Hostname: page-component-7c8c6479df-24hb2 Total loading time: 0 Render date: 2024-03-29T15:39:14.158Z Has data issue: false hasContentIssue false

Adipose tissue development during early life: novel insights into energy balance from small and large mammals

Published online by Cambridge University Press:  18 June 2012

Michael E. Symonds*
Affiliation:
The Early Life Nutrition Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital, Nottingham NG7 2UH, UK
Mark Pope
Affiliation:
The Early Life Nutrition Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital, Nottingham NG7 2UH, UK
Helen Budge
Affiliation:
The Early Life Nutrition Research Unit, Academic Child Health, School of Clinical Sciences, University Hospital, Nottingham NG7 2UH, UK
*
*Corresponding author: Professor Michael E. Symonds, fax +44 115 823 0626, email michael.symonds@nottingham.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Since the rediscovery of brown adipose tissue (BAT) in adult human subjects in 2007, there has been a dramatic resurgence in research interest in its role in heat production and energy balance. This has coincided with a reassessment of the origins of BAT and the suggestion that brown preadipocytes could share a common lineage with skeletal myoblasts. In precocial newborns, such as sheep, the onset of non-shivering thermogenesis through activation of the BAT-specific uncoupling protein 1 (UCP1) is essential for effective adaptation to the cold exposure of the extra-uterine environment. This is mediated by a combination of endocrine adaptations which accompany normal parturition at birth and further endocrine stimulation from the mother's milk. Three distinct adipose depots have been identified in all species studied to date. These contain either primarily white, primarily brown or a mix of brown and white adipocytes. The latter tissue type is present, at least, in the fetus and, thereafter, appears to take on the characteristics of white adipose tissue during postnatal development. It is becoming apparent that a range of organ-specific mechanisms can promote UCP1 expression. They include the liver, heart and skeletal muscle, and involve unique endocrine systems that are stimulated by cold exposure and/or exercise. These multiple pathways that promote BAT function vary with age and between species that may determine the potential to be manipulated in early life. Such interventions could modify, or reverse, the normal ontogenic pathway by which BAT disappears after birth, thereby facilitating BAT thermogenesis through the life cycle.

Type
Symposium on ‘Metabolic flexibility in animal and human nutrition’
Copyright
Copyright © The Authors 2012

Abbreviations:
BAT

brown adipose tissue

PRLR

prolactin receptor

UCP1

uncoupling protein

Following the ‘rediscovery’ of brown adipose tissue (BAT) in 2007( Reference Nedergaard, Bengtsson and Cannon 1 ) there has been a substantial amount of innovative research into the assessment of both its function and regulation( Reference Nedergaard, Bengtsson and Cannon 2 ). This work has largely been undertaken in human adults with little consideration of its developmental aspects( Reference Symonds, Pope and Sharkey 3 ). The majority of animal studies have utilised rodent models as these have the advantages of offering gene deletion approaches, have a short duration and can be constrained within a closely controlled and confined environment( Reference Symonds, Sebert and Budge 4 ). However, as will be considered in more detail later, there are fundamental differences in early life development between small (i.e. rodents) and large (i.e. sheep) mammals, which could have a pronounced impact on our understanding of BAT function throughout the life cycle.

In adult human subjects, BAT is primarily sited in the supraclavicular regions where it is present across a wide range of ages( Reference Cypess, Lehman and Williams 5 Reference Virtanen, Lidell and Orava 7 ). This is not unexpected given that this is one of the main sites of BAT originally identified( Reference Aherne and Hull 8 ) and more recently confirmed( Reference Hu, Tovar and Pavlova 9 ) in infants. The initiation of non-shivering thermogenesis in both human subjects and large mammals, such as sheep, is one critical factor in determining metabolic adaptation to the cold challenge of the extra-uterine environment( Reference Clarke, Heasman and Firth 10 ).

The substantial potential of BAT to contribute to energy balance is not limited to the period immediately after birth because its maximal rate of heat production through the life cycle is in the order of 300 W/kg compared with 1 W/kg in all other tissues( Reference Power 11 , Reference Symonds, Budge and Perkins 12 ). In the 1980s it was calculated that in adult human subjects, only 40–50 g of BAT would be required for it to be able to contribute 20% of daily energy expenditure( Reference Rothwell and Stock 13 , Reference Rothwell and Stock 14 ), an estimation that has now been confirmed( Reference Cannon and Nedergaard 15 ). Surprisingly, despite the important role of the early life nutritional environment on adipose tissue development, especially in large mammals such as sheep, there has been only a limited amount of research into its impact on BAT( Reference Symonds, Pope and Sharkey 3 ). However, in rodents, at least, there have now been three specific sub-types of adipose tissue identified:

  1. (1) ‘Classical’ BAT located within the interscapular region that possesses large amounts of the BAT-specific uncoupling protein 1 (UCP1)( Reference Cannon and Nedergaard 16 ) and arises from a mfy5, muscle-like cell lineage( Reference Seale, Bjork and Yang 17 ).

  2. (2) White adipose tissue located in a large number of different locations throughout the body.

  3. (3) Brown adipose in white tissue depots that are not derived from a myf5-positive lineage( Reference Petrovic, Walden and Shabalina 18 ).

The extent to which comparable classifications can be made for adipose tissue development in large mammals remains to be established but, as this review will go on to highlight, it is likely that a different relationship could be present. Extensive studies are now required to explore the complexity of the multiple pathways involved in BAT regulation, and the differential effects of short- and long-term challenges. These will need to be undertaken across a range of different species in order to realise transferable interventions designed to promote BAT function and, thus, prevent excess body fat accumulation. At present, however, the majority of rodent studies have investigated the effects of a ‘high-fat’ dietary challenge( Reference Symonds, Sebert and Budge 4 ) conducted in a thermoneutral environment resulting in variable outcomes within BAT( Reference Fromme and Klingenspor 19 ).

Metabolic adaptation to the extra-uterine environment

BAT is maximally recruited at birth when the rapid appearance of UCP1 initiates non-shivering thermogenesis( Reference Symonds, Bird and Clarke 20 ). It coincides with maximal heat production which is rarely matched again at any other stage of life. This process is highly dependent on the degree of maturation at birth and, in altricial species such as the rat, this maturation is delayed until around 7 d after birth when the hypothalamic–pituitary axis becomes fully functional( Reference Cannon, Connoley, Obregon, Kunzel and Jesen 21 ). If this maturation process is inhibited, hypothermia and death rapidly result, although in some genetic manipulations of imprinted genes, there are still a small number of survivors( Reference Charalambous, Ferron and da Rocha 22 ). Interestingly, surviving offspring that were generated to lack the imprinted delta-like homologue 1/preadipocyte factor, and iodothyronine deiodinase type 3, fail to adapt effectively to the change in diet at weaning and were subsequently unable to maintain UCP1 longer than 11 d after birth. The extent to which a comparable developmental window is apparent in precocial mammals remains to be demonstrated.

Differences in BAT development are mediated, in part, by changes in thyroid function that has been established in both small( Reference Hall, Ribich and Christoffolete 23 ) and large( Reference Schermer, Bird and Lomax 24 ) mammals to be critical for the onset of BAT thermogenesis. Thus, the latter is influenced by the ability of triiodothyronine to serve as a bipotential mediator of mitochondrial biogenesis( Reference Lee, Takahashi and Yasubuchi 25 ). In addition to the effects of triiodothyronine, the rapid increase in UCP1 around the time of birth in large mammals (as summarised in Fig. 1) is also dependent on the rapid appearance of a range of other endocrine stimulatory factors including cortisol, prolactin, leptin and catecholamines( Reference Symonds, Mostyn and Pearce 26 ). Their secretion in the fetus/newborn is dependent on the stress of the normal birth process which results in intense activation of the central nervous system( Reference Symonds, Bird and Clarke 20 ). The magnitude of response is, however, dependent on exposure of the offspring to the cool temperature challenge of the extra-uterine environment( Reference Clarke, Heasman and Firth 10 ). To date, the effect of this stimulus has only been studied in the perirenal–abdominal depot of the sheep. This depot, which represents about 80% of all adipose tissue in the newborn sheep, is characterised by the rapid loss of its UCP1 expression( Reference Clarke, Buss and Juniper 27 ) and its subsequent apparent morphological conversion to white adipose tissue, coincident with a greatly increased leptin synthetic capacity (Fig. 1).

Fig. 1. Summary of the changes in gene expression for markers of brown (i.e. uncoupling protein 1 (UCP1))( Reference Mostyn, Wilson and Dandrea 28 ), white (i.e. leptin)( Reference Bispham, Budge and Mostyn 29 ) and brown adipocytes in white (i.e. homeobox C9 (HOXC9)( Reference Walden, Hansen and Timmons 30 )) in perirenal–abdominal adipose tissue between fetal and postnatal life (term=145 d gestation (gd)) in the sheep.

In rodents, it has been suggested that the conversion of myf5 myogenic progenitors to brown adipocytes is regulated by bone morphogenetic protein 7 acting through PRDM16 (PRD1-BF1-RIZ1 homologous domain containing 16)( Reference Seale, Bjork and Yang 17 , Reference Tseng, Kokkotou and Schulz 31 ). The optimum marker for brown adipose in white adipocytes, however, appears to be homeobox C9( Reference Walden, Hansen and Timmons 30 ) and exhibits a similar ontogeny to leptin (Fig. 1). The extent to which brown adipose in white characteristics can be manipulated in early life remains to be established; although, in the adult mouse gene expression for homeobox C9 is unaffected by cold exposure( Reference Walden, Hansen and Timmons 30 ).

Milk composition and brown adipose tissue thermogenesis

Feeding also appears to be vital in switching on BAT function in rodents( Reference Dessolin, Schalling and Champigny 32 ) in which fat composition of milk peaks at birth and then declines up to 1 week after birth( Reference Nicholas and Hartmann 33 ). Then even after this stage of lactation, the fat content remains around three times higher than that of either sheep( Reference Fulkerson and McDowell 34 ) or human subjects( Reference Kulski and Hartmann 35 ). Protein concentrations are also elevated in rodents but, while these remain unchanged through lactation in the rat( Reference Nicholas and Hartmann 33 ) they fall dramatically over the first 4 d of lactation in human breast milk( Reference Kulski and Hartmann 35 ). This primarily reflects the very different composition of colostrum in human (and sheep) milk( Reference Perez, Sanchez and Aranda 36 ) compared with that of rodents and which is necessary for providing immune protection immediately after birth. Human milk is also characterised as having a high cortisol content( Reference Kulski and Hartmann 37 ) although the extent to which this could further promote UCP1 synthesis in the newborn, as demonstrated in the fetus( Reference Mostyn, Pearce and Budge 38 ), has yet to be investigated. Therefore, milk composition between small and large mammals (including human subjects) is very different (as summarised in Table 1). The impact of alterations in milk composition resulting from pre or postnatal changes in maternal diet within species has been largely overlooked.

Table 1. Comparison of the milk composition between the rat, sheep and human subjects

Human milk also has a high prolactin( Reference Cregan, Mitoulas and Hartmann 42 ) content which is implicated in promoting BAT development and ensuring BAT thermogenesis is maintained through postnatal development( Reference Symonds, Sebert and Budge 43 ). This could result in enhanced BAT activity during later life, preventing excess adipose tissue deposition around central organs and increasing BAT sensitivity to thermogenic challenges. At the same time, diurnal variation in prolactin uptake within the human breast( Reference Cregan, Mitoulas and Hartmann 42 ) may further regulate BAT function in the infant. Prolactin is known to have a rapid thermogenic effect in neonatal sheep( Reference Pearce, Budge and Mostyn 44 ), at least when administered intravenously, but whether this response is present when prolactin is ingested enterally or is modulated between BAT depots has yet to be examined. It appears that not only is prolactin an important potential regulator of UCP1 in the perinatal period( Reference Virtanen, Lidell and Orava 7 ), newly identified depots of BAT in the sternal and clavicular regions of juvenile sheep( Reference Symonds, Pope and Sharkey 3 ) may be highly sensitive to prolactin as gene expression for UCP1 across these depots is highly positively correlated with that for the long form of the prolactin receptor (PRLR) (r 2 0·95; P<0·01).

There is also increasing evidence of differences in feeding behaviour( Reference Li, Fein and Grummer-Strawn 45 ), preferences( Reference Townsend and Pitchford 46 ) and growth trajectory( Reference Crume, Ogden and Mayer-Davis 47 ) between breast and bottle-fed infants. It is, therefore, possible that the absence of BAT activating endocrine factors within formula milk differentially resets energy regulation between breast- and formula-fed infants.

One factor that has restricted our ability to assess BAT thermogenesis in larger cohorts of human subjects has been the limitations of methods available to assess BAT activity: positron emission tomography with F-18 fluorodeoxyglucose; single photon emission tomography scanning with tracers such as I-123-meta-iodobenzylguanidine or Tc-99m-tetrofosmin; and tissue biopsy( Reference Fukuchi, Ono and Nakahata 48 Reference Thackeray, Beanlands and Dasilva 50 ). These invasive techniques have substantial disadvantages relating to the administration of comparatively large amounts of radiopharmaceuticals to healthy individuals, the use of single substrates to identify uptake into BAT, together with the static information they reveal and their relatively high costs. We have now developed the use of thermal imaging as a rapid, safe, and acceptable technique for use in human subjects, confirming previous findings in both rodents( Reference Blessing, Mohammed and Ootsuka 51 ) and human subjects( Reference Ouellet, Labbe and Blondin 52 ) of the rapidity with which BAT thermogenesis is initiated and showing that there is a pronounced reduction in activity of BAT with age( Reference Symonds, Henderson and Elvidge 53 ).

Using thermal imaging( Reference Symonds, Henderson and Elvidge 53 ), we are now investigating the potential thermogenic effects of individual food ingredients. This has demonstrated a significant thermogenic effect of drinking milk in young children (Fig. 2), which results in up to 0·7°C rise in BAT temperature, thus indicating a role in dietary-induced thermogenesis. It is likely that the macronutrient composition determines the magnitude of response( Reference Scazzina, Del Rio and Benini 54 ) that has been shown in adults to be promoted by protein and reduced by fat( Reference Westerterp, Wilson and Rolland 55 ).

Fig. 2. (Colour online) Comparison of the change in temperature of brown adipose tissue located within the supraclavicular region of an 8-year-old child in the fed state before (A) and after (B) placing one hand in cold water for 5 min (A, B) or before (C) and after (D) drinking 100 ml warm (about 70°C) semi-skimmed milk (adapted from Symonds et al.( Reference Symonds, Henderson and Elvidge 53 )). Each challenge was accompanied with a mean increase in temperature of the supraclavicular region of about 0·7°C.

The prolactin receptor and brown adipose tissue development

PRLR are one potential mediator by which the maternal nutritional environment can alter adipose tissue composition. PRLR are highly expressed in fetal adipose tissue( Reference Symonds, Phillips and Anthony 56 ) where they are more responsive to external challenges than in the fetal liver( Reference Bispham, Heasman and Clarke 57 ), for example. The increased amount of fetal BAT deposited when maternal food intake is raised is accompanied by an increased PRLR abundance( Reference Budge, Bispham and Dandrea 58 ), whereas sub-optimal maternal food intake has the opposite effect( Reference Symonds, Phillips and Anthony 56 ). In adults, it is known that the plasma concentration of prolactin is inversely related to the abundance of its receptor( Reference Auchtung, Rius and Kendall 59 ) and could be important in explaining seasonal differences in BAT function in adult human subjects( Reference Au-Yong, Thorn and Ganatra 60 ). To date, studies on the direct effects of prolactin on fetal adipose tissue have not been undertaken. This results, in part, from the potential confounding influence of placental lactogen on adipose tissue development as, potentially, it can also bind to the PRLR. The relative binding of placental lactogen, as compared to prolactin, to the PRLR varies markedly between tissues and a 100–1000 times greater molar concentration of placental lactogen is required to achieve the same binding as prolactin in both the liver and ovary( Reference Cohick, Dai and Xu 61 ). In contrast, in fibroblasts, placental lactogen binding to the PRLR is 250-fold greater than that of prolactin( Reference Fowlkes and Freemark 62 ). To date, comparable studies have not been undertaken in adipocytes; although it is notable that placental lactogen only has a very limited lipolytic effect( Reference Fielder and Talamantes 63 , Reference Campbell, Kostyo and Scanes 64 ) suggesting a low affinity of placental lactogen for the PRLR in adipose tissue. Moreover, in the newborn sheep, direct stimulation of the PRLR promotes BAT thermogenesis( Reference Pearce, Budge and Mostyn 44 ). These results have been confirmed in a mouse PRLR knockout model which demonstrated that BAT growth and development were severely impaired, as was the expression of a number of brown adipogenic genes including UCP1( Reference Viengchareun, Servel and Feve 65 ).

Given the important role prolactin has in establishing and maintaining pregnancy, it is perhaps surprising that its exact role in fetal development is still uncertain( Reference Soares 66 ). In sheep, although maternal plasma prolactin is decreased 100-fold by short day length and offspring circulating prolactin and progesterone concentrations also decrease, there are few obvious consequences for pregnancy, birth weight or postnatal growth( Reference Helliwell, Wallace and Aitken 67 ).

PRLR are also expressed in white adipose tissue and have been implicated in the regulation of adult fat mass. Two recent genome-wide association studies have identified a variant, rs4712652, adjacent to the prolactin gene that is associated with excess body weight( Reference Meyre, Delplanque and Chevre 68 , Reference Nilsson, Olsson and Isomaa 69 ). Furthermore, as may be expected for a gene that is closely involved in reproductive function, this relationship is gender specific with the risk A allele of rs4712652 strongly correlated with BMI and fat mass in males but not females( Reference Nilsson, Olsson and Isomaa 69 ). Previous clinical studies have demonstrated that correction of hyperprolactinaemia in human subjects improves body weight; although it has a much greater effect on reducing body weight in males compared to females. However, obese females with a normal prolactin profile, have a short-term body weight reduction, associated with a lowering in prolactin following administration of the dopamine D2 receptor agonist bromocriptine( Reference Kok, Roelfsema and Frolich 70 ). Clearly, further studies are needed to explore this relationship further especially from a developmental perspective.

External factors stimulating brown adipose tissue thermogenesis

There is now increasing evidence in rodents that a number of other tissues and endocrine factors can promote BAT function in rodents. One important example is the liver as, in rats, the postnatal maturation of BAT has been shown to relate to the onset of feeding and initiation of hepatic function, mediated by the release of fibroblast growth factor 21, which can also promote BAT function( Reference Hondares, Rosell and Gonzalez 71 ). This factor has also been shown to have a physiological role in adipose tissue of adult mice in which gene expression within adipose tissue is raised by chronic cold exposure, whereas it is depressed in the liver( Reference Fisher, Kleiner and Douris 72 ). Fibroblast growth factor 21 also appears to promote the ‘browning’ of some white adipose tissue depots where it acts in an autocrine/paracrine manner to enhance the abundance of the nuclear receptor coactivator PPARα 1α. It may, however, have only a modest role in energy balance as deletion of the fibroblast growth factor 21 gene does not impair BAT gene expression and the normal diurnal variation in body temperature is maintained with cold exposure. This is accompanied by a small (but non-significant) reduction in temperature which may be linked to increased recruitment of shivering thermogenesis. However, as fibroblast growth factor 21 can have widespread metabolic effects and results in skeletal fragility( Reference Wei, Dutchak and Wang 73 ), it is unlikely to have immediate therapeutic potential.

An important link between muscle and BAT development has been highlighted from studies focused on the heart( Reference Whittle and Vidal-Puig 74 ) and skeletal muscle( Reference Bostrom, Wu and Jedrychowski 75 ). The heart, acting through cardiac natriuretic peptides, can regulate BAT thermogenesis in adult mice( Reference Bordicchia, Liu and Amri 76 ). This response appears to be mediated by ventricular, or cardiac B-type, natriuretic peptide, which promotes UCP1 expression; although this effect is potentially greater in inguinal white fat, compared to interscapular BAT. Given the relatively high expression of UCP1 in both human( Reference Sacks, Fain and Holman 77 ), ovine( Reference Symonds, Pope and Sharkey 3 ) and mouse( Reference Walden, Hansen and Timmons 30 ) epicardial fat, it will interesting to see whether this, and other depots, respond in a similar manner( Reference Rudolph 78 ).

Skeletal muscle also has a potential role in regulating BAT function. Exercise in adult mice and human subjects results in increased secretion of a new hormone, irisin, which promotes energy expenditure in mice( Reference Bostrom, Wu and Jedrychowski 75 ). Irisin is a membrane protein that is cleaved from FNDC5 (fibronectin type III domain containing 5) and promotes the expression of UCP1 in white adipose tissue, but not BAT. The magnitude of response differs across white fat depots in mice and is greatest within inguinal fat. This study also demonstrated some fascinating differences in the gene response of muscle to exercise between mice and human subjects with leucine-rich α-2-glycoprotein, a novel member of GTPase-activating proteins( Reference Vogt and Seiler 79 ), showing the greatest response in human subjects, but being unresponsive to exercise in mice( Reference Bostrom, Wu and Jedrychowski 75 ). Intriguingly, leucine-rich α-2-glycoprotein has previously been linked to cell growth and cancer( Reference Li, Zhang and Qiu 80 , Reference Ladd, Busald Buson and Johnson 81 ) and animal studies have shown cancer per se can promote BAT function( Reference Bing, Brown and King 82 ).

Birth could be considered to represent a period of rapid muscular activity and is associated with a dramatic rise in muscle oxygenation( Reference Gunn and Gluckman 83 ). In precocial mammals such as sheep, birth is also accompanied by an increase in voluntary muscular activity, and with the onset of shivering thermogenesis which is, in turn, dependent both on BAT function( Reference Alexander and Williams 84 ) and on the magnitude of thermal challenge( Reference Symonds, Andrews and Johnson 85 ). The process of adaptation at birth may, therefore, provide further insights into the cross-talk between different muscle and fat depots, together with their ability of other organs to promote BAT function in early life as summarised in Fig. 3.

Fig. 3. Summary of the primary factors that can activate non-shivering thermogenesis in brown adipose tissue of the neonatal sheep. HPA, hypothalamic–pituitary–adrenal; ↑, increase.

In conclusion, multiple pathways can be recruited to promote BAT function. These may also promote the ‘browning’ of white adipose tissue and, thus, reverse the normal ontogenic pathway by which BAT disappears from some depots after birth. An increased understanding of these interactions and especially the role of feeding in further activating BAT in the postnatal period could result in novel interventions aimed at enhancing BAT thermogenesis throughout the life cycle.

Acknowledgements

The authors declare no conflict of interest. M. E. S. developed and wrote the article and undertook the analysis summarised in Fig. 2. M. P. undertook the analysis summarised in Fig. 1 and assisted in editing the final article. H. B. contributed to the article's development and to its editing.

References

1. Nedergaard, J, Bengtsson, T & Cannon, B (2007) Unexpected evidence for active brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab 293, E444–E452.CrossRefGoogle ScholarPubMed
2. Nedergaard, J, Bengtsson, T & Cannon, B (2010) Three years with adult human brown adipose tissue. Ann N Y Acad Sci 1212, E20E36.CrossRefGoogle ScholarPubMed
3. Symonds, ME, Pope, M, Sharkey, D et al. . (2012) Adipose tissue and fetal programming. Diabetologia 55(6), 15971606.CrossRefGoogle ScholarPubMed
4. Symonds, ME, Sebert, S & Budge, H (2011) The obesity epidemic: from the environment to epigenetics – not simply a response to dietary manipulation in a thermoneutral environment. Front Epigenom 2, 24, doi: 10.3389/fgene.2011. 00024 Google ScholarPubMed
5. Cypess, AM, Lehman, S, Williams, G et al. . (2009) Identification and importance of brown adipose tissue in adult humans. N Engl J Med 360, 15091517.CrossRefGoogle ScholarPubMed
6. van Marken Lichtenbelt, WD, Vanhommerig, JW, Smulders, NM et al. . (2009) Cold-activated brown adipose tissue in healthy men. N Engl J Med 360, 15001508.CrossRefGoogle ScholarPubMed
7. Virtanen, KA, Lidell, ME, Orava, J et al. . (2009) Functional brown adipose tissue in healthy adults. N Engl J Med 360, 15181525.CrossRefGoogle ScholarPubMed
8. Aherne, W & Hull, D (1966) Brown adipose tissue and heat production in the newborn infant. J Pathol Bacteriol 91, 223234.CrossRefGoogle ScholarPubMed
9. Hu, HH, Tovar, JP, Pavlova, Z et al. (2011) Unequivocal identification of brown adipose tissue in a human infant. J Magn Reson Imag 35(4), 938942.CrossRefGoogle Scholar
10. Clarke, L, Heasman, L, Firth, K et al. . (1997) Influence of route of delivery and ambient temperature on thermoregulation in newborn lambs. Am J Physiol Regul Integr Comp Physiol 272, R1931–R1939.CrossRefGoogle ScholarPubMed
11. Power, G (1989) Biology of temperature: the mammalian fetus. J Dev Physiol 12, 295304.Google ScholarPubMed
12. Symonds, ME, Budge, H, Perkins, AC et al. . (2011) Adipose tissue development – impact of the early life environment. Prog Biophys Mol Biol 106, 300306.CrossRefGoogle ScholarPubMed
13. Rothwell, NJ & Stock, MJ (1979) A role for brown adipose tissue in diet-induced thermogenesis. Nature 281, 3135.CrossRefGoogle ScholarPubMed
14. Rothwell, NJ & Stock, MJ (1983) Luxuskonsumption, diet-induced thermogenesis and brown fat: the case in favour. Clin Sci (Lond) 64, 1923.CrossRefGoogle Scholar
15. Cannon, B & Nedergaard, J (2012) Yes, even human brown fat is on fire! J Clin Invest 122(2), 486489.CrossRefGoogle ScholarPubMed
16. Cannon, B & Nedergaard, J (2004) Brown adipose tissue: function and physiological significance. Physiol Rev 84, 277359.CrossRefGoogle ScholarPubMed
17. Seale, P, Bjork, B, Yang, W et al. . (2008) PRDM16 controls a brown fat/skeletal muscle switch. Nature 454, 961967.CrossRefGoogle Scholar
18. Petrovic, N, Walden, TB, Shabalina, IG et al. . (2010) Chronic peroxisome proliferator-activated receptor gamma activation of epididymally derived white adipocyte cultures reveals a population of thermogenically competent, UCP1-containing adipocytes molecularly distinct from classic brown adipocytes. J Biol Chem 285, 71537164.CrossRefGoogle ScholarPubMed
19. Fromme, T & Klingenspor, M (2011) Uncoupling protein 1 expression and high-fat diets. Am J Physiol Regul Integr Comp Physiol 300, R18.CrossRefGoogle ScholarPubMed
20. Symonds, ME, Bird, JA, Clarke, L et al. . (1995) Nutrition, temperature and homeostasis during perinatal development. Exp Phys 80, 907940.CrossRefGoogle ScholarPubMed
21. Cannon, B, Connoley, E, Obregon, M-J et al. . (1988) Perinatal activation of brown adipose tissue. In The Endocrine Control of the Fetus, pp. 306320 [Kunzel, W & Jesen, A, editors]. Berlin: Springer Verlag.CrossRefGoogle Scholar
22. Charalambous, M, Ferron, SR, da Rocha, ST et al. . (2012) Imprinted gene dosage is critical for the transition to independent life. Cell Metab 15, 209221.CrossRefGoogle ScholarPubMed
23. Hall, JA, Ribich, S, Christoffolete, MA et al. . (2010) Absence of thyroid hormone activation during development underlies a permanent defect in adaptive thermogenesis. Endocrinology 151, 45734582.CrossRefGoogle ScholarPubMed
24. Schermer, SJ, Bird, JA, Lomax, MA et al. . (1996) Effect of fetal thyroidectomy on brown adipose tissue and thermoregulation in newborn lambs. Reprod Fertil Dev 8, 9951002.CrossRefGoogle ScholarPubMed
25. Lee, JY, Takahashi, N, Yasubuchi, M et al. . (2012) Triiodothyronine induces UCP-1 expression and mitochondrial biogenesis in human adipocytes. Am J Physiol Cell Physiol 302, C463–C472.CrossRefGoogle ScholarPubMed
26. Symonds, ME, Mostyn, A, Pearce, S et al. . (2003) Endocrine and nutritional regulation of fetal adipose tissue development. J Endocrinol 179, 293299.CrossRefGoogle ScholarPubMed
27. Clarke, L, Buss, DS, Juniper, DS et al. . (1997) Adipose tissue development during early postnatal life in ewe-reared lambs. Exp Phys 82, 10151017.Google ScholarPubMed
28. Mostyn, A, Wilson, V, Dandrea, J et al. . (2003) Ontogeny and nutritional manipulation of mitochondrial protein abundance in adipose tissue and the lungs of postnatal sheep. Br J Nutr 90, 323328.CrossRefGoogle ScholarPubMed
29. Bispham, J, Budge, H, Mostyn, A et al. . (2002) Ambient temperature, maternal dexamethasone, and postnatal ontogeny of leptin in the neonatal lamb. Pediatr Res 52, 8590.CrossRefGoogle ScholarPubMed
30. Walden, TB, Hansen, IR, Timmons, JA et al. . (2012) Recruited vs. nonrecruited molecular signatures of brown, “brite”, and white adipose tissues. Am J Physiol Endocrinol Metab 302, E19–E31.CrossRefGoogle Scholar
31. Tseng, YH, Kokkotou, E, Schulz, TJ et al. . (2008) New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454, 10001004.CrossRefGoogle ScholarPubMed
32. Dessolin, S, Schalling, M, Champigny, O et al. . (1997) Leptin gene is expressed in rat brown adipose tissue at birth. FASEB J 11, 382387.CrossRefGoogle ScholarPubMed
33. Nicholas, KR & Hartmann, PE (1991) Milk secretion in the rat: progressive changes in milk composition during lactation and weaning and the effect of diet. Comp Biochem Physiol A Comp Physiol 98, 535542.CrossRefGoogle ScholarPubMed
34. Fulkerson, WJ & McDowell, GH (1974) Effect of oestrogen administered in early or late lactation on the yield and composition of milk in sheep. J Endocrinol 63, 175180.CrossRefGoogle ScholarPubMed
35. Kulski, JK & Hartmann, PE (1981) Changes in human milk composition during the initiation of lactation. Aust J Exp Biol Med Sci 59, 101114.CrossRefGoogle ScholarPubMed
36. Perez, MD, Sanchez, L, Aranda, P et al. . (1990) Synthesis and evolution of concentration of beta-lactoglobulin and alpha-lactalbumin from cow and sheep colostrum and milk throughout early lactation. Cell Mol Biol 36, 205212.Google ScholarPubMed
37. Kulski, JK & Hartmann, PE (1981) Changes in the concentration of cortisol in milk during different stages of human lactation. Aust J Exp Biol Med Sci 59, 769778.CrossRefGoogle ScholarPubMed
38. Mostyn, A, Pearce, S, Budge, H et al. . (2003) Influence of cortisol on adipose tissue development in the fetal sheep during late gestation. J Endocrinol 176, 2330.CrossRefGoogle ScholarPubMed
39. Banchero, GE, Quintans, G, Martin, GB et al. . (2004) Nutrition and colostrum production in sheep. 1. Metabolic and hormonal responses to a high-energy supplement in the final stages of pregnancy. Reprod Fertil Dev 16, 633643.CrossRefGoogle ScholarPubMed
40. Macy, IG (1949) Composition of human colostrum and milk. Am J Dis Child 78, 589603.Google ScholarPubMed
41. Michaelsen, KF, Skafte, L, Badsberg, JH et al. . (1990) Variation in macronutrients in human bank milk: influencing factors and implications for human milk banking. J Pediatr Gastroenterol Nutr 11, 229239.CrossRefGoogle ScholarPubMed
42. Cregan, MD, Mitoulas, LR & Hartmann, PE (2002) Milk prolactin, feed volume and duration between feeds in women breastfeeding their full-term infants over a 24 h period. Exp Physiol 87, 207214.CrossRefGoogle ScholarPubMed
43. Symonds, ME, Sebert, SP & Budge, H (2010) Nutritional regulation of fetal growth and implications for productive life in ruminants. Animal 4, 10571083.CrossRefGoogle ScholarPubMed
44. Pearce, S, Budge, H, Mostyn, A et al. . (2005) Prolactin, the prolactin receptor and uncoupling protein abundance and function in adipose tissue during development in young sheep. J Endocrinol 184, 351359.CrossRefGoogle ScholarPubMed
45. Li, R, Fein, SB & Grummer-Strawn, LM (2010) Do infants fed from bottles lack self-regulation of milk intake compared with directly breastfed infants? Pediatrics 125, e1386–e1393.CrossRefGoogle ScholarPubMed
46. Townsend, E & Pitchford, NJ (2012) Baby knows best? The impact of weaning style on food preferences and body mass index in early childhood in a case-controlled sample. BMJ Open 2, e000298.CrossRefGoogle Scholar
47. Crume, TL, Ogden, LG, Mayer-Davis, EJ et al. (2012) The impact of neonatal breast-feeding on growth trajectories of youth exposed and unexposed to diabetes in utero: the EPOCH Study. Int J Obes (Lond) 36(4), 529534.CrossRefGoogle ScholarPubMed
48. Fukuchi, K, Ono, Y, Nakahata, Y et al. . (2003) Visualization of interscapular brown adipose tissue using (99 m)Tc-tetrofosmin in pediatric patients. J Nucl Med 44, 15821585.Google Scholar
49. Hadi, M, Chen, CC, Whatley, M et al. . (2007) Brown fat imaging with (18)F-6-fluorodopamine PET/CT, (18)F-FDG PET/CT, and (123)I-MIBG SPECT: a study of patients being evaluated for pheochromocytoma. J Nucl Med 48, 10771083.CrossRefGoogle Scholar
50. Thackeray, JT, Beanlands, RS & Dasilva, JN (2007) Presence of specific 11C-meta-Hydroxyephedrine retention in heart, lung, pancreas, and brown adipose tissue. J Nucl Med 48, 17331740.CrossRefGoogle ScholarPubMed
51. Blessing, W, Mohammed, M & Ootsuka, Y (2012) Heating and eating: brown adipose tissue thermogenesis precedes food ingestion as part of the ultradian basic rest-activity cycle in rats. Physiol Behav 105, 966974.CrossRefGoogle ScholarPubMed
52. Ouellet, V, Labbe, SM, Blondin, DP et al. (2012) Brown adipose tissue oxidative metabolism contributes to energy expenditure during acute cold exposure in humans. J Clin Invest 122(2), 545552.CrossRefGoogle ScholarPubMed
53. Symonds, ME, Henderson, K, Elvidge, L et al. . (2012) Thermal imaging to assess age-related changes of skin temperature within the supraclavicular region co-locating with brown adipose tissue in healthy children. J Pediatr (In the Press).CrossRefGoogle ScholarPubMed
54. Scazzina, F, Del Rio, D, Benini, L et al. . (2010) The effect of breakfasts varying in glycemic index and glycemic load on dietary induced thermogenesis and respiratory quotient. Nutr Metab Cardiovasc Dis 21, 121125.CrossRefGoogle Scholar
55. Westerterp, KR, Wilson, SA & Rolland, V (1999) Diet induced thermogenesis measured over 24 h in a respiration chamber: effect of diet composition. Int J Obes Relat Metab Disord 23, 287292.CrossRefGoogle Scholar
56. Symonds, ME, Phillips, ID, Anthony, RV et al. . (1998) Prolactin receptor gene expression and foetal adipose tissue. J Neuroendocrinol 10, 885890.CrossRefGoogle ScholarPubMed
57. Bispham, J, Heasman, L, Clarke, L et al. . (1999) Effect of maternal dexamethasone treatment and ambient temperature on prolactin receptor abundance in Brown adipose and hepatic tissue in the foetus and new-born lamb. J Neuroendocrinol 11, 849856.CrossRefGoogle ScholarPubMed
58. Budge, H, Bispham, J, Dandrea, J et al. . (2000) Effect of maternal nutrition on brown adipose tissue and prolactin receptor status in the fetal lamb. Pediatr Res 47, 781786.CrossRefGoogle ScholarPubMed
59. Auchtung, TL, Rius, AG, Kendall, PE et al. . (2005) Effects of photoperiod during the dry period on prolactin, prolactin receptor, and milk production of dairy cows. J Dairy Sci 88, 121127.CrossRefGoogle ScholarPubMed
60. Au-Yong, IT, Thorn, N, Ganatra, R et al. . (2009) Brown adipose tissue and seasonal variation in humans. Diabetes 58, 25832587.CrossRefGoogle ScholarPubMed
61. Cohick, CB, Dai, G, Xu, L et al. . (1996) Placental lactogen-I variant utilizes the prolactin receptor signaling pathway. Mol Cell Endocrinol 116, 4958.CrossRefGoogle ScholarPubMed
62. Fowlkes, J & Freemark, M (1992) Binding of placental lactogen and growth hormone to fetal sheep fibroblasts. Pediatr Res 32, 200203.CrossRefGoogle ScholarPubMed
63. Fielder, PJ & Talamantes, F (1987) The lipolytic effects of mouse placental lactogen II, mouse prolactin, and mouse growth hormone on adipose tissue from virgin and pregnant mice. Endocrinology 121, 493497.CrossRefGoogle ScholarPubMed
64. Campbell, RM, Kostyo, JL & Scanes, CG (1990) Lipolytic and antilipolytic effects of human growth hormone, its 20-kilodalton variant, a reduced and carboxymethylated derivative, and human placental lactogen on chicken adipose tissue in vitro. Proc Soc Exp Biol Med 193, 269273.CrossRefGoogle ScholarPubMed
65. Viengchareun, S, Servel, N, Feve, B et al. . (2008) Prolactin receptor signaling is essential for perinatal brown adipocyte function: a role for insulin-like growth factor-2. PLoS ONE 3, e1535.CrossRefGoogle ScholarPubMed
66. Soares, MJ (2004) The prolactin and growth hormone families: pregnancy-specific hormones/cytokines at the maternal-fetal interface. Reprod Biol Endocrinol 2, 51.CrossRefGoogle ScholarPubMed
67. Helliwell, RJ, Wallace, JM, Aitken, RP et al. . (1997) The effect of prenatal photoperiodic history on the postnatal endocrine status of female lambs. Anim Reprod Sci 47, 303314.CrossRefGoogle ScholarPubMed
68. Meyre, D, Delplanque, J, Chevre, JC et al. . (2009) Genome-wide association study for early-onset and morbid adult obesity identifies three new risk loci in European populations. Nat Genet 41, 157159.CrossRefGoogle ScholarPubMed
69. Nilsson, L, Olsson, AH, Isomaa, B et al. . (2011) A common variant near the PRL gene is associated with increased adiposity in males. Mol Genet Metab 102, 7881.CrossRefGoogle ScholarPubMed
70. Kok, P, Roelfsema, F, Frolich, M et al. . (2006) Activation of dopamine D2 receptors simultaneously ameliorates various metabolic features of obese women. Am J Physiol Endocrinol Metab 291, E1038E1043.CrossRefGoogle ScholarPubMed
71. Hondares, E, Rosell, M, Gonzalez, FJ et al. . (2010) Hepatic FGF21 expression is induced at birth via PPARalpha in response to milk intake and contributes to thermogenic activation of neonatal brown fat. Cell Metab 11, 206212.CrossRefGoogle ScholarPubMed
72. Fisher, FM, Kleiner, S, Douris, N et al. . (2012) FGF21 regulates PGC-1alpha and browning of white adipose tissues in adaptive thermogenesis. Genes Dev 26, 271281.CrossRefGoogle ScholarPubMed
73. Wei, W, Dutchak, PA, Wang, X et al. (2012) Fibroblast growth factor 21 promotes bone loss by potentiating the effects of peroxisome proliferator-activated receptor γ. Proc Natl Acad Sci USA 109(8), 31433148.CrossRefGoogle ScholarPubMed
74. Whittle, AJ & Vidal-Puig, A (2012) NPs – heart hormones that regulate brown fat? J Clin Invest 122, 804807.CrossRefGoogle ScholarPubMed
75. Bostrom, P, Wu, J, Jedrychowski, MP et al. . (2012) A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463468.CrossRefGoogle ScholarPubMed
76. Bordicchia, M, Liu, D, Amri, EZ et al. . (2012) Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes. J Clin Invest 122, 10221036.CrossRefGoogle ScholarPubMed
77. Sacks, HS, Fain, JN, Holman, B et al. . (2009) Uncoupling protein-1 and related messenger ribonucleic acids in human epicardial and other adipose tissues: epicardial fat functioning as brown fat. J Clin Endocrinol Metab 94, 36113615.CrossRefGoogle ScholarPubMed
78. Rudolph, AM (1985) Distribution and regulation of blood flow in the fetal and neonatal lamb. Circ Res 57, 811821.CrossRefGoogle ScholarPubMed
79. Vogt, N & Seiler, S (2008) The RHO1-specific GTPase-activating protein LRG1 regulates polar tip growth in parallel to Ndr kinase signaling in Neurospora. Mol Biol Cell 19, 45544569.CrossRefGoogle ScholarPubMed
80. Li, Y, Zhang, Y, Qiu, F et al. . (2011) Proteomic identification of exosomal LRG1: a potential urinary biomarker for detecting NSCLC. Electrophoresis 32, 19761983.CrossRefGoogle ScholarPubMed
81. Ladd, J, Busald Buson, T, Johnson, M et al. (2012) Increased plasma levels of the APC-interacting protein MAPRE1, LRG1 and IGFBP2 preceding a diagnosis of colorectal cancer in women. Cancer Prev Res 5(4), 655664.CrossRefGoogle ScholarPubMed
82. Bing, C, Brown, M, King, P et al. . (2000) Increased gene expression of brown fat uncoupling protein (UCP)1 and skeletal muscle UCP2 and UCP3 in MAC16-induced cancer cachexia. Cancer Res 60, 24052410.Google ScholarPubMed
83. Gunn, T & Gluckman, PD (1995) Perinatal thermogenesis. Early Hum Dev 42, 169183.CrossRefGoogle ScholarPubMed
84. Alexander, G & Williams, D (1968) Shivering and nonshivering thermogenesis during summit metabolism in young lambs. J Physiol (Lond) 198, 251276.CrossRefGoogle ScholarPubMed
85. Symonds, ME, Andrews, DC & Johnson, PJ (1989) The control of thermoregulation in the developing lamb during slow wave sleep. J Dev Physiol 11, 289298.Google ScholarPubMed
Figure 0

Fig. 1. Summary of the changes in gene expression for markers of brown (i.e. uncoupling protein 1 (UCP1))(28), white (i.e. leptin)(29) and brown adipocytes in white (i.e. homeobox C9 (HOXC9)(30)) in perirenal–abdominal adipose tissue between fetal and postnatal life (term=145 d gestation (gd)) in the sheep.

Figure 1

Table 1. Comparison of the milk composition between the rat, sheep and human subjects

Figure 2

Fig. 2. (Colour online) Comparison of the change in temperature of brown adipose tissue located within the supraclavicular region of an 8-year-old child in the fed state before (A) and after (B) placing one hand in cold water for 5 min (A, B) or before (C) and after (D) drinking 100 ml warm (about 70°C) semi-skimmed milk (adapted from Symonds et al.(53)). Each challenge was accompanied with a mean increase in temperature of the supraclavicular region of about 0·7°C.

Figure 3

Fig. 3. Summary of the primary factors that can activate non-shivering thermogenesis in brown adipose tissue of the neonatal sheep. HPA, hypothalamic–pituitary–adrenal; ↑, increase.