Sleep Deprivation: Comparison
Please note this is a comparison between Version 2 by Camila Xu and Version 3 by Beatrice Ragnoli.

Sleep health and its adaptation to individual and environmental factors are crucial to promote physical and mental well-being across animal species.

  • sleep deprivation
  • immune system suppression
  • SARS-CoV-2 infection

1. Introduction

The need of sleep in all animal species suggests that sleep plays a pivotal role in preserving vital functions. Over the past decades, many studies have revealed the strict relationship between sleep and physical or mental well-being. In particular, sleep and the immune system appear to be bidirectionally linked, especially during the body’s defense against disease [1]. For example, it is well established how the host immune response to external pathogens, such as viruses, can be negatively influenced by deprivation of sleep [2]. Conversely, a regular sleep routine boosts the immune system, ensuring appropriate and effective immune responses. In this regard, some authors have identified different effects of total sleep deprivation (TSD) in rats [3][4][5], in particular, a TSD-induced defect of host defense [6][7][8][9][10]. In previous studies, the effects of sleep deprivation were demonstrated also in human subjects including enhancement of evoked potentials, heightening of fantasy in fantasy-impoverished subjects and alleviation of endogenous depression. Sleep has also been involved in the plastic cerebral transformations during the learning and memory processes [11][12][13]. More recent studies highlighted the role of sleep in maintaining metabolic homeostasis either in animal models than in humans and a possible correlation between sleep deprivation and the onset of a metabolic syndrome (MetS) as a consequence [14]. This complex network may furthermore be related with changes in immune system that may interfere with a physiological response increasing the susceptibility to viral infections.

2. Sleep Deprivation Effects in Rat Models

Some authors reported several major effects of sleep deprivation (SDES) arising in rats after TSD or paradoxical sleep deprivation (PSD) through the disk-over-water (DOW) method [3]. These effects listed in Table 1 were subsequently confirmed by other groups [4][5] (Table 1).
Table 1. Major sleep deprivation effects in rats.
Symptoms Effects
Mortality Rats died or presented signs of imminent death usually after two or three weeks of TSD, unless sleep was recovered. PSD rats also died after 4–6 week.
Food intake Despite increased food intake, with increased caloric values, TSD and PSD rats lost weight.
Appearance Rats progressively appeared scrawny and debilitated.
[6]. By contrast, Everson isolated opportunistic and facultative anaerobe microbes in the blood of 5/6 near-terminal TSD rats, but none in controls, indicating, for the first time, an association between TSD and impaired immune response [7]. Everson’s results were subsequently confirmed [8][9] even though in vivo studies have produced conflicting results [10] (Table 2).
Table 2. Host defense effects in TSD rats.
Immune System Effects Findings
Increased permeability of the gut wall Gut wall of TSD rats becomes porous letting bacteria migrate to the peritoneal cavity [8].
Generalized failure of immune function Skin lesions without inflammation and lack of fever [9]
Skin Rats progressively appeared scrawny and debilitated.
Thermoregulatory changes TSD rats showed an initial increase and subsequent decrease in waking Tip. PSD rats only showed Tip decline.
Paradoxical sleep Recovery from prolonged TSD determined large rebounds of PS. The same was demonstrated after only 2–4 days of TSD [3].
TSD: total sleep deprivation, PSD: paradoxical sleep deprivation, EE: energy expenditure, PS: paradoxical sleep, Tip: intraperitoneal temperature.
Altogether, these studies demonstrated that TSD and PSD inevitably lead to a sleep-related syndrome with harmful effects on psychophysical health. In particular, sleep-deprived rats tend to show specific characteristics of progressive energy burn, typical skin lesions, thermoregulatory unbalance, leading to death, features never described in stressed rats [3][4].
The past four decades have witnessed a major paradigm shift in the study of the effect of sleep disturbances on the host immune response to invading pathogens [4]. Early studies about alteration of immune functions in TSD and PSD rats, assessing the amount of splenocytes, mitogen-induced lymphocyte proliferation test, and in vitro and in vivo plaque formation in response to various antigens, did not report significant differences from control group, suggesting that sleep deprivation does not lead to immune suppression
Abnormal T lymphocyte-mediated response
TSD rats injected with allogenic tumor cells develop slow-growing tumors, which regress more rapidly than yoked controls. Time onset of the reaction is typical of a T mediated response [10].
TDS: total sleep deprivation.
Data supporting that the gut is one of the primary sites of immune system dysregulation came from experiments on TSD rats orally treated with a mix of broad-spectrum antibiotics, where no bacteria could be found in the gut, blood, liver, kidneys and mesenteric nodes. The fact that these antibiotic-treated TSD rats continued experiencing a body temperature decline, a profound catabolic state manifested by high food intake and weight loss, and died “on schedule” in the absence of systemic bacterial infection supports the notion that an impaired immune response, caused by TSD, may play a decisive role in rat death [8]. However, taking together published data on this matter, we do not have a clear picture of how specific the effects of TSD may be in these animals. Indeed, sleep is so physiological in mammals that effects of its deprivation could be quite similar since produced by similar pathways [3][4]. Moreover, Sun Q et al. showed the effects of sleep deprivation on different signaling pathways, in particular, reduced expression of insulin receptor substrate (IRS)/phosphoinositide 3-kinase (PI3K)/AKT and the mammalian target of rapamycin (mTOR) as well as FoxO1 signaling pathways. Another important axis, the one regulated by the long isoform of the leptin receptor (LepRb)-mediated JAK2/STAT3 resulted attenuated by sleep deprivation. Young rats sleep deprived also experienced an alteration of the genes involved in the transcriptional feedback loop regulated by CLOCK and BMAL1 proteins. All these events confirm that alteration of sleep length causes dysregulation of several pathways in the hypothalamus with consequences in the regulation of hunger and energy consumption [15].

3. Sleep Deprivation Effects in Humans

This section may be divided by subheadings. It should provide a concise and precise description of the experimental results, their interpretation, as well as the experimental conclusions that can be drawn. Many causes are responsible for loss or dysregulation of sleep and usually a multifactorial origin is recognized. Among the most common reported causes of sleep loss there are respiratory disorders like apnea, but also other neurological and psychiatric conditions such as insomnia, parasomnias, mood disturbances, restless leg syndrome, and psychosis. It has been shown that the sleep architecture may change with age; deep sleep (characterized by delta-waves) decreases while the proportion of lighter sleep increases; this may conduct to increased sleep dysregulation [16]. Sleep disorders are widely distributed, as reported by different studies. Sleep loss affects more than 50 million people in America [17]. Léger D et al. showed that in 20% of the young adults analyzed (range 25–45 years old), sleep was reduced by ninety minutes as compared to what is needed for wellbeing [18]. Other authors reported in the last thirty years a consistent decrease in the duration of sleep coming up to about 18 min for night [19][20]. The accelerated rhythms of our society, which keep us ever connected, for work or pleasure, using computers, mobile phones and other devices until late at night, will likely determine an increase of sleep disorders. The effects of sleep deprivation on health outcomes have also been heavily studied in humans. From these studies, it seems that the most frequently observed effect in both TSD rats and humans is increased hunger [11][12][13]. Moreover, a 72-h TSD in humans significantly increased urea excretion, as well as plasma urea nitrogen increased in TSD and PSD rats in TSD and PSD rats [21]. Increased appetite in PSD subjects was also reported by Dement and Sampson [22][23]. However, psychological effects of sleep loss are not as quickly evident in humans as in rats. If 13.6 h in rats vs. 8.0 h in humans are considered the mean sleep need, the effects of TSD should onset 1.7 time faster in rats than in humans [3]. Studies on PSD in humans did not show evident damaging symptoms but they were relatively short (range 1–16 nights) [24]. The first report on long-time PSD results was published by Wyatt et al., who chronically administered phenelzine, a monoamine oxidase inhibitor (MAOI), in seven narcoleptic subjects, inducing severe and prolonged PS loss. Based on periodic recordings, in two subjects, PS almost disappeared for more than a year, with only mild symptoms reported as side effects of the drug mainly increased weight for excessive eating [25]. It should, however, be pointed out that in the aforementioned studies, cataplexy occurred when there were present either short intervals of PS or PS like moments [3]. Further studies showed how PSD could affect cerebral activity. These effects included enhancement of evoked potentials, boosting of fantasy in subjects with poor imagination and relief of endogenous depression [26]. Sleep has also been involved in neuroplasticity and in the learning and memory processes. Many published data confirmed that sleep is implicated in the fixation of short-term memory. Subjects who were sleep-deprived during post-training nights showed virtually no performance improvement on the following days, whereas subjects allowed to sleep immediately after training displayed a significant performance enhancement [27].
Among risk factors for human healthiness, sleep deprivation is one of the most changeable.
The American Heart Association and the Centre for disease Control and Prevention (CDC) claim that a sleep period of seven hours/night is necessary for favouring well-being and decreased probability of diseases [28][29]. The effects of inadequate sleep are dangerous.
In fact, morbidity and mortality connected to cardiovascular diseases (CVD) are dangerously increased by sleep disorders [30][31]. Everyday rhythm is present in cardiac, smooth muscle, and endothelial cells, modulating heart rate, blood pressure, and endothelial activity, [32] all functions with daily variation. Endothelial dysfunction and platelet activation are the trigger of atherosclerosis processes, already present before clinical presentation of CVD [33][34][35]; and for this reason, a significant target for evaluating the initial effects of sleep deprivation on the cardiovascular system. Moreover, endothelial activity evaluated in coronary and peripheral arteries reveals following CVD episodes (i.e., myocardial infarction and stroke) and death [36][37]. This is particularly true in subjects with chronic respiratory diseases such as COPD during stability phases [38][39] and during exacerbations [40]. Sleep deprivation may favour the onset and progression of age-related endothelial dysfunction and through this process favour pathogenesis and development of CVD [41]. Consequently, understanding the involvement of sleep in maintaining endothelial function may be useful to reach a healthy vascular aging.
The impacts of different hormones levels have been found to have a pivotal role in sleep deprivation. In fact, a chronic deprivation of sleep seems to be associated with increased cortisol levels and conversely with decreased levels of testosterone. Testosterone is known to intensify the function of the main blocker of neurotransmissions (gamma-aminobutyric acid—GABA) and of serotonin, involved in the regulation stabilization of mood and depression. The reduced testosterone levels could be involved in the relationship between depression and anxiety. Furthermore, elevated serum cortisol levels are associated with depression, anxiety, hypertension, obesity, and type II diabetes. Chronic sleep deprivation is associated with high inflammatory mediators, which are elevated in both comorbid conditions and mental illnesses.
The subjective experience of sleep loss can be distressing and conduct to increased cortisol levels, which can increase blood sugar, blood pressure and cravings for carbohydrates causing weight gain and consequently either medical and psychiatric problems. For the aforementioned reasons sleep loss can have painful and detrimental effects depending on subjects and can worsen pre-existing conditions [42].

References

  1. Ibarra-Coronado, E.G.; Pantaleón-Martínez, A.M.; Velazquéz-Moctezuma, J.; Prospéro-García, O.; Méndez-Díaz, M.; Pérez-Tapia, M.; Pavón, L.; Morales-Montor, J. The Bidirectional Relationship between Sleep and Immunity against Infections. J. Immunol. Res. 2015, 2015, 678164.
  2. Asif, N.; Iqbal, R.; Nazir, C.F. Human immune system during sleep. Am. J. Clin. Exp. Immunol. 2017, 6, 92–96.
  3. Rechtschaffen, A.; Bergmann, B.M.; Everson, C.A.; Kushida, C.A.; Gilliland, M.A. Sleep Deprivation in the Rat: X. Integration and Discussion of the Findings. Sleep 1989, 12, 68–87.
  4. Rechtschaffen, A.; Bregmann, B.M. Sleep deprivation in the rat: An update of the 1989 paper. Sleep 2002, 25, 18–24.
  5. Rechtschaffen, A.; Bregmann, B.M. Sleep deprivation in the rat by the disk-over-water method. Behav. Brain Res. 1995, 69, 55–63.
  6. Benca, R.M.; Kushida, C.A.; Everson, C.A.; Kalski, R.; Bergmann, B.M.; Rechtschaffen, A. Sleep Deprivation in the Rat: VII. Immune Function. Sleep 1989, 12, 47–52.
  7. Everson, C.A. Sustained sleep deprivation impairs host defense. Am. J. Physiol. Integr. Comp. Physiol. 1993, 265, R1148–R1154.
  8. Bergmann, B.M.; Gilliland, M.A.; Feng, P.-F.; Russell, D.R.; Shaw, P.; Wright, M.; Rechtschaffen, A.; Alverdy, J.C. Are Physiological Effects of Sleep Deprivation in the Rat Mediated by Bacterial Invasion? Sleep 1996, 19, 554–562.
  9. Everson, C.A.; Toth, L.A. Systemic bacterial invasion induced by sleep deprivation. Am. J. Physiol. Integr. Comp. Physiol. 2000, 278, R905–R916.
  10. Bergmann, B.M.; Rechtschaffen, A.; Gilliland, M.A.; Quintans, J. Effect of extended sleep deprivation on tumor growth in rats. Am. J. Physiol. Integr. Comp. Physiol. 1996, 271, R1460–R1464.
  11. Wilkinson, R.T. Sleep Deprivation. In Physiology of Survival; Endholme, E.G., Bacharach, A.L., Eds.; London Academic Press: London, UK, 1965; pp. 399–430.
  12. Horne, J.A. Sleep function, with particular reference to sleep deprivation. Ann. Clin. Res. 1985, 17, 199–208.
  13. Horne, J.A. A review of the biological effects of total sleep deprivation in man. Biol. Psychol. 1978, 7, 55–102.
  14. Sharma, S.; Kavuru, M. Sleep and Metabolism: An Overview. Int. J. Endocrinol. 2010, 2010, 270832.
  15. Sun, Q.; Liu, Y.; Wei, W.; Wu, D.; Lin, R.; Wen, D.; Jia, L. Chronic Timed Sleep Restriction Attenuates LepRb-Mediated Signaling Pathways and Circadian Clock Gene Expression in the Rat Hypothalamus. Front. Neurosci. 2020, 14, 909.
  16. Roth, T. Insomnia: Definition, prevalence, etiology, and consequences. J. Clin. Sleep Med. 2007, 15 (Suppl. S5), S7–S10.
  17. Ferrie, J.E.; Kumari, M.; Salo, P.; Singh-Manoux, A.; Kivimaki, M. Sleep epidemiology--A rapidly growing field. Int. J. Epidemiol. 2011, 40, 1431–1437.
  18. Léger, D.; du Roscoat, E.; Bayon, V.; Guignard, R.; Pâquereau, J.; Beck, F. Short sleep in young adults: Insomnia or sleep debt? Prevalence and clinical description of short sleep in a representative sample of 1004 young adults from France. Sleep Med. 2011, 12, 454–462.
  19. Kronholm, E.; Partonen, T.; Laatikainen, T.; Peltonen, M.; Härmä, M.; Hublin, C.; Kaprio, J.; Aro, A.R.; Partinen, M.; Fogelholm, M.; et al. Trends in self-reported sleep duration and insomnia-related symptoms in Finland from 1972 to 2005: A comparative review and re-analysis of Finnish population samples. J. Sleep Res. 2008, 17, 54–62.
  20. Rowshan Ravan, A.; Bengtsson, C.; Lissner, L.; Lapidus, L.; Björkelund, C. Thirty-six-year secular trends in sleep duration and sleep satisfaction, and associations with mental stress and socioeconomic factors--results of the Population Study of Women in Gothenburg, Sweden. J. Sleep Res. 2010, 19, 496–503.
  21. Kant, G.J.; Genser, S.G.; Thorne, D.R.; Pfalser, H.L.; Mougey, E.H. Effects of 72 Hour Sleep Deprivation on Urinary Cortisol and Indices of Metabolism. Sleep 1984, 7, 142–146.
  22. Dement, W.C. The effect of dream deprivation. Science 1960, 131, 1705–1707.
  23. Sampson, H. Psychological effects of deprivation of dreaming sleep. J. Nerv. Ment. Dis. 1966, 143, 305–317.
  24. Vogel, G.W. A Review of REM Sleep Deprivation. Arch. Gen. Psychiatry 1975, 32, 749–761.
  25. Wyatt, R.J.; Fram, D.H.; Buchbinder, R.; Snyder, F. Treatment of Intractable Narcolepsy with a Monoamine Oxidase Inhibitor. N. Engl. J. Med. 1971, 285, 987–991.
  26. McGrath, M.J.; Cohen, D.B. REM sleep facilitation of adaptive waking behavior: A review of the literature. Psycol. Bull. 1978, 85, 24–57.
  27. Maquet, P. The Role of Sleep in Learning and Memory. Science 2001, 294, 1048–1052.
  28. Liu, Y.; Wheaton, A.G.; Chapman, D.P.; Cunningham, T.J.; Lu, H.; Croft, J.B. Prevalence of Healthy Sleep Duration among Adults—United States, 2014. MMWR. Morb. Mortal. Wkly. Rep. 2016, 65, 137–141.
  29. St-Onge, M.-P.; Grandner, M.A.; Brown, D.; Conroy, M.B.; Jean-Louis, G.; Coons, M.; Bhatt, D.L. Sleep Duration and Quality: Impact on Lifestyle Behaviors and Cardiometabolic Health: A Scientific Statement From the American Heart Association. Circulation 2016, 134, e367–e386.
  30. Covassin, N.; Singh, P. Sleep duration and cardiovascular disease risk: Epidemiologic and experimental evidence. Sleep Med. Clin. 2016, 11, 81–89.
  31. Itani, O.; Jike, M.; Watanabe, N.; Kaneita, Y. Short sleep duration and health outcomes: A systematic review, meta-analysis, and meta-regression. Sleep Med. 2017, 32, 246–256.
  32. Sato, M.; Matsuo, T.; Atmore, H.; Akashi, M. Possible contribution of chronobiology to cardiovascular health. Front. Physiol. 2014, 4, 409.
  33. Davignon, J.; Ganz, P. Role of Endothelial Dysfunction in Atherosclerosis. Circulation 2004, 109, III-27–III-32.
  34. Malerba, M.; Clini, E.; Malagola, M.; Avanzi, G.C. Platelet activation as a novel mechanism of atherothrombotic risk in chronic obstructive pulmonary disease. Expert Rev. Hematol. 2013, 6, 475–483.
  35. Malerba, M.; Radaeli, A.; Nardin, M.; Clini, E.; Carpagnano, G.E.; Sciatti, E.; Salghetti, F.; Bonadei, I.; Platto, F.; Vizzardi, E. Endothelial dysfunction assessment by noninvasive peripheral arterial tonometry in patients with chronic obstructive pulmonary disease compared with healthy subjects. Clin. Respir. J. 2018, 12, 1466–1472.
  36. Ras, R.T.; Streppel, M.T.; Draijer, R.; Zock, P.L. Flow-mediated dilation and cardiovascular risk prediction: A systematic review with meta-analysis. Int. J. Cardiol. 2013, 168, 344–351.
  37. Inaba, Y.; Chen, J.A.; Bergmann, S.R. Prediction of future cardiovascular outcomes by flow-mediated vasodilatation of brachial artery: A meta-analysis. Int. J. Cardiovasc. Imaging 2010, 26, 631–640.
  38. Malerba, M.; Nardin, M.; Radaeli, A.; Montuschi, P.; Carpagnano, G.E.; Clini, E. The potential role of endothelial dysfunction and platelet activation in the development of thrombotic risk in COPD patients. Expert Rev. Hematol. 2017, 10, 821–832.
  39. Malerba, M.; Olivini, A.; Radaeli, A.; Ricciardolo, F.L.M.; Clini, E. Platelet activation and cardiovascular comorbidities in patients with chronic obstructive pulmonary disease. Curr. Med Res. Opin. 2016, 32, 885–891.
  40. Polosa, R.; Malerba, M.; Cacciola, R.R.; Morjaria, J.B.; Maugeri, C.; Prosperini, G.; Gullo, R.; Spicuzza, L.; Radaeli, A.; Di Maria, G.U. Effect of acute exacerbations on circulating endothelial, clotting and fibrinolytic markers in COPD patients. Intern. Emerg. Med. 2013, 8, 567–574.
  41. Matsuzawa, Y.; Kwon, T.; Lennon, R.J.; Lerman, L.O.; Lerman, A. Prognostic Value of Flow-Mediated Vasodilation in Brachial Artery and Fingertip Artery for Cardiovascular Events: A Systematic Review and Meta-Analysis. J. Am. Heart Assoc. 2015, 4, e002270.
  42. Hanson, J.A.; Huecker, M.R. Sleep Deprivation; StatPearls Publishing: Treasure Island, FL, USA, 2019.
More
Video Production Service