<<

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector

Available online at www.sciencedirect.com

ScienceDirect

Impact of on and energy balance

Cristina Rabasa and Suzanne L Dickson

The stress response mobilizes the body’s energy stores in order to stress response. The dorsal cap and submagnocellular

respond to a threatening situation. A striking observation is the part of the PVN send projections to the brainstem and

diversity of metabolic changes that can occur in response to spinal nuclei controlling sympathetic and parasympa-

stress. On one hand acute intense stress is commonly associated thetic activity. During stress, activation of the sympa-

with feeding suppression and reduced body weight gain. The thetic (SNS) stimulates epinephrine and

activation of the hypothalamic–pituitary–adrenal axis and the a smaller, although significant norepinephrine release

release of corticotropin-releasing hormone (CRH) might partially from the , as well as norepinephrine

explain the anorexigenic effects of acute stress. CRH can also release from the sympathetic nerve endings [1]. These

stimulate the sympathetic nervous system and catecholamine catecholamines stimulate a(1–2) and b(1–3) receptors

release, inducing hypophagia and weight loss, through their expressed in different central and peripheral targets to

effects on the liver and on white and brown adipose tissue. On the coordinate cardiovascular responses (e.g. cardiac output

other hand, chronic stress can lead to dietary over-consumption and respiration are accelerated), redirect blood flow to

(especially palatable foods), increased visceral adiposity and arouse the brain and prepare the muscles and the

weight gain. These obesogenic effects of stress are mainly for a ‘fight or flight’ response. The parvocellular part of

explained by the chronic release of glucocorticoids and the PVN is the origin of neurons sending axons to the

neuropeptide Y. Stressful situations can activate all of these median eminence, where they release corticotropin re-

systems together, and the metabolic outcome of stress exposure leasing hormone (CRH) thereby activating the hypotha-

is determined by a host of intrinsic and external factors. If we are lamic–pituitary adrenal axis (HPA). CRH reaches the

to find new ways to limit the development of stress-linked and stimulates the synthesis and release

cardiometabolic diseases, we need to discover why, in some of adrenocorticotropic hormone (ACTH). Circulating

circumstances, the pro-obesogenic effects of stress outweigh its ACTH acts on melanocortin 2 (MC-2) receptors in the

anorexigenic effects. The equilibrium between the different

zona fasciculata of the , stimulating the

components of the stress response that accompany chronic

synthesis and release of glucocorticoids ( in

stress situations could be crucial to understand and prevent the

humans and corticosterone in rodents). These act in

long-lasting adverse metabolic effects induced by stress.

several tissues through the mineralocorticoid and gluco-

Address corticoid receptors (MR and GR) and exert negative

Department of Physiology, Institute of Neuroscience and Physiology, feedback on the HPA axis suppressing the stress re-

The Sahlgrenska Academy at the University of Gothenburg, sponse [2].

Medicinaregatan 11, PO Box 434, SE-405 30 Gothenburg, Sweden

Collectively all of the stress-linked changes in the SNS

Corresponding author: Rabasa, Cristina ([email protected])

and HPA axes mobilize the body’s energy stores in order

to allow a rapid response that favors survival under acute

Current Opinion in Behavioral Sciences 2016, 9:71–77 threatening situations. However, modern life stressors

This review comes from a themed issue on Diet, behavior and brain are typically chronic, including financial worries, work

function problems, family responsibilities or health concerns [3].

Edited by Dana M Small and Susanne E la Fleur In this context, the stress response is not beneficial but

rather becomes harmful, predisposing individuals to

For a complete overview see the Issue and the Editorial

visceral obesity and cardiometabolic disease [4,5], a

Available online 7th February 2016

leading cause of death worldwide. Still, the effects of

http://dx.doi.org/10.1016/j.cobeha.2016.01.011

stress on body weight and food intake are controversial.

2352-1546/# 2016 The Authors. Published by Elsevier Ltd. This is an Although there is a ink between stress and changes in

open access article under the CC BY-NC-ND license (http://creative-

eating habits in both men and women [6], not everyone

commons.org/licenses/by-nc-nd/4.0/).

gains weight under stress: about 40% increase and 40%

decrease their food intake [7]. Many studies have con-

tributed to understand the individual differences be-

Relevance of the stress response and stress tween those that gain and those that lose weight [6,8–

hormones for energy balance 10]. However, other factors like the chronicity and

Stress is the response of an to factors actually or intensity of the stressors, factors that are difficult to

symbolically endangering its integrity. Irrespective of manipulate in humans, can be of major importance and

the type of stress, information about the situation con- could help to understand the physiological and neuro-

verges on the paraventricular nucleus (PVN) of the biological mechanisms underpinning the metabolic re-

, a brain region crucially involved in the sponse to stress.

www.sciencedirect.com Current Opinion in Behavioral Sciences 2016, 9:71–77

72 Diet, behavior and brain function

Figure 1

In this review we attempt to untangle the complex

relationship between stress and food intake, which tends

to favor an anorexigenic response when the stress is acute Control - NS IMOch - NS CUS - NS

Control - IMOa IMOch - IMOa CUS - IMOa

and severe. We adopt the view that under chronic or 30

repeated stress, (neuro)physiological adaptations occur

that give way to an orexigenic response. We describe 20

some of the known mechanisms and signaling systems

10

and highlight the important role of CRH, glucocorticoids

and NPY. Given that a number of recent reviews describe

0

the interaction between obesity and stress, we limit this

Body weight change (g) PS1 PS2 PS3 PS4

review to specific, perhaps less understood aspects of this –10

relationship that include the interactions between the Days after the last stress exposure

anorexigenic and orexigenic effectors of the HPA and Current Opinion in Behavioral Sciences

SNS axes during the stress response.

Influence of prior chronic stress exposure on body weight change

Severe stress induces anorexia after an acute immobilization (IMO). Rats were exposed to 9 days of

The most commonly reported result in rodents is that handling (control), chronic unpredictable stress (CUS) or chronic

immobilization (IMOch). On day 10, half of the rats in each group were

stress reduces food intake and body weight in a manner

exposed to handling (non-stress condition; Control-NS, IMOch-NS and

that is directly related to the stress severity [11]. A single

CUS-NS), or acutely exposed to 1 h of IMO (control-IMOa, IMOch-

exposure to restraint [12] or immobilization [13] induces a IMOa and CUS-IMOa). Data are represented as means + S.E.M.

reduction in body weight that can persist >10 days, even (n = 6–9) of cumulative body weight changes, relative to the body

weight achieved before the acute IMO exposure (day 10). Only the

if the food intake is quickly normalized [12,13]. More-

rats exposed to stress for the first time (Control-IMOa) lose weight

over, these body weight-lowering effects can be extreme-

after IMO ( p < 0.001 vs Control-NS group), while rats with either

ly long lasting after repeated exposure to the stressor.

chronic IMO or CUS are protected from the body weight reduction

Strikingly, rats do not catch up to the weight of non-stress induced by IMO. PS indicates the day post-stress (Pastor-Ciurana



controls even by 80 days after 3 consecutive days of et al. [15 ]).

restraint [14]. Thus, after stress, rats appear to have a

capacity to defend a new lower set-point for energy

homeostasis despite returning to normal caloric intake potent anorexigenic effects [18,19]. Despite this, CRHR1

and so energy expenditure must also be adjusted. How- or CRHR2 knockout mice do not differ from wildtypes

ever, even although chronically stressed rats do not catch regarding their food intake and body weight under non-

up to the weight of control never-stressed rats, prelimi- stressed conditions, suggesting that CRH receptors are

nary observations from our group demonstrate that rats not needed for the basal regulation of these parameters

exposed to 1 h/day of forced swim lose weight only on the [20,21]. However, severe impairments in the HPA axis

first days of stress exposure, but then start to gain weight response have been observed in CRHR1 knockout mice

afterwards. Interestingly, this chronic stress-linked exposed to restraint, social defeat or forced swim stress

weight gain occurs despite a reduction in food intake, [20,22] but only minor alterations in the CRHR2 knock-

suggesting that metabolic adaptations occur during stress out mice [20,23]. The lack of CRHR1 also protects

which increase caloric efficiency. Supportively, Pastor-Ciur- against stress-induced hypophagia [24]. Consistent with



ana et al. [15 ] demonstrated that prior chronic unpredict- this, delivery of a non-specific CRH antagonist into the

able or repeated stress experience protects rats from the third [25], but not in the lateral or the fourth ventricle

anorectic effects of a subsequent acute and intense [25,26] prevents the weight loss induced by stress, impli-

stressor like immobilization (Figure 1). Therefore, al- cating the hypothalamus as the likely target for this effect.

though intense stress seems to induce immediate body Indeed, this lends support to the hypothesis that the

weight loss, compensatory mechanisms exist that restore effects of stress on weight loss involve CRH release

energy balance. within the PVN and the inhibition of NPY-neurons

[27]. However, infusions of CRHR2 antagonists in the

Short-term anorexia: a role for CRH and Urocortins bed nucleus of the stria terminalis (BNST) and lateral

The CRH system initiates the HPA axis response to stress septum (LS) [28] or CRHR1 antagonists to the basolateral

and it has been extensively linked to stress-induced amygdala (BLA) [29] can also prevent stress-induced

anorexia. CRH and its analogs Urocortins (UCN) 1–3 anorexia. It should be noted, however, that these benefi-

act as neurotransmitters and activate CRHR1 and cial effects of the CRH antagonists are rather mild and

CRHR2 receptors that have a role in food intake, weight short lived in the stress condition [30]; most studies report



gain and metabolic control [16 ,17]. Under basal condi- effects at 1-hour post stress in food-deprived rats [31,32].

tions, peripheral and i.c.v. injections of CRH or Urocor- Future studies should aim to follow the effects of these

tins decrease food intake and body weight gain for up to CRH receptor antagonist drugs for a longer period after

48 hours after injection, with Ucn1 having the most the stress.

Current Opinion in Behavioral Sciences 2016, 9:71–77 www.sciencedirect.com

Stress and metabolism Rabasa and Dickson 73

Body weight reduction and energetic status forced swim induce very strong stress responses, and it is

It is widely accepted that manipulations causing in- possible that these rodent stressors are modeling a se-

creased sympathetic tone (such as that induced by stress) verely traumatic situation. In addition, when these stress-

are linked to increased thermogenesis in brown adipose ors are repeatedly presented, rodents adapt very fast [50].

tissue (BAT), decreased food intake and body weight Human studies typically involve milder but chronic

[33]. Catecholamines stimulate lipolysis and the release stressors [3], and they demonstrate that stress can increase

of fatty acids from the adipose tissue, and also glycogen- body weight and preference for palatable food in many

olysis and gluconeogenesis in the liver [34,35]. CRH and ways [9,10,51,52]. Stress engages pathways that confer the

ACTH contribute to stimulate the SNS and the noradren- reward value of foods, such as the mesocorticolimbic

aline release [36,37], and extrahypothalamic brain areas dopamine pathway. By activating this pathway, glucocor-

containing CRH immunoreactive neurons, like BNST ticoids increase motivating aspects of drugs [53], and

and central amygdala, participate through the simulation potentially also for palatable food. In this regard, the

of the locus coeruleus (LC) [38]. Energy expenditure ‘Comfort Food’ hypothesis proposed by Dallman [54]

increases while the respiratory quotient decreases during suggests that eating palatable high fat/sugar food can

restraint or chronic mild stress [14,39]. This increase in relieve negative emotions linked to stress and reduce

energy expenditure is coupled to an increase in body and the HPA axis response [52,54,55]. Firstly, it was sug-

BAT temperature under stress in rodents [36,40,41], gested that the visceral fat might act as a signal of energy



mediated by the glutamatergic stimulation of the dor- stores that feeds back to inhibit the HPA axis [54,56 ],

somedial hypothalamus to the rostral medullary raphe but this hypothesis has been updated. Using a model of

region (rMR) that participates in the sympathetic stimu- limited access to sucrose, which reduces the stress re-



lation of BAT [40]. Stress-linked can be sponse, Ulrich-Lai et al. [57 ] propose that sucrose intake

prevented with a receptor agonist of 5-HT1A in the rMR, alters signaling between the PVN and the basolateral/

by CRH antagonists delivery to the brain ventricles [36] medial amygdala on one side and the BNST on the other,

and also by antagonists to b-3 adrenoceptor highly resulting in a diminished response to stress.

expressed in BAT [40–42]. Indeed, stress hyperthermia

seems to be mediated by BAT thermogenesis. Moreover, Apart from increasing the motivation to eat palatable

acute and chronic immobilization increase UCP-1 activity food, increasing visceral obesity is one way in which

in BAT, and the effect is stronger after chronic stress [43]; chronic stress increases the risk of metabolic syndrome,

this fits with the increased response of BAT thermogen- since abdominal obesity is one of its core symptoms

esis to norepinephrine after repeated stress [44]. On the (together with hypertension, high fasting glucose levels

other hand, GCs can oppose these effects; dexametha- and triglycerides and low HDL cholesterol levels), meta-

sone inhibits adrenergic stimulation of the functioning of bolic alterations strongly related with the development of

human brown adipocytes [45], and corticosterone attenu- type-2 diabetes and cardiovascular disease [58].

ates the stimulatory effects of ACTH and norepinephrine

on BAT (increase UCP1 expression) in vitro and in vivo in Acute and chronic effects of glucocorticoids

mice through the GR receptors [37]. Glucocorticoids (GC) have a complex role in energy

balance and metabolic control. Acutely, they are con-

The catabolic effects of SNS activation during stress are sidered catabolic hormones, that increase lipolysis and

not only observed in BAT but also in white adipose tissue the release of fatty acids from WAT [59], and stimulate

(WAT). In WAT, the release of catecholamines under hepatic glycogenolysis and gluconeogenesis, increasing

stress and the consequent stimulation of b adrenergic the circulating glucose concentrations and the glycogen

receptors increases lipolysis and inhibits the proliferation deposition [60]. However, an anabolic effect of GCs



of preadipocytes [46], while the lack of norepinephrine within WAT has been demonstrated [61 ]. People suf-

increases the fat cell number and the WAT mass [47]. In fering from Cushing syndrome, endogenous hypercor-

addition, repeated immobilization stimulates catechol- tisolism, have abdominal obesity, increased body fat,

amine biosynthesis in visceral WAT [48], and chronic high blood pressure, insulin resistance and preference



cold stress [49 ] or in vitro culture with ACTH [37] can for fat food [62,63]. It has been hypothesized that

change the WAT to BAT-like morphology, with high insulin, frequently released as a consequence of GC

expression levels of UCP-1 perhaps increasing thermo- action [64–66], might be crucial in food choice and fat

genesis. accumulation in stress [67]. In absence of insulin, GC

increase the intake of chow, rich in carbohydrates [65],

Chronic stress increases body weight and risk while in presence of insulin, GC increase the preference

for cardiometabolic disease for more palatable options [68,69]. However, when the

Whereas in rodents stress usually induces weight loss, in GC levels are constantly high as is the case of chronic

humans, this effect is not always observed, and depends stress, insulin resistance is observed. Thus, other

upon which experimental designs are used to explore the factors have to be responsible for the fat preference

stressor. In rodents, the classic stressors like restraint, or in the long term.

www.sciencedirect.com Current Opinion in Behavioral Sciences 2016, 9:71–77

74 Diet, behavior and brain function

Figure 2

Brain Food intake

Body weight PVN BNST Motivation to eat palatable food AMY Liver CRH Gluconeogenesis Glycogenolysis

BAT ACTH BAT induced

Sympathetic neuron thermogenesis Glucocorticoids Preadipocyte differentiation Stress hyperthermia

Epinephrine WAT Norepinephrine Lipolysis Preadipocyte NPY differentiation Adipocyte

proliferation Stimulation Inhibition

Current Opinion in Behavioral Sciences

The different systems involved in the stress effects on energy balance and their stimulatory and inhibitory role in different organs. PVN:

paraventricular nucleus of the hypothalamus; AMY: amygdale; BNST: bed nucleus of the stria terminalis; CRH: corticotropin-releasing hormone;

ACTH: adrenocorticotropic hormone; NPY: neuropeptide Y; BAT: brown adipose tissue; WAT: white adipose tissue.

Glucocorticoids also affect BAT, which is involved in of the SNS, from where it is released as an adrenergic

and energy expenditure. BAT-induced cotransmitter under some stress situations [73]. Centrally,

thermogenesis can contribute to a reduction in body fat NPY potently increases food intake through its effects in

[70]. GCs appear to be required for BAT cell differentia- the hypothalamus, that can be considered a fight against

tion (in vitro) [45,71] yet in high concentrations, gluco- starvation under stress. Additionally, central NPY exerts

corticoids have been shown to impair BAT-induced protective effects under stress by reducing anxiety and

thermogenesis in vivo [72]. Consistent with this, GCs depressive-like behaviors [74] and can increase fat intake

inhibits the adrenergic stimulation of the functioning of when is infused in the Nucleus Accumbens [75]. Simulta-

the brown adipocytes in vitro [45,71]. Therefore, GCs neously, injections of a synthetic GC or one session of

may impact on energy balance through their actions in footshock increase NPY content in the ARC, and decrease

WAT and BAT. However, under stressful situations, the amount of CRH in the mediobasal hypothalamus

other factors like the activation of the SNS can oppose [64,76], both of which could contribute to a feeding re-

these effects, and the net result can depend on the sponse. Likewise, chronic restraint increases the expres-

combination of both systems. sion of the orexigenic peptide agouti-related peptide

(AgRP, which colocalises with NPY) in the lateral hypo-

Neuropeptide Y is engaged in the stress response at thalamus and down-regulates the melanocortin receptor 4

many levels (MC4) in ARC, that responds to the anorexigenic proo-

Another important peptide linking stress and adiposity is piomelanocortin (POMC) [77], potentiating orexigenic

neuropeptide Y (NPY), a peptide produced both in the systems. However, a parallel stimulation of anorexigenic

arcuate nucleus (ARC) of the hypothalamus and in neurons pathways has been also observed in the ARC, since acute

Current Opinion in Behavioral Sciences 2016, 9:71–77 www.sciencedirect.com

Stress and metabolism Rabasa and Dickson 75

footshock or restraint down-regulates AgRP [76,77], and different types of stress with food intake and energy

acute restraint increases the expression of POMC in the balance will facilitate the development of strategies that

whole hypothalamus [78]. circumvent the development of stress-linked metabolic

disease.

NPY can be released as an adrenergic cotransmitter,

acting by itself or modulating the effects of norepineph-

Conflict of interest statement

rine and ATP, also released from the sympathetic term-

Nothing declared.

inals. For example, in WAT, NPY appears to oppose the

effects of norepinephrine, inhibiting lipolysis induced by

 Acknowledgements

a b-agonist in cultured adipocytes [79]. Kuo et al. [49 ]

We gratefully acknowledge support from the EC projects Nudge-it and

demonstrated that NPY is upregulated in WAT after

Full4Health (under grant agreement numbers 607310 and 266408

chronic cold and social defeat stress in mice fed the high respectively), from the Swedish Research Council for Medicine (2012-1758)

and from Forskning och Utveklingsarbete (ALFGBG-138741). Thanks to

fat-high sucrose diet. Besides, the increase in abdominal

Dr Erik Schele for his help with the design of the Figure 2, and to Prof

fat and the symptoms resembling metabolic syndrome in

John-Olov Jansson for reading and commenting on the text.

these mice were reversed by delivery of a NPY2R antag-

onist to the abdominal fat. They show that NPY increases

References and recommended reading

under stress, and stimulates angiogenesis and adipogen-

Papers of particular interest, published within the period of review,

esis through NPY2R. Moreover, when sympathetic neural have been highlighted as:

cells are treated with dexamethasone, the NPY expres-

  of special interest

sion is remarkedly increased [49 ], and could be one of

 of outstanding interest

the mechanisms explaining some of the obesogenic

effects of GCs. 1. Kvetnansky R, Sabban EL, Palkovits M: Catecholaminergic

systems in stress: structural and molecular genetic

approaches. Physiol Rev 2009, 89:535-606.

Conclusions: is stress altering energy

balance? 2. Armario A: The hypothalamic–pituitary–adrenal axis: what can

it tell us about stressors? CNS Neurol Disord Drug Targets 2006,

It seems evident that stress can impact on energy balance 5:485-501.

in many diverse ways and at different levels (Figure 2). 3. American Psychological Association. Stress in America, Paying

with our health. http://www.apa.org/news/press/releases/stress/

Stress can trigger both orexigenic-like and anorexigenic-

2014/stress-report.pdf 2014.

like responses reflecting a variety of intrinsic and external

4. Rosmond R: Role of stress in the pathogenesis of the

factors such as individual differences, (palatable) food

metabolic syndrome. Psychoneuroendocrinology 2005, 30:1-10.

availability and/or the type of stress. Chronic and acute

5. von Kanel R: Psychosocial stress and cardiovascular risk:

stressors recruit some overlapping but also divergent

current opinion. Swiss Med Wkly 2012, 142:w13502.

systems relevant for metabolic control. From an evolu-

6. Adam TC, Epel ES: Stress, eating and the reward system.

tionary perspective, it seems reasonable that chronic

Physiol Behav 2007, 91:449-458.

stressors promote the activation of pro-obesogenic mech-

7. NPR/Robert Wood Johnson Foundation. Harvard School of Public

anisms that favor the accumulation of central/visceral fat. Health. The burden of stress in America. http://media.npr.org/

documents/2014/july/npr_rwfj_harvard_stress_poll.pdf. 2014.

This is because constant activation of the stress response

can be interpreted as living in a constant unsafe situation, 8. Weinstein SE, Shide DJ, Rolls BJ: Changes in food intake in

response to stress in men and women: psychological factors.

for which it is beneficial to store central (easy to use) fat

Appetite 1997, 28:7-18.

and glycogen in the liver and hence, help supply the brain

9. Wardle J, Steptoe A, Oliver G, Lipsey Z: Stress, dietary restraint

with energy on short demand. This apparently simple

and food intake. J Psychosomatic Res 2000, 48:195-202.

idea is difficult to demonstrate experimentally in rodents,

10. Epel E, Lapidus R, McEwen B, Brownell K: Stress may add bite to

probably because classical stressors do not mimic the appetite in women: a laboratory study of stress-induced

human situations and it is difficult to find direct compar- cortisol and eating behavior. Psychoneuroendocrinology 2001,

26:37-49.

isons between acute and chronic stress. Most rodent stress

11. Martı´ O, Martı´ J, Armario A: Effects of chronic stress on food

models that increase adiposity like social crowding [80]

  intake in rats: influence of stressor intensity and duration of

social defeat [49 ,81] or cold stress [49 ] seem to po- daily exposure. Physiol Behav 1994, 55:747-753.

tently stimulate NPY secretion, that could be an impor-

12. Rybkin II, Zhou Y, Volaufova J, Smagin GN, Ryan DH, Harris RBS:

tant factor in future studies that attempt to model the Effect of restraint stress on food intake and body weight is

determined by time of day. Am J Physiol Regul Integr Comp

metabolic alterations seen in humans. Future experi-

Physiol 1997, 273:R1612-R1622.

ments aimed to directly compare the acute and chronic

13. Valle` s A, Martı´ O, Garcı´a A, Armario A: Single exposure to

effects of these stressors on the HPA and SNS interac-

stressors causes long-lasting, stress-dependent reduction of

tions regarding energy balance are needed. It is impera- food intake in rats. Am J Physiol Regul Integr Comp Physiol

2000:R1138-R1144.

tive to know whether differences in the adaptations of the

anorexigenic and orexigenic responses along chronic 14. Harris RB, Palmondon J, Leshin S, Flatt WP, Richard D: Chronic

disruption of body weight but not of stress peptides or

stress can explain the final impact on metabolism. Ulti-

receptors in rats exposed to repeated restraint stress. Horm

mately, an increased understanding of the links between Behav 2006, 49:615-625.

www.sciencedirect.com Current Opinion in Behavioral Sciences 2016, 9:71–77

76 Diet, behavior and brain function

15. Pastor-Ciurana J, Rabasa C, Ortega-Sanchez JA et al.: Prior 32. Sekino A, Ohata H, Mano-Otagiri A, Arai K, Shibasaki T: Both

 exposure to repeated immobilization or chronic unpredictable corticotropin-releasing factor receptor type 1 and type 2 are

stress protects from some negative sequels of an acute involved in stress-induced inhibition of food intake in rats.

immobilization. Behav Brain Res 2014, 265:155-162. Psychopharmacology (Berl) 2004, 176:30-38.

This paper demonstrates that chronic stress can protect from the nega-

tive consequences of an intense but acute stressor. They show that after 33. Bray GA: Reciprocal relation of food intake and sympathetic

chronic stress, the rats do not show the anorectic effects expected after activity: experimental observations and clinical implications.

immobilization, suggesting metabolic adaptations that protect from the Int J Obes Relat Metab Disord 2000, 24(Suppl2):S8-S17.

stress induced reductions in the body weight.

34. Bartness TJ, Shrestha YB, Vaughan CH, Schwartz GJ, Song CK:

16. Harris RB: Chronic and acute effects of stress on energy Sensory and sympathetic nervous system control of white

 balance: are there appropriate animal models? Am J Physiol adipose tissue lipolysis. Mol Cell Endocrinol 2010, 318:34-43.

Regul Integr Comp Physiol 2015, 308:R250-R265.

35. Sherwin RS, Sacca L: Effect of epinephrine on glucose

Review on the effects of stress in metabolism and energy balance. This

metabolism in humans: contribution of the liver. Am J Physiol

review present the different animal models to study the metabolic altera-

Endocrinol Metab 1984, 247:E157-E165.

tions occurred after chronic and acute stress, and whether they induce

reduction or increase in the body weight gain.

36. Morimoto A, Nakamori T, Morimoto K, Tan N, Murakami N: The

central role of corticotrophin-releasing factor (CRF-41) in

17. Stengel A, Tache´ YF: CRF and urocortin peptides as

in rats. J Physiol 1993, 460:221-229.

modulators of energy balance and feeding behavior during

stress. Front Neurosci 2014:8.

37. van den Beukel JC, Grefhorst A, Quarta C et al.: Direct activating

effects of adrenocorticotropic hormone (ACTH) on brown

18. Spina M, Merlo-Pich E, Chan RK, Basso AM, Rivier J, Vale W,

adipose tissue are attenuated by corticosterone. FASEB J

Koob GF: Appetite-suppressing effects of urocortin, a CRF-

2014, 28:4857-4867.

related neuropeptide. Science 1996, 273:1561-1564.

38. Kova´ cs KJ: CRH: the link between hormonal-, metabolic- and

19. Tanaka C, Asakawa A, Ushikai M et al.: Comparison of the

behavioral responses to stress. J Chem Neuroanat 2013, 54:25-33.

anorexigenic activity of CRF family peptides. Biochem Biophys

Res Commun 2009, 390:887-891.

39. Garcia-Diaz DF, Campion J, Milagro FI, Lomba A, Marzo F,

Martinez JA: Chronic mild stress induces variations in

20. Preil J, Muller MB, Gesing A et al.: Regulation of the

locomotive behavior and metabolic rates in high fat fed rats. J

hypothalamic–pituitary–adrenocortical system in mice

Physiol Biochem 2007, 63:337-346.

deficient for CRH receptors 1 and 2. Endocrinology 2001,

142:4946-4955.

40. Kataoka N, Hioki H, Kaneko T, Nakamura K: Psychological stress

activates a dorsomedial hypothalamus-medullary raphe

21. Muller MB, Keck ME, Zimmermann S, Holsboer F, Wurst W:

circuit driving brown adipose tissue thermogenesis and

Disruption of feeding behavior in CRH receptor 1-deficient

hyperthermia. Cell Metab 2014, 20:346-358.

mice is dependent on glucocorticoids. Neuroreport 2000,

11:1963-1966.

41. Lkhagvasuren B, Nakamura Y, Oka T, Sudo N, Nakamura K: Social

defeat stress induces hyperthermia through activation of

22. Muller MB, Landgraf R, Preil J, Sillaber I, Kresse AE, Keck ME,

thermoregulatory sympathetic premotor neurons in the

Zimmermann S, Holsboer F, Wurst W: Selective activation of the

medullary raphe region. Eur J Neurosci 2011, 34:1442-1452.

hypothalamic vasopressinergic system in mice deficient for

the corticotropin-releasing hormone receptor 1 is dependent

42. Mohammed M, Ootsuka Y, Blessing W: Brown adipose tissue

on glucocorticoids. Endocrinology 2000, 141:4262-4269.

thermogenesis contributes to emotional hyperthermia in a

23. Bale TL, Contarino A, Smith GW, Chan R, Gold LH, Sawchenko PE, resident rat suddenly confronted with an intruder rat. Am J

Koob GF, Vale WW, Lee KF: Mice deficient for corticotropin- Physiol Regul Integr Comp Physiol 2014, 306:R394-R400.

releasing hormone receptor-2 display anxiety-like behaviour

43. Gao B, Kikuchi-Utsumi K, Ohinata H, Hashimoto M, Kuroshima A:

and are hypersensitive to stress. Nat Genet 2000, 24:410-414.

Repeated immobilization stress increases uncoupling protein

24. Chotiwat C, Kelso EW, Harris RB: The effects of repeated 1 expression and activity in Wistar rats. Jpn J Physiol 2003,

restraint stress on energy balance and behavior of mice with 53:205-213.

selective deletion of CRF receptors. Stress 2010, 13:203-213.

44. Nozu T, Okano S, Kikuchi K, Yahata T, Kuroshima A: Effect of

25. Smagin GN, Howell LA, Redmann S Jr, Ryan DH, Harris RB: immobilization stress on in vitro and in vivo thermogenesis of

Prevention of stress-induced weight loss by third ventricle brown adipose tissue. Jpn J Physiol 1992, 42:299-308.

CRF receptor antagonist. Am J Physiol 1999, 276:R1461-R1468.

45. Barclay JL, Agada H, Jang C, Ward M, Wetzig N, Ho KK: Effects of

26. Miragaya JR, Harris RB: Antagonism of corticotrophin- glucocorticoids on human brown adipocytes. J Endocrinol

releasing factor receptors in the fourth ventricle modifies 2015, 224:139-147.

responses to mild but not restraint stress. Am J Physiol Regul

46. Jones DD, Ramsay TG, Hausman GJ, Martin RJ: Norepinephrine

Integr Comp Physiol 2008, 295:R404-R416.

inhibits rat pre-adipocyte proliferation. Int J Obes Relat Metab

27. Heinrichs SC, Menzaghi F, Pich EM, Hauger RL, Koob GF: Disord 1992, 16:349-354.

Corticotropin-releasing factor in the paraventricular nucleus

47. Bowers RR, Festuccia WT, Song CK, Shi H, Migliorini RH,

modulates feeding induced by neuropeptide Y. Brain Res 1993,

611:18-24. Bartness TJ: Sympathetic innervation of white adipose tissue

and its regulation of fat cell number. Am J Physiol Regul Integr

28. Ohata H, Shibasaki T: Involvement of CRF2 receptor in the brain Comp Physiol 2004, 286:R1167-R1175.

regions in restraint-induced anorexia. Neuroreport 2011,

22:494-498. 48. Vargovic P, Ukropec J, Laukova M, Kurdiova T, Balaz M, Manz B,

Ukropcova B, Kvetnansky R: Repeated immobilization stress

29. Jochman KA, Newman SM, Kalin NH, Bakshi VP: Corticotropin- induces catecholamine production in rat mesenteric

releasing factor-1 receptors in the basolateral amygdala adipocytes. Stress 2013, 16:340-352.

mediate stress-induced anorexia. Behav Neurosci 2005,

119:1448-1458. 49. Kuo LE, Kitlinska JB, Tilan JU et al.: Neuropeptide Y acts directly

 in the periphery on fat tissue and mediates stress-induced

30. Chotiwat C, Harris RB: Antagonism of specific corticotropin- obesity and metabolic syndrome. Nat Med 2007, 13:803-811.

releasing factor receptor subtypes selectively modifies weight This paper demonstrates that the combination of chronic stress and high

loss in restrained rats. Am J Physiol Regul Integr Comp Physiol fat diet induces metabolic-like syndrome in mice through the effects of

2008, 295:R1762-R1773. NPY and NPY2R in white adipose tissue.

31. Krahn DD, Gosnell BA, Grace M, Levine AS: CRF antagonist 50. Rabasa C, Gagliano H, Pastor-Ciurana J, Fuentes S, Belda X,

partially reverses CRF- and stress-induced effects on feeding. Nadal R, Armario A: Adaptation of the hypothalamus–pituitary–

Brain Res Bull 1986, 17:285-289. adrenal axis to daily repeated stress does not follow the rules

Current Opinion in Behavioral Sciences 2016, 9:71–77 www.sciencedirect.com

Stress and metabolism Rabasa and Dickson 77

of habituation: a new perspective. Neurosci Biobehav Rev 2015, 65. Fleur SEl, Akana SF, Manalo SL, Dallman MF: Interaction

56:35-49. between corticosterone and insulin in obesity: regulation of

lard intake and fat stores. Endocrinology 2004, 145:2174-2185.

51. Oliver G, Wardle J, Gibson EL: Stress and food choice: a

laboratory study. Psychosom Med 2000, 62:853-865. 66. Nicod N, Giusti V, Besse C, Tappy L: Metabolic adaptations to

dexamethasone-induced insulin resistance in healthy

52. Pecoraro N, Reyes F, Gomez F, Bhargava A, Dallman MF: Chronic

volunteers. Obesity Res 2003, 11:625-631.

stress promotes palatable feeding, which reduces signs of

stress: feedforward and feedback effects of chronic stress. 67. Dallman MF, Warne JP, Foster MT, Pecoraro NC:

Endocrinology 2004, 145:3754-3762. Glucocorticoids and insulin both modulate caloric intake

through actions on the brain. J Physiol 2007, 583:431-436.

53. Piazza PV, Le Moal M: The role of stress in drug self-

administration. Trends Pharmacol Sci 1998, 19:67-74.

68. la Fleur SE, Houshyar H, Roy M, Dallman MF: Choice of lard, but

not total lard calories, damps adrenocorticotropin responses

54. Dallman MF, Pecoraro N, Akana SF et al.: Chronic stress and

to restraint. Endocrinology 2005, 146:2193-2199.

obesity: a new view of ‘‘comfort food’’. Proc Natl Acad Sci U S A

2003, 100:11696-11701.

69. Warne JP: Shaping the stress response: interplay of palatable

food choices, glucocorticoids, insulin and abdominal obesity.

55. Gibson EL: The psychobiology of comfort eating: implications

Mol Cell Endocrinol 2009, 300:137-146.

for neuropharmacological interventions. Behav Pharmacol

2012, 23:442-460.

70. Yoneshiro T, Aita S, Matsushita M, Kayahara T, Kameya T,

Kawai Y, Iwanaga T, Saito M: Recruited brown adipose tissue

56. de Kloet AD, Krause EG, Solomon MB et al.: Adipocyte

as an antiobesity agent in humans. J Clin Invest 2013, 123:3404-

 glucocorticoid receptors mediate fat-to-brain signaling.

3408.

Psychoneuroendocrinology 2015, 56:110-119.

Using mice with reduced expression of GR receptors in the WAT, the

71. Soumano K, Desbiens S, Rabelo R, Bakopanos E, Camirand A,

authors demonstrate that these receptors in the adipose tissue are

Silva JE: Glucocorticoids inhibit the transcriptional response

involved in the stress response, in the negative feedback exerted by

of the uncoupling protein-1 gene to adrenergic stimulation in a

the GCs and in the body weight increase in DIO mice.

brown adipose cell line. Mol Cell Endocrinol 2000, 165:7-15.

57. Ulrich-Lai YM, Christiansen AM, Wang X, Song S, Herman JP:

72. Strack AM, Bradbury MJ, Dallman MF: Corticosterone

 Statistical modeling implicates neuroanatomical circuit

decreases nonshivering thermogenesis and increases lipid

mediating stress relief by ‘comfort’ food. Brain Struct Funct

2015. storage in brown adipose tissue. Am J Physiol 1995, 268:R183-

R191.

This paper aims to explain the brain pathways involved in the comfort

food hypothesis using a statistical model. The results obtained suggest

73. Hirsch D, Zukowska Z: NPY and stress 30 years later: the

that sucrose (comfort food) limits stress responses via plastic changes to

peripheral view. Cell Mol Neurobiol 2012, 32:645-659.

the structure and function of stress-regulatory neural circuits including

basolateral–medial amygdala and BNST.

74. Baldock PA, Lin S, Zhang L et al.: Neuropeptide y attenuates

stress-induced bone loss through suppression of

58. National Cholesterol Education Program Expert Panel on

noradrenaline circuits. J Bone Miner Res 2014, 29:2238-2249.

Detection E, and Treatment of High Blood Cholesterol in

Adults (Adult Treatment Panel III). Third report of the national

75. van den Heuvel JK, Furman K, Gumbs MC et al.: Neuropeptide Y

cholesterol education program (NCEP) expert panel on

activity in the nucleus accumbens modulates feeding behavior

detection, evaluation, and treatment of high blood cholesterol

and neuronal activity. Biol Psychiatry 2015, 77:633-641.

in adults (adult treatment panel III) final report. Circulation 2002,

106:3143-3421.

76. Kas MJH, Bruijnzeel AW, Haanstra JR, Wiegant VM, Adan RAH:

Differential regulation of agouti-related protein and

59. Peckett AJ, Wright DC, Riddell MC: The effects of

neuropeptide Y in hypothalamic neurons following a stressful

glucocorticoids on adipose tissue lipid metabolism.

event. J Mol Endocrinol 2005, 35:159-164.

Metabolism 2011, 60:1500-1510.

77. Chagra SL, Zavala JK, Hall MV, Gosselink KL: Acute and

60. Baxter JD: Glucocorticoid hormone action. Pharmacol Therap

repeated restraint differentially activate orexigenic pathways

B: Gen Syst Pharmacol 1976, 2:605-659.

in the rat hypothalamus. Regul Pept 2011, 167:70-78.

61. Lee M-J, Pramyothin P, Karastergiou K, Fried SK: Deconstructing

78. Calvez J, Fromentin G, Nadkarni N, Darcel N, Even P, Tome D,

 the roles of glucocorticoids in adipose tissue biology and the

Ballet N, Chaumontet C: Inhibition of food intake induced by

development of central obesity. Biochim Biophys Acta (BBA)

acute stress in rats is due to satiation effects. Physiol Behav

Mol Basis Disease 2014, 1842:473-481.

2011, 104:675-683.

The authors review the systemic and local effects of GC in the adipose

tissue, presenting an updated version of possible molecular mechanisms

79. Turtzo LC, Marx R, Lane MD: Cross-talk between sympathetic

involved in the regulation of the adipose tissue by the GCs.

neurons and adipocytes in coculture. Proc Natl Acad Sci U S A

62. Castonguay TW: Glucocorticoids as modulators in the control 2001, 98:12385-12390.

of feeding. Brain Res Bull 1991, 27:423-428.

80. Lin EJ, Sun M, Choi EY, Magee D, Stets CW, During MJ: Social

63. Prague JK, May S, Whitelaw BC: Cushing’s syndrome. BMJ overcrowding as a chronic stress model that increases

2013, 346:f945. adiposity in mice. Psychoneuroendocrinology 2015, 51:318-330.

64. Zakrzewska KE, Cusin I, Stricker-Krongrad A, Boss O, Ricquier D, 81. Bartolomucci A, Cabassi A, Govoni P et al.: Metabolic

Jeanrenaud B, Rohner-Jeanrenaud F: Induction of obesity and consequences and vulnerability to diet-induced obesity in

hyperleptinemia by central glucocorticoid infusion in the rat. male mice under chronic social stress. PLoS One 2009,

Diabetes 1999, 48:365-370. 4:e4331.

www.sciencedirect.com Current Opinion in Behavioral Sciences 2016, 9:71–77