, we present recent developments on the protective effects and mechanisms of exogenous and endogenous H2S in preclinical models of transplant IRI and evaluate the potential of H2S therapy in organ preservation as great promise for renal transplant recipients in the future. Antioxid. Redox Signal. 28, 1503-1515.
Collapse
Affiliation(s)
- George J Dugbartey
- 1 Department of Medicine, Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,2 Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen , Groningen, Netherlands
| | - Hjalmar R Bouma
- 2 Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen , Groningen, Netherlands
| | - Manujendra N Saha
- 3 Matthew Mailing Center for Translational Transplant Studies, Western University , London, Canada .,4 Department of Surgery, Division of Urology, London Health Sciences Center, Western University , London, Canada .,5 Department of Microbiology and Immunology, London Health Sciences Center, Western University , London, Canada
| | - Ian Lobb
- 3 Matthew Mailing Center for Translational Transplant Studies, Western University , London, Canada
| | - Robert H Henning
- 2 Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen , Groningen, Netherlands
| | - Alp Sener
- 3 Matthew Mailing Center for Translational Transplant Studies, Western University , London, Canada .,4 Department of Surgery, Division of Urology, London Health Sciences Center, Western University , London, Canada .,5 Department of Microbiology and Immunology, London Health Sciences Center, Western University , London, Canada .,6 London Health Sciences Center, Western University , London, Canada
| |
Collapse
|
36
|
Effect of Sodium Thiosulfate Postconditioning on Ischemia-Reperfusion Injury Induced Mitochondrial Dysfunction in Rat Heart. J Cardiovasc Transl Res 2018; 11:246-258. [PMID: 29721767 DOI: 10.1007/s12265-018-9808-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/23/2018] [Indexed: 12/21/2022]
Abstract
The recent research on the therapeutic applications of sodium thiosulfate (STS) has gained importance in the treatment of cardiovascular diseases. Progressively through the present work, we have demonstrated that postconditioning of isolated rat heart subjected to ischemia-reperfusion injury using STS had preserved the mitochondrial structure, function, and number. Heart comprising of two mitochondrial subpopulations interfibrillar (IFM-involved in contractile function) and subsarcolemmal (SSM-involved in metabolic function), STS postconditioning imparted a state of hypometabolism to SSM, thereby reducing the metabolic demand of the reperfused heart. The IFM, on the other hand, provided the energy required to maintain contraction. Moreover, the hypometabolic state induced in SSM can lower the free radical release in addition to STS innate ability to act as an antioxidant and radical scavenger, all of which collectively provided cardioprotection. Therefore, drugs targeting IFM specifically or those reducing the energy demand for SSM can be suitable targets for myocardial ischemia-reperfusion injury.
Collapse
|
37
|
Viblanc VA, Schull Q, Roth JD, Rabdeau J, Saraux C, Uhlrich P, Criscuolo F, Dobson FS. Maternal oxidative stress and reproduction: Testing the constraint, cost and shielding hypotheses in a wild mammal. Funct Ecol 2018. [DOI: 10.1111/1365-2435.13032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | - Quentin Schull
- Université de Strasbourg CNRS IPHC UMR 7178 Strasbourg France
| | - Jeffrey D. Roth
- Department of Biological Sciences Auburn University Auburn AL USA
| | | | - Claire Saraux
- UMR MARBEC IFREMER (Institut Français de Recherche pour l'Exploitation de la Mer) Sète France
| | - Pierre Uhlrich
- Université de Strasbourg CNRS IPHC UMR 7178 Strasbourg France
| | | | | |
Collapse
|
38
|
Abstract
In some organisms and cells, oxygen availability influences oxygen consumption. In this review, we examine this phenomenon of hypoxic hypometabolism (HH), discussing its features, mechanisms, and implications. Small mammals and other vertebrate species exhibit "oxyconformism," a downregulation of metabolic rate and body temperature during hypoxia which is sensed by the central nervous system. Smaller body mass and cooler ambient temperature contribute to a high metabolic rate in mammals. It is this hypermetabolic state that is suppressed by hypoxia leading to HH. Larger mammals including humans do not exhibit HH. Tissues and cells also exhibit reductions in respiration during hypoxia in vitro, even at oxygen levels ample for mitochondrial oxidative phosphorylation. The mechanisms of cellular HH involve intracellular oxygen sensors including hypoxia-inducible factors, AMP-activated protein kinase (AMPK), and mitochondrial reactive oxygen species (ROS) which downregulate mitochondrial activity and ATP utilization. HH has a profound impact on cardiovascular, respiratory, and metabolic physiology in rodents. Therefore, caution should be exercised when extrapolating the results of rodent hypoxia studies to human physiology.
Collapse
|
39
|
Dugbartey GJ, Hardenberg MC, Kok WF, Boerema AS, Carey HV, Staples JF, Henning RH, Bouma HR. Renal Mitochondrial Response to Low Temperature in Non-Hibernating and Hibernating Species. Antioxid Redox Signal 2017; 27:599-617. [PMID: 28322600 DOI: 10.1089/ars.2016.6705] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SIGNIFICANCE Therapeutic hypothermia is commonly applied to limit ischemic injury in organ transplantation, during cardiac and brain surgery and after cardiopulmonary resuscitation. In these procedures, the kidneys are particularly at risk for ischemia/reperfusion injury (IRI), likely due to their high rate of metabolism. Although hypothermia mitigates ischemic kidney injury, it is not a panacea. Residual mitochondrial failure is believed to be a key event triggering loss of cellular homeostasis, and potentially cell death. Subsequent rewarming generates large amounts of reactive oxygen species that aggravate organ injury. Recent Advances: Hibernators are able to withstand periods of profoundly reduced metabolism and body temperature ("torpor"), interspersed by brief periods of rewarming ("arousal") without signs of organ injury. Specific adaptations allow maintenance of mitochondrial homeostasis, limit oxidative stress, and protect against cell death. These adaptations consist of active suppression of mitochondrial function and upregulation of anti-oxidant enzymes and anti-apoptotic pathways. CRITICAL ISSUES Unraveling the precise molecular mechanisms that allow hibernators to cycle through torpor and arousal without precipitating organ injury may translate into novel pharmacological approaches to limit IRI in patients. FUTURE DIRECTIONS Although the precise signaling routes involved in natural hibernation are not yet fully understood, torpor-like hypothermic states with increased resistance to ischemia/reperfusion can be induced pharmacologically by 5'-adenosine monophosphate (5'-AMP), adenosine, and hydrogen sulfide (H2S) in non-hibernators. In this review, we compare the molecular effects of hypothermia in non-hibernators with natural and pharmacologically induced torpor, to delineate how safe and reversible metabolic suppression may provide resistance to renal IRI. Antioxid. Redox Signal. 27, 599-617.
Collapse
Affiliation(s)
- George J Dugbartey
- 1 Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands .,2 Division of Cardiology, Cincinnati Children's Hospital Medical Center , Cincinnati, Ohio
| | - Maarten C Hardenberg
- 1 Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands
| | - Wendelinde F Kok
- 1 Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands
| | - Ate S Boerema
- 3 Groningen Institute for Evolutionary Life Sciences, University of Groningen , Groningen, the Netherlands .,4 Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands
| | - Hannah V Carey
- 5 Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin , Madison, Wisconsin
| | - James F Staples
- 6 Department of Biology, University of Western Ontario , London, Canada
| | - Robert H Henning
- 1 Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands
| | - Hjalmar R Bouma
- 1 Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands .,7 Department of Internal Medicine, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands
| |
Collapse
|
40
|
Munro D, Treberg JR. A radical shift in perspective: mitochondria as regulators of reactive oxygen species. ACTA ACUST UNITED AC 2017; 220:1170-1180. [PMID: 28356365 DOI: 10.1242/jeb.132142] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondria are widely recognized as a source of reactive oxygen species (ROS) in animal cells, where it is assumed that over-production of ROS leads to an overwhelmed antioxidant system and oxidative stress. In this Commentary, we describe a more nuanced model of mitochondrial ROS metabolism, where integration of ROS production with consumption by the mitochondrial antioxidant pathways may lead to the regulation of ROS levels. Superoxide and hydrogen peroxide (H2O2) are the main ROS formed by mitochondria. However, superoxide, a free radical, is converted to the non-radical, membrane-permeant H2O2; consequently, ROS may readily cross cellular compartments. By combining measurements of production and consumption of H2O2, it can be shown that isolated mitochondria can intrinsically approach a steady-state concentration of H2O2 in the medium. The central hypothesis here is that mitochondria regulate the concentration of H2O2 to a value set by the balance between production and consumption. In this context, the consumers of ROS are not simply a passive safeguard against oxidative stress; instead, they control the established steady-state concentration of H2O2 By considering the response of rat skeletal muscle mitochondria to high levels of ADP, we demonstrate that H2O2 production by mitochondria is far more sensitive to changes in mitochondrial energetics than is H2O2 consumption; this concept is further extended to evaluate how the muscle mitochondrial H2O2 balance should respond to changes in aerobic work load. We conclude by considering how differences in the ROS consumption pathways may lead to important distinctions amongst tissues, along with briefly examining implications for differing levels of activity, temperature change and metabolic depression.
Collapse
Affiliation(s)
- Daniel Munro
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada R3T 2N2.,Centre on Aging, University of Manitoba, Winnipeg, MB, Canada R3T 2N2
| | - Jason R Treberg
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada R3T 2N2 .,Centre on Aging, University of Manitoba, Winnipeg, MB, Canada R3T 2N2.,Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada R3T 2N2
| |
Collapse
|
41
|
Abstract
Although it is generally believed that oxidative phosphorylation and adequate oxygenation are essential for life, human development occurs in a profoundly hypoxic environment and "normal" levels of oxygen during embryogenesis are even harmful. The ability of embryos not only to survive but also to thrive in such an environment is made possible by adaptations related to metabolic pathways. Similarly, cancerous cells are able not only to survive but also to grow and spread in environments that would typically be fatal for healthy adult cells. Many biological states, both normal and pathological, share underlying similarities related to metabolism, the electron transport chain, and reactive species. The purpose of Part I of this review is to review the similarities among embryogenesis, mammalian adaptions to hypoxia (primarily driven by hypoxia-inducible factor-1), ischemia-reperfusion injury (and its relationship with reactive oxygen species), hibernation, diving animals, cancer, and sepsis, with a particular focus on the common characteristics that allow cells and organisms to survive in these states.
Collapse
Affiliation(s)
- Robert H Thiele
- From the Department of Anesthesiology, University of Virginia, Charlottesville, VA
| |
Collapse
|
42
|
Bhowmick S, Moore JT, Kirschner DL, Drew KL. Arctic ground squirrel hippocampus tolerates oxygen glucose deprivation independent of hibernation season even when not hibernating and after ATP depletion, acidosis, and glutamate efflux. J Neurochem 2017; 142:160-170. [PMID: 28222226 DOI: 10.1111/jnc.13996] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/16/2017] [Accepted: 02/08/2017] [Indexed: 12/20/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) triggers a cascade of uncontrolled cellular processes that perturb cell homeostasis. The arctic ground squirrel (AGS), a seasonal hibernator resists brain damage following cerebral I/R caused by cardiac arrest and resuscitation. However, it remains unclear if tolerance to I/R injury in AGS depends on the hibernation season. Moreover, it is also not clear if events such as depletion of ATP, acidosis, and glutamate efflux that are associated with anoxic depolarization are attenuated in AGS. Here, we employ a novel microperfusion technique to test the hypothesis that tolerance to I/R injury modeled in an acute hippocampal slice preparation in AGS is independent of the hibernation season and persists even after glutamate efflux. Acute hippocampal slices were harvested from summer euthermic AGS, hibernating AGS, and interbout euthermic AGS. Slices were subjected to oxygen glucose deprivation (OGD), an in vitro model of I/R injury to determine cell death marked by lactate dehydrogenase (LDH) release. ATP was assayed using ENLITEN ATP assay. Glutamate and aspartate efflux was measured using capillary electrophoresis. For acidosis, slices were subjected to pH 6.4 or ischemic shift solution (ISS). Acute hippocampal slices from rats were used as a positive control, susceptible to I/R injury. Our results indicate that when tissue temperature is maintained at 36°C, hibernation season has no influence on OGD-induced cell death in AGS hippocampal slices. Our data also show that tolerance to OGD in AGS hippocampal slices occurs despite loss of ATP and glutamate release, and persists during conditions that mimic acidosis and ionic shifts, characteristic of cerebral I/R. Read the Editorial Comment for this article on page 10.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, Alaska, USA.,Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - Jeanette T Moore
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - Daniel L Kirschner
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, Alaska, 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
|
43
|
Gorr TA. Hypometabolism as the ultimate defence in stress response: how the comparative approach helps understanding of medically relevant questions. Acta Physiol (Oxf) 2017; 219:409-440. [PMID: 27364602 DOI: 10.1111/apha.12747] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/28/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022]
Abstract
First conceptualized from breath-hold diving mammals, later recognized as the ultimate cell autonomous survival strategy in anoxia-tolerant vertebrates and burrowing or hibernating rodents, hypometabolism is typically recruited by resilient organisms to withstand and recover from otherwise life-threatening hazards. Through the coordinated down-regulation of biosynthetic, proliferative and electrogenic expenditures at times when little ATP can be generated, a metabolism turned 'down to the pilot light' allows the re-balancing of energy demand with supply at a greatly suppressed level in response to noxious exogenous stimuli or seasonal endogenous cues. A unifying hallmark of stress-tolerant organisms, the adaptation effectively prevents lethal depletion of ATP, thus delineating a marked contrast with susceptible species. Along with disengaged macromolecular syntheses, attenuated transmembrane ion shuttling and PO2 -conforming respiration rates, the metabolic slowdown in tolerant species usually culminates in a non-cycling, quiescent phenotype. However, such a reprogramming also occurs in leading human pathophysiologies. Ranging from microbial infections through ischaemia-driven infarcts to solid malignancies, cells involved in these disorders may again invoke hypometabolism to endure conditions non-permissive for growth. At the same time, their reduced activities underlie the frequent development of a general resistance to therapeutic interventions. On the other hand, a controlled induction of hypometabolic and/or hypothermic states by pharmacological means has recently stimulated intense research aimed at improved organ preservation and patient survival in situations requiring acutely administered critical care. The current review article therefore presents an up-to-date survey of concepts and applications of a coordinated and reversibly down-regulated metabolic rate as the ultimate defence in stress responses.
Collapse
Affiliation(s)
- T. A. Gorr
- Institute of Veterinary Physiology; Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| |
Collapse
|
44
|
Otis JP, Pike AC, Torrealba JR, Carey HV. Hibernation reduces cellular damage caused by warm hepatic ischemia-reperfusion in ground squirrels. J Comp Physiol B 2017; 187:639-648. [PMID: 28144740 DOI: 10.1007/s00360-017-1056-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/06/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022]
Abstract
During the hibernation season, livers from 13-lined ground squirrels (Ictidomys tridecemlineatus) are resistant to damage induced by ex vivo, cold ischemia-warm reperfusion (IR) compared with livers from summer squirrels or rats. Here, we tested the hypothesis that hibernation also reduces damage to ground squirrel livers in an in vivo, warm IR model, which more closely resembles complications associated with traumatic injury or surgical interventions. We also examined whether protection is mediated by two metabolites, inosine and biliverdin, that are elevated in ground squirrel liver during interbout arousals. Active squirrels in spring and hibernators during natural arousals to euthermia (body temperature 37 °C) were subject to liver IR or sham treatments. A subset of hibernating squirrels was pre-treated with compounds that inhibit inosine synthesis/signaling or biliverdin production. This model of liver IR successfully induced hepatocellular damage as indicated by increased plasma liver enzymes (ALT, AST) and hepatocyte apoptosis index compared to sham in both seasons, with greater elevations in spring squirrels. In addition, liver congestion increased after IR to a similar degree in spring and hibernating groups. Microvesicular steatosis was not affected by IR within the same season but was greater in sham squirrels in both seasons. Plasma IL-6 increased ~twofold in hibernators pre-treated with a biliverdin synthesis inhibitor (SnPP) prior to IR, but was not altered by IR in untreated squirrels. The results show that hibernation provides protection to ground squirrel livers subject to warm IR. Further research is needed to clarify mechanisms responsible for endogenous protection of liver tissue under ischemic stress.
Collapse
Affiliation(s)
- Jessica P Otis
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA
| | - Amanda C Pike
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA
| | - Jose R Torrealba
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hannah V Carey
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| |
Collapse
|
45
|
Glossmann HH, Lutz OMD. Torpor: The Rise and Fall of 3-Monoiodothyronamine from Brain to Gut-From Gut to Brain? Front Endocrinol (Lausanne) 2017; 8:118. [PMID: 28620354 PMCID: PMC5450037 DOI: 10.3389/fendo.2017.00118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022] Open
Abstract
3-Monoiodothyronamine (T1AM), first isolated from rat brain, is reported to be an endogenous, rapidly acting metabolite of thyroxine. One of its numerous effects is the induction of a "torpor-like" state in experimental animals. A critical analysis of T1AM, to serve as an endogenous cryogen, is given. The proposed biosynthetic pathway for formation of T1AM, which includes deiodinases and ornithine decarboxylase in the upper intestinum, is an unusual one. To reach the brain via systemic circulation, enterohepatic recycling and passage through the liver may occur. The possible role of gut microbiota is discussed. T1AM concentrations in human serum, measured by a specific monoclonal assay are up to three orders of magnitude higher compared to values obtained by MS/MS technology. The difference is explained by the presence of a high-affinity binder for T1AM (Apolipoprotein B-100) in serum, which permits the immunoassay to measure the total concentration of the analyte but limits MS/MS technology to detect only the unbound (free) analyte, a view, which is contested here.
Collapse
Affiliation(s)
- Hartmut H. Glossmann
- Institut für Biochemische Pharmakologie, Innsbruck, Austria
- *Correspondence: Hartmut H. Glossmann,
| | | |
Collapse
|
46
|
Cooper ST, Sell SS, Fahrenkrog M, Wilkinson K, Howard DR, Bergen H, Cruz E, Cash SE, Andrews MT, Hampton M. Effects of hibernation on bone marrow transcriptome in thirteen-lined ground squirrels. Physiol Genomics 2016; 48:513-25. [PMID: 27207617 PMCID: PMC4967218 DOI: 10.1152/physiolgenomics.00120.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Mammalian hibernators adapt to prolonged periods of immobility, hypometabolism, hypothermia, and oxidative stress, each capable of reducing bone marrow activity. In this study bone marrow transcriptomes were compared among thirteen-lined ground squirrels collected in July, winter torpor, and winter interbout arousal (IBA). The results were consistent with a suppression of acquired immune responses, and a shift to innate immune responses during hibernation through higher complement expression. Consistent with the increase in adipocytes found in bone marrow of hibernators, expression of genes associated with white adipose tissue are higher during hibernation. Genes that should strengthen the bone by increasing extracellular matrix were higher during hibernation, especially the collagen genes. Finally, expression of heat shock proteins were lower, and cold-response genes were higher, during hibernation. No differential expression of hematopoietic genes involved in erythrocyte or megakaryocyte production was observed. This global view of the changes in the bone marrow transcriptome over both short term (torpor vs. IBA) and long term (torpor vs. July) hypothermia can explain several observations made about circulating blood cells and the structure and strength of the bone during hibernation.
Collapse
Affiliation(s)
- Scott T Cooper
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin;
| | - Shawn S Sell
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Molly Fahrenkrog
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Kory Wilkinson
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - David R Howard
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Hannah Bergen
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Estefania Cruz
- Biology Department, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Steve E Cash
- Hematology/Oncology, Gundersen Lutheran Medical Foundation, La Crosse, Wisconsin
| | - Matthew T Andrews
- Department of Biology, University of Minnesota-Duluth, Duluth, Minnesota; and
| | - Marshall Hampton
- Department of Mathematics and Statistics, University of Minnesota-Duluth, Duluth, Minnesota
| |
Collapse
|
47
|
Yin Q, Ge H, Liao CC, Liu D, Zhang S, Pan YH. Antioxidant Defenses in the Brains of Bats during Hibernation. PLoS One 2016; 11:e0152135. [PMID: 27010916 PMCID: PMC4806925 DOI: 10.1371/journal.pone.0152135] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/09/2016] [Indexed: 01/12/2023] Open
Abstract
Hibernation is a strategy used by some mammals to survive a cold winter. Small hibernating mammals, such as squirrels and hamsters, use species- and tissue-specific antioxidant defenses to cope with oxidative insults during hibernation. Little is known about antioxidant responses and their regulatory mechanisms in hibernating bats. We found that the total level of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in the brain of each of the two distantly related hibernating bats M. ricketti and R. ferrumequinum at arousal was lower than that at torpid or active state. We also found that the levels of malondialdehyde (product of lipid peroxidation) of the two hibernating species of bats were significantly lower than those of non-hibernating bats R. leschenaultia and C. sphinx. This observation suggests that bats maintain a basal level of ROS/RNS that does no harm to the brain during hibernation. Results of Western blotting showed that hibernating bats expressed higher amounts of antioxidant proteins than non-hibernating bats and that M. ricketti bats upregulated the expression of some enzymes to overcome oxidative stresses, such as superoxide dismutase, glutathione reductase, and catalase. In contrast, R. ferrumequinum bats maintained a relatively high level of superoxide dismutase 2, glutathione reductase, and thioredoxin-2 throughout the three different states of hibernation cycles. The levels of glutathione (GSH) were higher in M. ricketti bats than in R. ferrumequinum bats and were significantly elevated in R. ferrumequinum bats after torpor. These data suggest that M. ricketti bats use mainly antioxidant enzymes and R. ferrumequinum bats rely on both enzymes and low molecular weight antioxidants (e.g., glutathione) to avoid oxidative stresses during arousal. Furthermore, Nrf2 and FOXOs play major roles in the regulation of antioxidant defenses in the brains of bats during hibernation. Our study revealed strategies used by bats against oxidative insults during hibernation.
Collapse
Affiliation(s)
- Qiuyuan Yin
- Laboratory of Molecular Ecology and Evolution, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai, China
| | - Hanxiao Ge
- Laboratory of Molecular Ecology and Evolution, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai, China
| | - Chen-Chong Liao
- Proteomic Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Di Liu
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, China
| | - Shuyi Zhang
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, China
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
- * E-mail: (YP); (SZ)
| | - Yi-Hsuan Pan
- Laboratory of Molecular Ecology and Evolution, School of Life Sciences, East China Normal University, Shanghai, China
- * E-mail: (YP); (SZ)
| |
Collapse
|
48
|
Abstract
Many environmental conditions can constrain the ability of animals to obtain sufficient food energy, or transform that food energy into useful chemical forms. To survive extended periods under such conditions animals must suppress metabolic rate to conserve energy, water, or oxygen. Amongst small endotherms, this metabolic suppression is accompanied by and, in some cases, facilitated by a decrease in core body temperature-hibernation or daily torpor-though significant metabolic suppression can be achieved even with only modest cooling. Within some ectotherms, winter metabolic suppression exceeds the passive effects of cooling. During dry seasons, estivating ectotherms can reduce metabolism without changes in body temperature, conserving energy reserves, and reducing gas exchange and its inevitable loss of water vapor. This overview explores the similarities and differences of metabolic suppression among these states within adult animals (excluding developmental diapause), and integrates levels of organization from the whole animal to the genome, where possible. Several similarities among these states are highlighted, including patterns and regulation of metabolic balance, fuel use, and mitochondrial metabolism. Differences among models are also apparent, particularly in whether the metabolic suppression is intrinsic to the tissue or depends on the whole-animal response. While in these hypometabolic states, tissues from many animals are tolerant of hypoxia/anoxia, ischemia/reperfusion, and disuse. These natural models may, therefore, serve as valuable and instructive models for biomedical research.
Collapse
Affiliation(s)
- James F Staples
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
49
|
Drew KL, Wells M, McGee R, Ross AP, Kelleher-Andersson J. Arctic ground squirrel neuronal progenitor cells resist oxygen and glucose deprivation-induced death. World J Biol Chem 2016; 7:168-177. [PMID: 26981205 PMCID: PMC4768121 DOI: 10.4331/wjbc.v7.i1.168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 11/26/2015] [Accepted: 01/11/2016] [Indexed: 02/05/2023] Open
Abstract
AIM: To investigate the influence of ischemia/reperfusion on arctic ground squirrel (AGS) neuronal progenitor cells (NPCs), we subjected these cultured cells to oxygen and glucose deprivation.
METHODS: AGS NPCs were expanded and differentiated into NPCs and as an ischemia vulnerable control, commercially available human NPCs (hNPCs) were seeded from thawed NPCs. NPCs, identified by expression of TUJ1 were seen at 14-21 d in vitro (DIV). Cultures were exposed to control conditions, hypoxia, oxygen and glucose deprivation or glucose deprivation alone or following return to normal conditions to model reperfusion. Cell viability and death were assessed from loss of ATP as well as from measures of alamarBlue® and lactate dehydrogenase in the media and from counts of TUJ1 positive cells using immunocytochemistry. Dividing cells were identified by expression of Ki67 and phenotyped by double labeling with GFAP, MAP2ab or TUJ1.
RESULTS: We report that when cultured in NeuraLife™, AGS cells remain viable out to 21 DIV, continue to express TUJ1 and begin to express MAP2ab. Viability of hNPCs assessed by fluorescence alamarBlue (arbitrary units) depends on both glucose and oxygen availability [viability of hNPCs after 24 h oxygen glucose deprivation (OGD) with return of oxygen and glucose decreased from 48151 ± 4551 in control cultures to 43481 ± 2413 after OGD, P < 0.05]. By contrast, when AGS NPCs are exposed to the same OGD with reperfusion at 14 DIV, cell viability assessed by alamarBlue increased from 165305 ± 11719 in control cultures to 196054 ± 13977 after OGD. Likewise AGS NPCs recovered ATP (92766 ± 6089 in control and 92907 ± 4290 after modeled reperfusion; arbitrary luminescence units), and doubled in the ratio of TUJ1 expressing neurons to total dividing cells (0.11 ± 0.04 in control cultures vs 0.22 ± 0.2 after modeled reperfusion, P < 0.05). Maintaining AGS NPCs for a longer time in culture lowered resistance to injury, however, did not impair proliferation of NPCs relative to other cell lineages after oxygen deprivation followed by re-oxygenation.
CONCLUSION: Ischemic-like insults decrease viability and increase cell death in cultures of human NPCs. Similar conditions have less affect on cell death and promote proliferation in AGS NPCs.
Collapse
|
50
|
Multifactorial processes to slowing the biological clock: Insights from a comparative approach. Exp Gerontol 2015; 71:27-37. [DOI: 10.1016/j.exger.2015.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/20/2015] [Accepted: 08/29/2015] [Indexed: 02/07/2023]
|