<<

The Mechanism Enabling in 3

Yuuki Horii, Takahiko Shiina, and Yasutake Shimizu

Abstract (CIRP) in the hearts of hibernating . Some including squirrels and ham- The CIRP mRNA is constitutively expressed sters undergo hibernation. During hibernation, in the heart of a nonhibernating euthermic body temperature drops to only a few degrees with several different forms probably above ambient temperature. The suppression due to alternative splicing. The short product of whole-body energy expenditure is associ- contained the complete open reading frame ated with regulated, but not passive, reduction for full-length CIRP, while the long product of cellular . The heart retains the had inserted sequences containing a stop ability to beat constantly, although body tem- codon, suggesting production of a C-terminal perature drops to less than 10 °C during hiber- deletion isoform of CIRP. In contrast to nonhi- nation. Cardiac myocytes of hibernating bernating hamsters, only the short product mammals are characterized by reduced Ca2+ was found in hibernating animals. Thus, these entry into the cell membrane and a concomi- results indicate that CIRP expression in the tant enhancement of Ca2+ release from and hamster heart is regulated at the level of alter- reuptake by the sarcoplasmic reticulum. These native splicing, which would permit a rapid adaptive changes would help in preventing increment of functional CIRP when entering excessive Ca2+ entry and its overload and in hibernation. We will summarize the current maintaining the resting levels of intracellular understanding of the cold-resistant property of Ca2+. Adaptive changes in gene expression in the heart in hibernating animals. the heart prior to hibernation may be indis- pensable for acquiring cold resistance. In Keywords addition, protective effects of cold-shock pro- Hibernation · Cold-shock protein · teins are thought to have an important role. We recently reported the unique expression pat- tern of cold-inducible RNA-binding protein Abbreviations

Y. Horii · T. Shiina · Y. Shimizu (*) CIRP Cold-inducing RNA-binding protein Department of Basic Veterinary Science, Laboratory CNS The central nervous system of Physiology, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan ECG Electrocardiograms e-mail: [email protected] HNF Hepatocyte nuclear factor

© Springer Nature Singapore Pte Ltd. 2018 45 M. Iwaya-Inoue et al. (eds.), Survival Strategies in Extreme Cold and Desiccation, Advances in Experimental Medicine and Biology 1081, https://doi.org/10.1007/978-981-13-1244-1_3 46 Y. Horii et al.

HP Hibernation-specific protein Table 3.1 Hibernating mammals ICV Intracerebroventricular Body RBM3 RNA-binding motif 3 temperature in SERCA2 asarco(endo)plasmic reticulum Ca2+- hibernation Order Species (°C) ATPase 2a Monotremata Echidna 4 Marsupialia Pygmy possum, 1.3–7.1 feathertail glider, Chiloe opossum 3.1 Hibernation of Mammals Eulipotyphla 1–9.7 Afrosoricida Tenrec 8.6–15 Chiroptera Most mammals have the ability to maintain their −2 to 13.9 6.5–9.3 body temperature within a narrow range even in a Carnivora Badger, 28–32.5 cold environment. In a cold environment, ther- Rodentia , , −2.9 to 15 moregulatory responses to minimize heat loss woodchuck, ground (e.g., peripheral vasoconstriction and piloerec- squirrel, , tion) are evoked (Tansey and Johnson 2015). In pocket mouse, kangaroo mouse, addition, heat-producing responses in skeletal hamster, jerboa, muscles (shivering thermogenesis) and in brown dormouse adipose tissue (non-shivering or metabolic ther- Ruf and Geiser (2015) mogenesis) are activated, and thereby a drop in body temperature is prevented (Tansey and Johnson 2015). If the body temperature of homo- contrast, mammalian hibernators possess a ther- therms drops extremely, they cannot survive moregulatory mechanism similar to that of non- because the heart cannot keep beating, and organs hibernators, and they can control their body fall into ischemia (Ivanov 2000). temperature in a nonhibernating state despite On the other hand, several mammalian species exposure to a wide range of surrounding temper- undergo hibernation (Carey et al. 2003; Ruf and atures (Carey et al. 2003; Ruf and Geiser 2015). Geiser 2015) (see Table 3.1). During hibernation, Even in an extremely cold environment, they do body temperature drops to only a few degrees not necessarily undergo hibernation if enough above ambient temperature (Carey et al. 2003; food is available. Furthermore, mammalian Ruf and Geiser 2015). Hibernating animals stay hibernators do not always continue in a hibernat- unmoving and usually show a curly shape to min- ing condition throughout winter; they sometimes imize heat dissipation from the body surface interrupt hibernation and spontaneously recover (Fig. 3.1). The hypothermic condition of mam- their body temperature even though they are con- malian hibernators is fundamentally different sistently exposed to a cold environment (Carey from that of poikilotherms ( and rep- et al. 2003). This behavior provides further evi- tiles). The body temperature of poikilothermic dence for the notion that hypothermia during animals directly correlates with changes in ambi- mammalian hibernation is actively induced, since ent temperature due to a lack of efficacious passively induced hypothermia may not recover mechanisms for maintaining body temperature unless ambient temperature is increased. Thus, (Jackson and Ultsch 2010; Malan 2014). As a hibernation of mammals is considered to be an result, body temperature drops passively in adaptive strategy to survive in a severe environ- response to a decrease in ambient temperature. In ment during winter. 3 The Mechanism Enabling Hibernation in Mammals 47

Fig. 3.1 Hibernating hamster. Pictures show curly shape that is usually observed during hibernation in Syrian hamsters

Fig. 3.2 A schema of body temperature during hibernation

3.2 Variation of Hibernation (Elephantulus myurus) reached 5–10 °C during daily (Mzilikazi et al. 2002). Some species During hibernation, the degree of body tempera- including tenrec and mouse adopt either ture reduction and duration of the hypothermic daily torpor or hibernation depending on the state vary widely among animal species (Carey ambient temperature (Lovegrove and Génin et al. 2003; Ruf and Geiser 2015). In black , 2008; Kobbe and Dausmann 2009; Kobbe et al. the body temperature during hibernation is 2011). A typical deep hibernation is character- around 33 °C, which is much higher than that in ized by extended duration of torpor bouts. As small hibernators (Carey et al. 2003; Ruf and shown in Fig. 3.2, the hypothermic state during Geiser 2015). In contrast, the body temperature deep hibernation is interrupted by periods of of arctic ground squirrels drops to as low as arousals to euthermia, so-called interbout arous- −3 °C during hibernation (Barnes 1989). Several als. The duration of torpor bouts is from a few mammalian species undergo daily torpor, in days to up to 5 weeks. The interbout arousals are which duration of the hypothermic state is less maintained for 12–24 h before reentry into torpor than 24 h (Breukelen and Martin 2015). In daily (Carey et al. 2003). The periodic hibernation-­ torpor, reduction of body temperature is rela- arousal cycles suggest that the central nervous tively moderate compared with that in deep system (CNS) is continuously operated even at a hibernation. Exceptionally, it has been reported low temperature during hibernation. that body temperature of the rock elephant 48 Y. Horii et al.

Table 3.2 Physiological parameters in active and hiber- metabolic rate of the hibernating nating hamsters (n = 6) is reduced to less than 5% of that observed in the Active Hibernation Hibernation nonhibernating euthermic counterpart (Wang and control in summer in winter Lee 1996). The suppression of whole-body Body 35.2 ± 0.6 5.0 ± 0.9 5.5 ± 0.3 temperature energy expenditure is associated with regulated, (°C) but not passive, reduction of cellular metabolism. Heart rate 369 ± 13 15.8 ± 3.1 15.0 ± 2.7 It has been demonstrated that a serine/threonine (beats/min) protein kinase, Akt (also known as protein kinase Respiratory 92.2 ± 8.5 2.3 ± 1.7 3.0 ± 1.4 B), is inactivated by dephosphorylation in hiber- rate (breaths/ min) nating animal organs, typically in skeletal mus- cles and the liver (Abnous et al. 2008). Considering that Akt activation plays an impor- It is known that seasonal hibernators, e.g., tant role in anabolic and catabolic responses in Richardson’s ground squirrel ( various cells, the dephosphorylation of Akt in richardsonii) and Siberian chipmunk (Tamias hibernating animal cells would be suitable for a sibiricus asiatics), rarely hibernate in summer decrease in metabolic activity. Interestingly, the even if they are placed in a cold condition (Kondo dephosphorylation is promoted immediately 1987). This suggests that the endogenous circan- prior to entering hibernation (Hoehn et al. 2004). nual rhythm plays a critical role in the induction Accordingly, the reduction of cellular metabo- of hibernation in seasonal hibernators. In con- lism during hibernation does not arise as a conse- trast, hamsters hibernated even in summer when quence of lowered temperature (i.e., general they were placed in a condition suitable for suppression of enzyme activity). Rather, cellular induction of hibernation (Miyazawa et al. 2008). metabolism is suppressed actively before enter- No significant differences in parameters includ- ing hibernation, and this can therefore be a cause ing body temperature, heart rate, respiratory rate of decrease in body temperature. Consistent with (Table 3.2), and incidence of ECG abnormalities this, Akt activity is increased during arousal from were found between hibernation in summer and hibernation (Lee et al. 2002; Fleck and Carey that in winter (Miyazawa et al. 2008). Therefore, 2005). the endogenous circannual rhythm might only Some species do not feed during hibernation, have a minor contribution, if any, to the induction whereas other species store food and feed during of hibernation in this species. Of course, this does interbout arousals (Humphries et al. 2003; Geiser not necessarily rule out the possibility of involve- 2004). Regardless of these differences, hiberna- ment of the circannual rhythm in the induction of tion in both groups of species can be considered hibernation in hamsters. It may be appropriate to as a fasting condition (Humphries et al. 2003). To consider that the relative importance of endoge- tolerate the long-term fasting condition, major nous and environmental factors varies among metabolic substrate switches from glucose to species and that this variation is a determinant for lipid occur during the hibernation period in seasonal or nonseasonal hibernators. ground squirrels and black bears (Serkova et al. 2007; Andrews et al. 2009) as evidenced by the fact that respiratory quotient values are about 3.3 Metabolic Regulation 0.7 in hibernating animals (Fedorov et al. 2009). During Hibernation Global analysis of gene expression by using DNA microarrays would allow speculation It is generally accepted that the primary purpose regarding differences in metabolic conditions of hibernation is to decrease metabolic activity, between hibernating hypothermic animals and allowing energy expenditure to be balanced with active euthermic animals (Williams et al. 2005). reduced energy supply due to limited food avail- In the liver of hibernating bears, expression levels ability during the winter season. For instance, the of key glucogenic enzymes are increased, 3 The Mechanism Enabling Hibernation in Mammals 49 whereas expression levels of glycolytic enzymes role in the entrance phase of hibernation in ham- are decreased (Fedorov et al. 2009). A similar sters (Tamura et al. 2005). The importance of shift from glycolysis to gluconeogenesis was central adenosine is suggested by the fact that observed at the mRNA and protein levels in the intracerebroventricular (ICV) injection of an ade- liver of hibernating ground squirrels (Yan et al. nosine A1-receptor antagonist to hamsters in the 2008). These changes would contribute to the process of dropping body temperature inhibits provision of glucose as an energy source for the entrance to hibernation (Tamura et al. 2005). The brain and other tissues in fasting conditions dur- effect of adenosine would be related to hiberna- ing hibernation. Also, genes involved in cellular tion onset but not to maintenance of a hypother- respiration are downregulated during hibernation mic condition, because decreased body (Williams et al. 2005; Yan et al. 2008; Fedorov temperature cannot be reversed in animals in et al. 2009). This is consistent with the reduced which deep hypothermia has already been estab- metabolic rate in hibernating animals (Carey lished (Tamura et al. 2005). Vice versa, ICV et al. 2003). Reduction of gene expression for injection of an A1-receptor agonist to euthermic anabolic enzymes with concomitant induction of hamsters decreases body temperature (Miyazawa gene expression for catabolic enzymes is also the et al. 2008). In the CNS, adenosine acts as a neu- case in lipid metabolism. A coordinated induc- romodulator, and the A1-receptor mediates the tion of genes involved in fatty acid β-oxidation presynaptic inhibition of neurotransmission. and downregulation of genes involved in lipid Thus, activation of the A1-receptor would act as biosynthesis at transcriptional (Williams et al. a suppressor of the thermoregulatory mechanism 2005; Yan et al. 2008) and proteomic levels (Shao in the CNS. In accordance with this, it has been et al. 2010) have been shown in the livers of reported that activation of the A1-receptor pro- hibernating bears and ground squirrels. In con- motes sedation and depression of locomotor trast, genes involved in amino acid catabolism activity (Radulovacki et al. 1982; Wauquier et al. are downregulated during hibernation (Fedorov 1987; Nikodijevic et al. 1991; Ticho and et al. 2009). Reduction of amino acid breakdown Radulovacki 1991; Malhotra and Gupta 1997). would be reasonable, since genes involved in Similar approaches to identify possible regu- protein biosynthesis in the liver and skeletal mus- lators of hibernation have revealed that opioid cles are increased in this state (Fedorov et al. peptides such as β-endorphin and endomorphine 2009). The enhanced protein biosynthesis is con- in the hypothalamus are related to the mainte- sidered to be a molecular adaptation that contrib- nance phase via the μ1-opioid receptor in ham- utes to the ability to reduce muscle atrophy over sters (Tamura et al. 2005) and via the δ-opioid prolonged periods of immobility during receptor in ground squirrels (Yu and Cai 1993a, hibernation. b). The contribution of opioid peptides to mainte- nance of the hypothermic state leads an interest- ing hypothesis that continuous release of opioid 3.4 Endogenous Regulators peptides during hypothermia may induce toler- of Hibernation ance, and therefore hamsters cannot maintain hypothermia for a long time (Tamura et al. 2005). 3.4.1 Factors Related Although further study is needed to verify this to Hibernation hypothesis, it provides a rational explanation for the presence of energy-demanding interbout Although the precise mechanism responsible for arousals. regulating hibernation remains unknown, a num- Several lines of evidence suggest that hista- ber of studies have revealed important factors mine in the hippocampus is involved in the main- controlling hibernation behavior. It has been tenance of hibernation. In general, histamine demonstrated that adenosine acting through the decreases sleep and promotes wakefulness adenosine A1-receptor in the CNS plays a key (Nishino et al. 2001). However, infusion of hista- 50 Y. Horii et al. mine into the dorsal hippocampus brings about 1992; Takamatsu et al. 1993). HP-20, HP-25, and prolonged duration of the torpor bout. This find- HP-27 contain an N-terminal collagen-like ing is also interesting since it supports the idea domain and a C-terminal globular domain homol- that hibernation is an arousal state distinct from ogous to the complement C1q (Takamatsu et al. any known euthermic arousal state, rather than 1993). The proteins can be detected in the plasma being homologous to sleep (Kilduff et al. 1993). of hibernators, but not in nonhibernators, includ- The preferential use of lipids during hiberna- ing tree squirrels and rats (Kondo and Kondo tion seems to suggest that excessive fat accumu- 1992; Takamatsu et al. 1993). The lack of HP in lation is appropriate for entering hibernation. tree squirrels (Callosciurus caniceps) is interest- However, it has been demonstrated that a high ing because tree squirrels are a species closely body mass inhibits the induction of hibernation related to but do not undergo hiberna- (Bieber et al. 2014; Zervanos et al. 2014). tion (Kojima et al. 2001). This provides support Conversely, a reduction of body mass triggers the for the pivotal role of HP in hibernation. entrance to hibernation in order to reduce the The plasma level of the HP complex decreases consumption of limited amounts of stored fat. markedly in hibernating chipmunks (Kondo and Thus, the decision of whether or not to enter Kondo 1992; Takamatsu et al. 1993). hibernation depends on the body mass and Concomitantly, HP gene expression in the liver, amount of fat deposits (Humphries et al. 2003; in which HP is exclusively produced, is down- Chayama et al. 2016). One possible hormonal regulated (Takamatsu et al. 1993). However, mediator that reflects the amount of fat deposits reduction of the circulating HP complex level is leptin (Houseknecht et al. 1998). In line with would not be totally dependent on reduced pro- this, a high circulating level of leptin negatively duction in the liver. In contrast to the plasma level impacts the induction of hibernation. In little of the HP complex, the level of a heterotrimer brown , dissociation of leptin secretion and composed of HP-20, HP-25, and HP-27 (called adiposity is found during the pre-hibernation HP20c) is increased in the brain (Kondo et al. period, and the decreased leptin level in the 2006). Therefore, transport to the brain is attrib- absence of a decrease in body mass permits the utable to the reduced circulating level of the HP entrance to hibernation (Kronfeld-Schor et al. complex. The currently accepted mechanism for 2000). Accordingly, leptin can be considered to activation of HP involves dissociation of the HP be an important regulator of hibernation. complex to HP20c and HP-55. HP20c, being free from HP-55, can be actively transported to the brain, where it regulates brain functions for 3.4.2 Hibernation-Specific Protein hibernation. In support of this model, a neutral- izing antibody against HP20c decreases the dura- Many studies have been conducted to identify tion of hibernation (Kondo et al. 2006). factors responsible for hibernation (Wang et al. Furthermore, hibernation is never induced in ani- 1988; Shintani et al. 2005; Tamura et al. 2005, mals lacking an increase in HP20c even in a cold 2006, 2012; Kondo 2007; Chayama et al. 2016). environment (Kondo et al. 2006). The precise The most typical factors that may play a role in action of HP20c in regulation of hibernation physiological adaptation prior to the onset of remains to be elucidated. hibernation are hibernation-specific proteins Interestingly, HP gene expression levels in the (HP), originally discovered in the chipmunk liver, as well as plasma HP levels, show seasonal (Tamias asiaticus) in 1992 (Kondo and Kondo oscillations even when chipmunks are kept under 1992). The protein identified in the plasma of the a warm condition with a 12-h photoperiod chipmunk is a 140-kDa protein complex that con- (Kondo et al. 2006). This indicates that gene sists of four components: three highly homolo- expression of HP is regulated by endogenous gous proteins (HP-20, HP-25 and HP-27) and a circannual rhythms, rather than environmental proteinase inhibitor (HP-55) (Kondo and Kondo factors (Kondo et al. 2006). It has been demon- 3 The Mechanism Enabling Hibernation in Mammals 51 strated that hepatocyte nuclear factor 4 (HNF-4) the degree of Ca2+ increment even if the ampli- activates HP-25 transcription (Kojima et al. tude of the cytosolic Ca2+ transient remains simi- 2000). In nonhibernating chipmunks, HNF-4 lar. It is therefore considered that a rise in the binds to the HP-25 promoter, leading to HP-25 basal Ca2+ concentration is an underlying basis transcriptional activation. On the other hand, for cardiac dysfunction at a cold temperature. small heterodimer partner (SHP), which is a Interestingly, in the ground squirrel, a typical ­negative regulator of HNF-4, is upregulated in hibernator, it has been reported that the basal the liver of hibernating chipmunks, resulting in intracellular Ca2+ concentration of myocardia is the dissociation of HNF-4 from the HP-25 pro- not increased at a cold temperature (Wang et al. moter and the repression of HP-25 gene tran- 2002). In addition, amplitude of the Ca2+ tran- scription (Tsukamoto et al. 2017). Accordingly, sient is increased at a cold temperature (Wang SHP, which controls HNF-4 binding to the HP- et al. 2002). In agreement with this, myocardial 25 gene promoter, would be one of the key regu- contractile force at a low temperature is greater lators of HP gene expression. than that at a temperature comparable to body temperature of the active state. The greater myo- cardial contractile force at a low temperature 3.5 Regulation of Cardiac would be reasonable as a compensatory mecha- Function During Hibernation nism for the marked reduction of heart rate. The remarkable differences in Ca2+ dynamics between 3.5.1 Innate Characteristics hibernators and nonhibernators suggest that an of the Heart of Hibernators ability to maintain cardiac contractility under an extremely hypothermic condition can be recog- Although heart rate in hibernating animals is dra- nized as an inherent feature of hibernators. matically lowered compared with that in euther- The cold-resistant nature of the heart of a mic counterparts, normal sinus rhythm is hibernator has also become apparent from exper- fundamentally maintained (Harris and Milsom iments in which attempts were made to induce 1995; Milsom et al. 1999; Mertens et al. 2008). artificial hypothermia in both hibernators and This is in contrast to nonhibernating mammals, in nonhibernators. When extreme hypothermia was which ventricular dysfunction and arrhythmias forcibly induced by pentobarbital anesthesia such as atrioventricular block and ventricular combined with cooling of the whole body, car- fibrillation develop when their body temperature diac contractility was maintained in hamsters drops to less than 20 °C (Johansson 1996; (Miyazawa et al. 2008). This is in sharp contrast Fedorov et al. 2008). Contraction of cardiac mus- to nonhibernators, in which cardiac arrest is usu- cle, analogous to that of , is ally induced at a low temperature (Duker et al. induced by intracellular Ca2+ transients (Kurihara 1983; Caprette and Senturia 1984; Johansson 1994). Hence, a rise in intracellular Ca2+ concen- 1996). In fact, the same procedure for inducing tration sufficient for inducing contraction is artificial hypothermia in hamsters was lethal in needed to maintain heart function at a cold tem- rats (Miyazawa et al. 2008). In addition to the perature. In the rat myocardia, basal intracellular cold resistance, the heart of a hibernator is known Ca2+ concentration, which is usually about to be resistant to various harmful stimuli. For 140 nM at 30–35 °C, is increased to 200–300 nM instance, the heart of one of the hibernators, in response to a cold temperature around 10 °C hedgehog dog, is hardly affected by manipula- (Liu et al. 1991; Wang and Zhou 1999). Such a tions that elicit atrial fibrillation (e.g., aconitine 2+ rise in the basal Ca concentration would nega- administration, high concentration of CaCl2 tively impact cardiac function, since it enhances administration, or ligation of the hepatic artery) basal tone, resulting in insufficient dilation dur- (Johansson 1985, 1996). ing the diastolic filling period. Furthermore, a rise in the basal Ca2+ concentration also lowers 52 Y. Horii et al.

3.5.2 Adaptive Changes increased release, that reuptake of Ca2+ by the in the Heart Prior sarcoplasmic reticulum is enhanced (Belke et al. to Hibernation 1991). Collectively, suppression of channel activ- ity in the plasma membrane with concomitant As mentioned above, the specific innate charac- activation of store function enables efficacious teristics of the heart of hibernators would be Ca2+ cycling at a cold temperature. important for enabling hibernation. It should be noted, however, that maintenance of cardiac function during hibernation does not totally 3.5.3 Molecular Basis depend on the innate ability of the heart. It is for the Adaptive Changes believed that adaptive changes that occur in in the Heart of Hibernating response to a short photoperiod and cold ambient Animals temperature are also essential for entering deep hibernation, as well as for keeping a hypothermic As mentioned above, cardiac myocytes of hiber- state and for recovery to a euthermic state. nating mammals are characterized by reduced Therefore, numerous studies have been carried Ca2+ entry into the cell membrane (Alekseev out to reveal remarkable differences between et al. 1996; Yatani et al. 2004; Dibb et al. 2005) hibernating animals and their euthermic and a concomitant enhancement of Ca2+ release counterparts. from and reuptake by the sarcoplasmic reticulum The most striking adaptive changes in the (Kondo and Shibata 1984; Belke et al. 1991; heart of hibernating animals are alterations in Wang et al. 2002). These adaptive changes would intracellular Ca2+ mobilization involving cardiac help in preventing excessive Ca2+ entry and its excitation-contraction coupling (Kondo and overload and in maintaining the resting levels of Shibata 1984; Lakatta and Guarnieri 1993). In intracellular Ca2+ (Wang et al. 2002). The molec- general, intracellular Ca2+ for contraction of car- ular basis of reduced Ca2+ entry into the cell diac muscle is supplied by its entry into the cell membrane would not be due to reduced expres- through the L-type Ca2+ channel followed by Ca2+ sion of the L-type Ca2+ channel protein but rather release from the sarcoplasmic reticulum, a Ca2+ due to a decrease in channel activity by phos- storage organelle (Kurihara 1994). In hibernating phorylation of the molecule (Kokoz et al. 2000) chipmunks, it has been demonstrated that activity (Fig. 3.3). of the L-type Ca2+ channel is suppressed and As for the increased release of Ca2+ from intra- thereby entry of extracellular Ca2+ is limited cellular stores, there has been a study demon- (Kondo and Shibata 1984). Since excessive Ca2+ strating that the density of ryanodine receptors is entry and its overload would damage cardiomyo- increased in the sarcoplasmic reticulum, although cytes through induction of apoptosis and/or the expression level of the receptors remains necrosis, maintenance of Ca2+ homeostasis is unchanged (Milner et al. 1991). The ryanodine essential for preventing profound arrhythmia and receptor is the major Ca2+ release channel on the ventricular fibrillation (Lakatta and Guarnieri sarcoplasmic reticulum required for excitation-­ 1993). Thus, it can be considered that suppres- contraction coupling in cardiac muscle (Kurihara sion of the L-type Ca2+ channel activity is an 1994). In addition, expression of sarco(endo) appropriate adaptive event for hibernating ani- plasmic reticulum Ca2+-ATPase 2a (SERCA2a) is mals. Meanwhile, suppression of the channel upregulated, and a negative regulator of activity may have a negative impact on cardiac SERCA2a, phospholamban (PLB), is downregu- contractility. To compensate for the reduced Ca2+ lated during hibernation (Brauch et al. 2005). entry, release of Ca2+ from intracellular stores is These changes enable a prompt removal of cyto- enhanced during hibernation (Kondo and Shibata solic Ca2+, thereby ensuring diastolic filling 1984). It is also important, in addition to the (Fig. 3.3). 3 The Mechanism Enabling Hibernation in Mammals 53

Fig. 3.3 Adaptive changes in molecules related to main- RyR ryanodine receptors, SERCA2a sarco(endo)plasmic tenance of intracellular Ca2+, contractility or synchronous reticulum Ca2+-ATPase 2a, PLB phospholamban, α-MHC contraction in cardiac myocytes of hibernating animals. myosin heavy chain-α, Cx connexin

Moreover, the expression of functional pro- that in hibernating animals can be successfully teins related to contractility (e.g., myosin heavy produced (Miyazawa et al. 2008). This procedure chain-α, ventricular myosin light chain, and the may reproduce a hypothermic condition without troponin C) and the expression of proteins promoting possible autonomic functions that involved in synchronous contraction (e.g., con- would usually be triggered in natural nexin43) have been shown to be upregulated or hibernation. downregulated appropriately in hibernating ani- Even after sufficient exposure to an environ- mals (Saitongdee et al. 2000; Brauch et al. 2005; ment that is appropriate for induction of adaptive Fedorov et al. 2005). Importantly, the onset of changes, hamsters show abnormal electrocardio- these changes precedes the onset of hibernation grams (ECG) such as J wave and/or signs related (Kondo 1987; Saitongdee et al. 2000), indicating to atrioventricular block when the hypothermic that these changes in gene expression and subse- condition is forcibly induced (Miyazawa et al. quent functional remodeling are preparatory pro- 2008). The J wave, which is typically described cesses for entering hibernation and are therefore in hypothermia in nonhibernating mammals indispensable for acquiring cold resistance (Brunson et al. 2005), is a wave located at the (Fig. 3.3). point of the end of the QRS complex and occupy- ing the initial part of the ST segment (Gussak et al. 1995). The origin of the J wave during 3.5.4 Autonomic Regulation hypothermia has been attributed to injury current, of the Heart delayed ventricular depolarization and early During Hibernation repolarization, tissue anoxia, and acidosis (Brunson et al. 2005). If the adaptive changes The adaptive changes prior to hibernation would exclusively contribute to cold tolerance of the alone be insufficient to maintain cardiac pulsatil- heart, heart pulsatility of well-adapted hamsters ity under an extremely hypothermic condition can be maintained appropriately not only during during hibernation, although these changes are natural hibernation but also during a forcibly undoubtedly indispensable. The operation of induced hypothermic condition. Therefore, the autonomic regulation for maintaining proper car- fact that the J wave as well as abnormal ECG diac pulsatility during hibernation has been sug- signs related to conduction block are not observed gested by experiments focusing on artificially in natural hibernation (Mertens et al. 2008; induced hypothermia in hamsters. By combining Miyazawa et al. 2008) can be rationally explained pentobarbital anesthesia with cooling of the ani- by the operation of regulatory mechanisms dur- mal, forced hypothermia that is comparable to ing natural hibernation to coordinate the cardiac 54 Y. Horii et al. conducting system properly and to prevent car- such as black bears (Fedorov et al. 2009, 2011) diac impairment caused by hypothermia. The and ground squirrels (Epperson et al. 2004; precise regulatory mechanisms have so far Williams et al. 2005) and plays an important role remained elusive. in neuroprotection (Tong et al. 2013; Peretti et al. 2015). Also, CIRP is expressed in response to a cold stress in the treefrog (Sugimoto and Jiang 3.6 Mechanism of Protection 2008). Accordingly, cold-shock proteins might Against Cold Temperature help to protect organs including the heart against a harmful low temperature during hibernation. 3.6.1 Cold-Shock Proteins-­ Associated Protection During Hibernation 3.6.2 Hibernation-Specific Alternative Splicing Generally, the heart of mammalians cannot keep of the CIRP Gene beating in a deep hypothermic condition (Ivanov 2000), suggesting that a cold temperature is We recently reported the unique expression pat- harmful to the heart. In contrast, the heart of tern of CIRP in the hearts of hibernating hamsters hibernating animals is capable of maintaining (Sano et al. 2015). In our study, RT-PCR analysis constant beating despite a decrease in body tem- revealed that CIRP mRNA is constitutively perature to less than 10 °C during hibernation expressed in the heart of a nonhibernating euther- (Carey et al. 2003). Therefore, in addition to the mic hamster with several different forms proba- adaptive changes prior to entering hibernation bly due to alternative splicing. The short product and the operation of autonomic regulatory mech- contained the complete open reading frame for anisms during hibernation, protection of cardio- full-length CIRP. On the other hand, the long myocytes against harmful effects of a cold product had inserted sequences containing a stop temperature would be essential to maintain heart codon, suggesting production of a C-terminal function under a condition of extreme hypother- deletion isoform of CIRP. The RNA-binding mia. In relation to the protective mechanism, domain in the N-terminal region (Lleonart 2010) recent studies have been focused on functional is conserved in the long isoform, indicating that roles of cold-shock proteins, including cold-­ the isoform possesses RNA-binding activity inducing RNA-binding protein (CIRP) and RNA-­ equal to that of full-length CIRP. However, the binding motif 3 (RBM3) (Zhu et al. 2016). It has isoform lacks critical phosphorylation and meth- been demonstrated that CIRP and RBM3 are ylation sites located in the C-terminal region, the induced by cold stress in cultured cells phosphorylation and/or methylation of which is (Nishiyama et al. 1997; Gotic et al. 2016; Zhu related to activation of CIRP (De Leeuw et al. et al. 2016). These proteins regulate gene expres- 2007; Lleonart 2010). It is thus probable that the sion at the level of translation (i.e., mRNA splic- C-terminally truncated isoform plays a dominant-­ ing, stability, and transport) and thus allow cells negative role over the full-length CIRP. In con- to respond rapidly to cold stress (Lleonart 2010; trast to nonhibernating hamsters, only the short Zhu et al. 2016). Accumulating evidence indi- product is expressed in hibernating animals. It is cates that CIRP and RBM3 play important roles therefore speculated that the dominant-negative in the protection of various types of cells against regulation is important to mask the function of harmful effects of a cold temperature (Gualerzi CIRP under a nonhibernating condition. The et al. 2003; Saito et al. 2010). dominant-negative regulation combined with The prominent action of cold-shock proteins, constitutively active transcription may permit which was originally revealed in cells of nonhi- rapid expression of CIRP function by switching bernators, has been shown to function during the splicing pattern, leading to avoidance of hibernation. For example, it has been reported hypothermic damage in the heart (Fig. 3.4). that RBM3 is increased in hibernating mammals 3 The Mechanism Enabling Hibernation in Mammals 55

Fig. 3.4 Alternative splicing of cold-inducible RNA-binding protein (CIRP) gene in nonhibernating euthermic and hibernating hypothermic hamsters. (The figure was modified from our published article Sano et al. 2015)

It would be of interest to uncover the factors and Kloner 2011; Tissier et al. 2012). Mild hypo- causing the shift in alternative splicing of CIRP. thermia, 32–35 °C, is very potent for reducing Recent evidence from cultured cells suggests that myocardial infarct size in some experimental ani- a mild cold temperature (32 °C) is a possible trig- mal models such as rabbits, dogs, sheep, pigs, ger for splicing regulation of the CIRP gene, and rats (Tissier et al. 2012). In addition, induced since mild cold exposure increases the expression hypothermia has been shown to reduce the risk of of CIRP mRNA without affecting its pre-mRNA cerebral ischemic damage both in animal studies levels (Gotic et al. 2016). This assumption can be and in humans, who have been resuscitated fol- applicable to the shift in alternative splicing of lowing cardiac arrest (Galvin et al. 2015). Thus, CIRP under the condition of natural hibernation, it is important to devise a method by which hypo- since animals go through a gradual decrease in thermia can be induced safely and simply in non- body temperature, and they would maintain mild hibernating mammals including humans. hypothermia, about 25–30 °C, for several hours Therapeutic hypothermia is generally induced (Horwitz et al. 2013). Taken together, it is reason- by a combination of anesthesia with cooling in able to consider that mild hypothermia during the the patient (Galvin et al. 2015). In addition, safe induction period of hibernation might induce and simple pharmacological approaches to hibernation-specific alternative splicing of CIRP achieve therapeutic hypothermia have been in the hamster heart. investigated. For instance, hydrogen sulfide can induce a state of hypothermia in mice by inhibit- ing cytochrome oxidase, which decreases their 3.7 Induction of Artificial metabolic rate and core body temperature (Guo Hypothermia et al. 2012). Administration of capsaicin also in Nonhibernating Animals reduces body temperature by about 2–3 °C (Jakab et al. 2005; Swanson et al. 2005; Jones et al. 3.7.1 Significance of Therapeutic 2009; Dow et al. 2014) since capsaicin is an ago- Hypothermia nist of TRPV1, which can detect a painful hot temperature (>42 °C) (Montell and Caterina Hypothermia results in a reduction of cellular 2007) and would be recognized as heat exposure, metabolic rate and oxygen consumption, indicat- leading to reduction of body temperature medi- ing that it may have therapeutic efficacy (Hale ated by the thermoregulatory center. It should be 56 Y. Horii et al. noted, however, that the target temperature is nation mechanisms are considered to be a poten- generally about 30 °C, which is categorized as tial therapeutic target for the treatment of several mild hypothermia, and that it is difficult to induce diseases. Although the application of this unique hibernation-like extreme hypothermia even by phenomenon to medical fields has been strongly these methods. desired, a poor understanding of the mechanisms limits the progress toward developing novel ther- apeutic strategies. A large number of previous 3.7.2 Induction of Hypothermia experiments focused on adaptive changes in the by Activation of Central heart prior to hibernation. It is clear that adaptive Adenosine A1-Receptor changes are involved in the beneficial properties of hibernating animals. However, it remains One of the profound problems that occur during unclear whether these changes are solely respon- induction of artificial hypothermia is heart dys- sible for the establishment of a hibernating con- function such as ventricular fibrillation and car- dition. For instance, it is uncertain whether the diac arrest. Even in hibernators such as hamsters, changes at the molecular level (see Fig. 3.3) are abnormal ECG is recorded during nonhiberna- sufficient for maintaining cardiac pulsatility tion artificial hypothermia induced by pentobar- under an extremely hypothermic condition. On bital anesthesia and cooling (Miyazawa et al. the other hand, artificially induced hypothermia 2008). To devise a safe method for induction of may provide a valuable tool to answer the ques- hypothermia, elucidation of the mechanisms for tion. The method for inducing hypothermia forc- tolerance to cold stress during hibernation would ibly in hamsters allows reproduction of a provide valuable information. Central adenosine hypothermic condition in the absence of possible A1-receptor-mediated signals play a role in the hibernation-specific reactions. Unlike hypother- induction and maintenance of hibernation mia in natural hibernation, the forced induction (Tamura et al. 2005; Jinka et al. 2011; Iliff and of hypothermia causes irreversible injury of the Swoap 2012). The predominant role of adenosine myocardium (Miyazawa et al. 2008). Comparison A1-receptor-mediated signals leads to the idea of the heart in forced hypothermia with that dur- that activation of adenosine A1-receptors would ing hibernation would be valuable for identifying induce hypothermia in both hibernating and non- critical factors related to cold resistance of the hibernating mammals. In accordance with this, heart. Thus, it is expected that further studies central administration of an adenosine using artificial hypothermia may provide a break- A1-receptor agonist and subsequent cooling through in understanding the hibernation induces extreme hypothermia in hamsters mechanisms. (Miyazawa et al. 2008) and rats (Tupone et al. 2013; Shimaoka et al. 2018) without accompany- Acknowledgments The reviewed results obtained in our ing atrioventricular block or abnormal laboratory were supported in part by JSPS KAKENHI ECG. These findings suggest that central adenos- Grant numbers JP15K14876 and JP25660249 to Y.S. and JP17J02251 to Y.H., and the Sasakawa Scientific ine A1-receptor-mediated signals would provide Research Grant from The Japan Science Society to Y.H. an appropriate condition for maintaining normal sinus rhythm during extreme hypothermia. References

3.8 Conclusion Abnous K, Dieni CA, Storey KB (2008) Regulation of Akt during hibernation in Richardson’s ground squir- Entering hibernation in mammals confers resis- rels. Biochim Biophys Acta 1780:185–193 Alekseev AE, Markevich NI, Korystova AF, Terzic A, tance to various harmful events such as low body Kokoz YM (1996) Comparative analysis of the kinetic temperature, severe ischemia, bacterial infection, characteristics of L-type calcium channels in cardiac irradiation, and muscle disuse. Therefore, hiber- cells of hibernators. Biophys J 70:786–797 3 The Mechanism Enabling Hibernation in Mammals 57

Andrews MT, Russeth KP, Drewes LR, Henry PG (2009) tion in the golden-mantled ground squirrel. Mol Cell Adaptive mechanisms regulate preferred utilization of Proteomics 3:920–933 ketones in the heart and brain of a hibernating mam- Fedorov VV, Li L, Glukhov A, Shishkina I, Aliev RR, mal during arousal from torpor. Am J Physiol Regul Mikheeva T, Nikolski VP, Rosenshtraukh LV, Efimov Integr Comp Physiol 296:R383–R393 IR (2005) Hibernator Citellus undulates maintains Barnes BM (1989) Freeze avoidance in a : body safe cardiac conduction and is protected against temperatures below 0 degrees C in an Arctic hiberna- tachyarrhythmias during extreme hypothermia: pos- tor. Science 244:1593–1595 sible role of Cx43 and Cx45 up-regulation. Heart Belke DD, Milner RE, Wang LC (1991) Seasonal varia- Rhythm 2:966–975 tions in the rate and capacity of cardiac SR calcium Fedorov VV, Glukhov AV, Sudharshan S, Egorov accumulation in a hibernating species. Y, Rosenshtraukh LV, Efimov IR (2008) 28:354–363 Electrophysiological mechanisms of antiarrhythmic Bieber C, Lebl K, Stalder G, Geiser F, Ruf T (2014) Body protection during hypothermia in winter hibernat- mass dependent use of hibernation: why not prolong ing versus nonhibernating mammals. Heart Rhythm the active season, if they can? Funct Ecol 28:167–177 5:1587–1596 Brauch KM, Dhruv ND, Hanse EA, Andrews MT (2005) Fedorov VB, Goropashnaya AV, Tøien Ø, Stewart NC, Digital transcriptome analysis indicates adaptive Gracey AY, Chang CL, Qin SZ, Pertea G, Quackenbush mechanisms in the heart of a hibernating mammal. J, Showe LC, Showe MK, Boyer BB, Barnes BM Physiol Genomics 23:227–234 (2009) Elevated expression of protein biosynthesis Breukelen FV, Martin SL (2015) The hibernation con- genes in liver and muscle of hibernating black bears tinuum: physiological and molecular aspects of meta- (Ursus americanus). Physiol Genomics 37:108–118 bolic plasticity in mammals physiology. Physiology Fedorov VB, Goropashnaya AV, Tøien O, Stewart NC, 30:273–281 Chang C, Wang H, Yan J, Showe LC, Showe MK, Brunson CE, Abbud E, Osman K, Skelton TN, Markov Barnes BM (2011) Modulation of gene expression AK (2005) Osborn (J) wave appearance on the electro- in heart and liver of hibernating black bears (Ursus cardiogram in relation to potassium transfer and myo- americanus). BMC Genomics 12:171 cardial metabolism during hypothermia. J Investig Fleck CC, Carey HV (2005) Modulation of apoptotic Med 53:434–437 pathways in intestinal mucosa during hibernation. Am Caprette DR, Senturia JB (1984) Isovolumetric perfor- J Physiol Regul Integr Comp Physiol 289:R586–R595 mance of isolated ground squirrel and rat hearts at low Galvin IM, Levy R, Boyd JG, Day AG, Wallace MC temperature. Am J Physiol Regul Integr Comp Physiol (2015) Cooling for cerebral protection during brain 247:R722–R727 surgery. Cochrane Database Syst Rev 1:CD006638 Carey HV, Andrews MT, Martin SL (2003) Mammalian Geiser F (2004) Metabolic rate and body temperature hibernation: cellular and molecular responses to reduction during hibernation and daily torpor. Annu depressed metabolism and low temperature. Physiol Rev Physiol 66:239–274 Rev 83:1153–1181 Gotic I, Omidi S, Fleury-Olela F, Molina N, Naef F, Chayama Y, Ando L, Tamura Y, Miura M, Yamaguchi Y Schibler U (2016) Temperature regulates splicing (2016) Decreases in body temperature and body mass efficiency of the cold-inducible RNA-binding protein constitute pre-hibernation remodelling in the Syrian gene Cirbp. Genes Dev 30:2005–2017 golden hamster, a facultative mammalian hibernator. Gualerzi CO, Giuliodori AM, Pon CL (2003) R Soc Open Sci 3:160002 Transcriptional and posttranscriptional control of De Leeuw F, Zhang T, Wauquier C, Huez G, Kruys V, cold-shock genes. J Mol Biol 331:527–539 Gueydan C (2007) The cold inducible RNA-binding Guo W, Kan JT, Cheng ZY, Chen JF, Shen YQ, Xu J, Wu protein migrates from the nucleus to cytoplasmic D, Zhu YZ (2012) Hydrogen sulfide as an endogenous stress granules by a methylation-dependent mecha- modulator in mitochondria and mitochondria dysfunc- nism and acts as a translational repressor. Exp Cell tion. Oxidative Med Cell Longev 2012:878052 Res 313:4130–4144 Gussak I, Bjerregaard P, Egan TM, Chaitman BR (1995) Dibb KM, Hagarty CL, Loudon AS, Trafford AW (2005) ECG phenomenon called the J wave. History, patho- Photoperioddependent modulation of cardiac excita- physiology, and clinical significance. J Electrocardiol tion contraction coupling in the Siberian hamster. Am 28:49–58 J Physiol Regul Integr Comp Physiol 288:R607–R614 Hale SL, Kloner RA (2011) Mild hypothermia as a car- Dow J, Simkhovich BZ, Hale SL, Kay G, Kloner RA dioprotective approach for acute myocardial infarc- (2014) Capsaicin-induced cardioprotection. Is hypo- tion: laboratory to clinical application. J Cardiovasc thermia or the salvage kinase pathway involved? Pharmacol Ther 16:131–139 Cardiovasc Drugs Ther 28:295–301 Harris MB, Milsom WK (1995) Parasympathetic influ- Duker GD, Olsson SO, Hecht NH, Senturia JB, Johansson ence on heart rate in euthermic and hibernating ground BW (1983) Ventricular fibrillation in hibernators and squirrels. J Exp Biol 198:931–937 nonhibernators. Cryobiology 20:407–420 Hoehn KL, Hudachek SF, Summers SA, Florant GL Epperson LE, Dahl TA, Martin SL (2004) Quantitative (2004) Seasonal, tissue-specific regulation of Akt/ analysis of liver protein expression during hiberna- protein kinase B and glycogen synthase in hiber- 58 Y. Horii et al.

nators. Am J Physiol Regul Integr Comp Physiol scription of the chipmunk HP-25 gene. Eur J Biochem 286:R498–R504 267:4635–4641 Horwitz BA, Chau SM, Hamilton JS, Song C, Gorgone Kojima M, Shiba T, Kondo N, Takamatsu N (2001) J, Saenz M, Horowitz JM, Chen CY (2013) Temporal The tree squirrel HP-25 gene is a pseudogene. Eur relationships of blood pressure, heart rate, baroreflex J Biochem 268:5997–6002 function, and body temperature change over a hiber- Kokoz YM, Grichenko AS, Korystova AF, Lankina DA, nation bout in Syrian hamsters. Am J Physiol Regul Pimenov OY, Markevich NI (2000) The regulation of Integr Comp Physiol 305:R759–R768 L-type Ca2+ currents in cardiac myocytes of hibernat- Houseknecht KL, Baile CA, Matteri RL, Spurlock ME ing animals. Membr Cell Biol 14:277–284 (1998) The biology of leptin: a review. J Anim Sci Kondo N (1987) Identification of a pre-hibernating state 76:1405–1420 in myocardium from nonhibernating chipmunks. Humphries MM, Thomas DW, Kramer DL (2003) The Experientia 43:873–875 role of energy availability in mammalian hiberna- Kondo N (2007) Endogenous circannual clock and HP tion: a cost-benefit approach. Physiol Biochem Zool complex in a hibernation control system. Cold Spring 76:165–179 Harb Symp Quant Biol 72:607–613 Iliff BW, Swoap SJ (2012) Central adenosine receptor Kondo N, Kondo J (1992) Identification of novel blood signaling is necessary for daily torpor in mice. Am proteins specific for mammalian hibernation. J Biol J Physiol Regul Integr Comp Physiol 303:R477–R484 Chem 267:473–478 Ivanov KP (2000) Physiological blocking of the mecha- Kondo N, Shibata S (1984) Calcium source for excitation-­ nisms of cold death: theoretical and experimental con- contraction coupling in myocardium of nonhibernat- siderations. J Therm Biol 25:467–479 ing and hibernating chipmunks. Science 225:641–643 Jackson DC, Ultsch GR (2010) Physiology of hibernation Kondo N, Sekijima T, Kondo J, Takamatsu N, Tohya K, under the ice by turtles and . J Exp Zool A Ecol Ohtsu T (2006) Circannual control of hibernation by Genet Physiol 313:311–327 HP complex in the brain. Cell 125:161–172 Jakab B, Helyes Z, Varga A, Bölcskei K, Szabó A, Sándor Kronfeld-Schor N, Richardson C, Silvia BA, Kunz TH, K, Elekes K, Börzsei R, Keszthelyi D, Pintér E, Petho Widmaier EP (2000) Dissociation of leptin secretion G, Németh J, Szolcsányi J (2005) Pharmacological and adiposity during prehibernatory fattening in little characterization of the TRPV1 receptor antagonist brown bats. Am J Physiol Regul Integr Comp Physiol JYL1421 (SC0030) in vivo and in vitro in the rat. Eur 279:R1277–R1281 J Pharmacol 517:35–44 Kurihara S (1994) Regulation of cardiac muscle contrac- Jinka TR, Tøien Ø, Drew KL (2011) Season primes the tion by intracellular Ca2+. Jpn J Physiol 44:591–611 brain in an arctic hibernator to facilitate entrance into Lakatta EG, Guarnieri T (1993) Spontaneous myocardial torpor mediated by adenosine A1 receptors. J Neurosci calcium oscillations: are they linked to ventricular 31:10752–10758 fibrillation? J Cardiovasc Electrophysiol 4:473–489 Johansson BW (1985) Ventricular repolarization and Lee M, Choi I, Park K (2002) Activation of stress signal- fibrillation threshold in hibernating species. Eur Heart ing molecules in bat brain during arousal from hiber- J 6(Suppl D):53–62 nation. J Neurochem 82:867–873 Johansson BW (1996) The hibernator heart–nature’s Liu B, Wang LC, Belke DD (1991) Effect of low tem- model of resistance to ventricular fibrillation. perature on the cytosolic free Ca2+ in rat ventricular Cardiovasc Res 31:826–832 myocytes. Cell Calcium 12:11–18 Jones WK, Fan GC, Liao S, Zhang JM, Wang Y, Weintraub Lleonart ME (2010) A new generation of proto-­ NL, Kranias EG, Schultz JE, Lorenz J, Ren X (2009) oncogenes: cold-inducible RNA binding proteins. Peripheral nociception associated with surgical inci- Biochim Biophys Acta 1805:43–52 sion elicits remote nonischemic cardioprotection via Lovegrove BG, Génin F (2008) Torpor and hibernation in a neurogenic activation of protein kinase C signaling. basal placental mammal, the Lesser Hedgehog Tenrec Circulation 120(suppl 1):S1–S9 Echinops telfairi. J Comp Physiol B 178:691–698 Kilduff TS, Krilowicz B, Milsom WK, Trachsel L, Wang Malan A (2014) The evolution of mammalian hibernation: LC (1993) Sleep and mammalian hibernation: homol- lessons from comparative acid-base physiology. Integr ogous adaptations and homologous processes? Sleep Comp Biol 54:484–496 16:372–386 Malhotra J, Gupta YK (1997) Effect of adenosine receptor Kobbe S, Dausmann K (2009) Hibernation in Malagasy modulation on pentylenetetrazole-induced seizures in mouse lemurs as a strategy to counter environmental rats. Br J Pharmacol 120:282–288 challenge. Naturwissenschaften 96:1221–1227 Mertens A, Stiedl O, Steinlechner S, Meyer M (2008) Kobbe S, Ganzhorn J, Dausmann K (2011) Extreme Cardiac dynamics during daily torpor in the Djungarian individual flexibility of in free-ranging hamster (Phodopus sungorus). Am J Physiol Regul Malagasy mouse lemurs (Microcebus griseorufus). Integr Comp Physiol 294:R639–R650 J Comp Physiol B 181:165–173 Milner RE, Michalak M, Wang LC (1991) Altered proper- Kojima M, Takamatsu N, Ishii T, Kondo N, Shiba T (2000) ties of calsequestrin and the ryanodine receptor in the HNF-4 plays a pivotal role in the liver-specific tran- cardiac sarcoplasmic reticulum of hibernating mam- mals. Biochim Biophys Acta 1063:120–128 3 The Mechanism Enabling Hibernation in Mammals 59

Milsom WK, Zimmer MB, Harris MB (1999) Regulation Shao C, Liu Y, Ruan H, Li Y, Wang H, Kohl F, of cardiac rhythm in hibernating mammals. Comp Goropashnaya AV, Fedorov VB, Zeng R, Barnes BM, Biochem Physiol A Mol Integr Physiol 124:383–391 Yan J (2010) Shotgun proteomic analysis of hiber- Miyazawa S, Shimizu Y, Shiina T, Hirayama H, Morita nating arctic ground squirrels. Mol Cell Proteomics H, Takewaki T (2008) Central A1-receptor activa- 9:313–326 tion associated with onset of torpor protects the heart Shimaoka H, Kawaguchi T, Morikawa K, Sano Y, Naitou against low temperature in the Syrian hamster. Am K, Nakamori H, Shiina T, Shimizu Y (2018) Induction J Physiol Regul Integr Comp Physiol 295:R991–R996 of hibernation-like hypothermia by central activation Montell C, Caterina MJ (2007) : chan- of the A1 adenosine receptor in a non-hibernator, the nels that are cool to the core. Curr Biol 17:R885–R887 rat. J Physiol Sci 68:425–430 Mzilikazi N, Lovegrove BG, Ribble DO (2002) Shintani M, Tamura Y, Monden M, Shiomi H (2005) Exogenous passive heating during torpor arousal Thyrotropin-releasing hormone induced thermogen- in free-ranging rock elephant , Elephantulus esis in Syrian hamsters: site of action and receptor myurus. Oecologia (Berl) 133:307–314 subtype. Brain Res 1039:22–29 Nikodijevic O, Sarges R, Daly JW, Jacobson KA (1991) Sugimoto K, Jiang H (2008) Cold stress and light signals Behavioral effects of A1- and A2-selective adenosine induce the expression of cold-inducible RNA binding agonists and antagonists: evidence for synergism and protein (cirp) in the brain and eye of the Japanese tree- antagonism. J Pharmacol Exp Ther 259:286–294 (Hyla japonica). Comp Biochem Physiol A Mol Nishino S, Fujiki N, Ripley B, Sakurai E, Kato M, Integr Physiol 151:628–636 Watanabe T, Mignot E, Yanai K (2001) Decreased Swanson DM, Dubin AE, Shah C, Nasser N, Chang L, Dax brain histamine content in hypocretin/orexin recep- SL, Jetter M, Breitenbucher JG, Liu C, Mazur C, Lord tor-­2 mutated narcoleptic dogs. Neurosci Lett B, Gonzales L, Hoey K, Rizzolio M, Bogenstaetter M, 313:125–128 Codd EE, Lee DH, Zhang SP, Chaplan SR, Carruthers Nishiyama H, Itoh K, Kaneko Y, Kishishita M, Yoshida NI (2005) Identification and biological evaluation of O, Fujita J (1997) A glycinerich RNA-binding protein 4-(3-trifluoromethylpyridin-2-yl)amide, a high affin- mediating cold-inducible suppression of mammalian ity TRPV1 (VR1) vanilloid receptor antagonist. J Med cell growth. J Cell Biol 137:899–908 Chem 48:1857–1872 Peretti D, Bastide A, Radford H, Verity N, Molloy Takamatsu N, Ohba K, Kondo J, Kondo N, Shiba T (1993) C, Martin MG, Moreno JA, Steinert JR, Smith T, Hibernation-associated gene regulation of plasma Dinsdale D, Willis AE, Mallucci GR (2015) RBM3 proteins with a collagen-like domain in mammalian mediates structural plasticity and protective effects of hibernators. Mol Cell Biol 13:1516–1521 cooling in neurodegeneration. Nature 518:236–239 Tamura Y, Shintani M, Nakamura A, Monden M, Shiomi Radulovacki M, Miletich RS, Green RD (1982) N6 H (2005) Phase-specific central regulatory systems of (L-phenylisopropyl) adenosine (L-PHA) increases hibernation in Syrian hamsters. Brain Res 1045:88–96 slow-wave sleep (S2) and decreases wakefulness in Tamura Y, Monden M, Shintani M, Kawai A, Shiomi rats. Brain Res 246:178–180 H (2006) Neuroprotective effects of hibernation-­ Ruf T, Geiser F (2015) Daily torpor and hibernation in regulating substances against low-temperature-­ and mammals. Biol Rev Camb Philos Soc induced cell death in cultured hamster hippocampal 90:891–926 neurons. Brain Res 1108:107–116 Saito K, Fukuda N, Matsumoto T, Iribe Y, Tsunemi Tamura Y, Shintani M, Inoue H, Monden M, Shiomi H A, Kazama T, Yoshida-Noro C, Hayashi N (2010) (2012) Regulatory mechanism of body temperature Moderate low temperature preserves the stemness of in the central nervous system during the maintenance neural stem cells and suppresses apoptosis of the cells phase of hibernation in Syrian hamsters: involvement via activation of the cold inducible RNA binding pro- of β-endorphin. Brain Res 1448:63–70 tein. Brain Res 1358:20–29 Tansey EA, Johnson CD (2015) Recent advances in ther- Saitongdee P, Milner P, Becker DL, Knight GE, Burnstock moregulation. Adv Physiol Educ 39:139–148 G (2000) Increased connexin43 gap junction protein in Ticho SR, Radulovacki M (1991) Role of adenosine in hamster cardiomyocytes during cold acclimatization sleep and temperature regulation in the preoptic area and hibernation. Cardiovasc Res 47:108–115 of rats. Pharmacol Biochem Behav 40:33–40 Sano Y, Shiina T, Naitou K, Nakamori H, Shimizu Y Tissier R, Ghaleh B, Cohen MV, Downey JM, Berdeaux A (2015) Hibernation-specific alternative splicing of the (2012) Myocardial protection with mild hypothermia. mRNA encoding cold-inducible RNA-binding pro- Cardiovasc Res 94:217–225 tein in the hearts of hamsters. Biochem Biophys Res Tong G, Endersfelder S, Rosenthal LM, Wollersheim Commun 462:322–325 S, Sauer IM, Bührer C, Berger F, Schmitt KR Serkova NJ, Rose JC, Epperson LE, Carey HV, Martin (2013) Effects of moderate and deep hypothermia SL (2007) Quantitative analysis of liver metabolites on RNA-binding proteins RBM3 and CIRP expres- in three stages of the circannual hibernation cycle in sions in murine hippocampal brain slices. Brain Res 13-lined ground squirrels by NMR. Physiol Genomics 1504:74–84 31:15–24 Tsukamoto D, Ito M, Takamatsu N (2017) HNF-4 par- ticipates in the hibernation-associated transcriptional 60 Y. Horii et al.

regulation of the chipmunk hibernation-related pro- administration of mioflazine, a nucleoside transport tein gene. Sci Rep 7:44279. https://doi.org/10.1038/ inhibitor. Psychopharmacology (Berlin) 91:434–439 srep44279 Williams DR, Epperson LE, Li W, Hughes MA, Taylor R, Tupone D, Madden CJ, Morrison SF (2013) Central acti- Rogers J, Martin SL, Cossins AR, Gracey AY (2005) vation of the A1 adenosine receptor (A1AR) induces Seasonally hibernating phenotype assessed through a hypothermic, torpor-like state in the rat. J Neurosci transcript screening. Physiol Genomics 24:13–22 33:14512–14525 Yan J, Barnes BM, Kohl F, Marr TG (2008) Modulation of Wang LCH, Lee TF (1996) Torpor and hibernation in gene expression in hibernating arctic ground squirrels. mammals: metabolic, physiological, and biochemi- Physiol Genomics 32:170–181 cal adaptations. In: Fregley MJ, Blatteis CM (eds) Yatani A, Kim SJ, Kudej RK, Wang Q, Depre C, Irie K, Handbook of physiology: environmental physiology. Kranias EG, Vatner SF, Vatner DE (2004) Insights into Oxford University Press, New York, pp 507–532 cardioprotection obtained from study of cellular Ca2+ Wang SQ, Zhou ZQ (1999) Alpha-stat calibration of handling in myocardium of true hibernating mammals. indo-1 fluorescence and measurement of intracellular Am J Physiol Heart Circ Physiol 286:H2219–H2228 free calcium in rat ventricular cells at different tem- Yu LC, Cai YP (1993a) Arousal following intra-preoptic peratures. Life Sci 65:871–877 area administration of naltrexone, ICI 174864 or nor-­ Wang LC, Belke D, Jourdan ML, Lee TF, Westly J, BNI in hibernating ground squirrels. Behav Brain Res Nurnberger F (1988) The “hibernation induction 57:31–35 trigger”: specificity and validity of bioassay using Yu LC, Cai YP (1993b) Effects of opioid receptors antag- the 13-lined ground squirrel. Cryobiology 1988 onists administration to suprachiasmatic nucleus on 25:355–362 hibernation of ground squirrels Citellus dauricus. Wang SQ, Lakatta EG, Cheng H, Zhou ZQ (2002) Comp Biochem Physiol C 104:249–252 Adaptive mechanisms of intracellular calcium Zervanos SM, Maher CR, Florant GL (2014) Effect of homeostasis in mammalian hibernators. J Exp Biol body mass on hibernation strategies of woodchucks 205:2957–2962 (Marmota monax). Integr Comp Biol 54:443–451 Wauquier A, Van Belle H, Van den Broeck WA, Janssen Zhu X, Bührer C, Wellmann S (2016) Cold-inducible pro- PA (1987) Sleep improvement in dogs after oral teins CIRP and RBM3, a unique couple with activities far beyond the cold. Cell Mol Life Sci 73:3839–3859