Review
Neural innervation of white adipose tissue and the control of lipolysis

https://doi.org/10.1016/j.yfrne.2014.04.001Get rights and content

Highlights

  • Principal stimulator of lipolysis in mammals is the sympathetic nervous system (SNS).

  • White adipose tissue (WAT) has sensory innervation of spinal origin.

  • Alterations in SNS drive affects adipocyte proliferation in addition to lipolysis.

  • WAT does not appear to possess parasympathetic innervation.

  • Insulin and adenosine inhibit SNS-induced lipolysis.

Abstract

White adipose tissue (WAT) is innervated by the sympathetic nervous system (SNS) and its activation is necessary for lipolysis. WAT parasympathetic innervation is not supported. Fully-executed SNS–norepinephrine (NE)-mediated WAT lipolysis is dependent on β-adrenoceptor stimulation ultimately hinging on hormone sensitive lipase and perilipin A phosphorylation. WAT sympathetic drive is appropriately measured electrophysiologically and neurochemically (NE turnover) in non-human animals and this drive is fat pad-specific preventing generalizations among WAT depots and non-WAT organs. Leptin-triggered SNS-mediated lipolysis is weakly supported, whereas insulin or adenosine inhibition of SNS/NE-mediated lipolysis is strongly supported. In addition to lipolysis control, increases or decreases in WAT SNS drive/NE inhibit and stimulate white adipocyte proliferation, respectively. WAT sensory nerves are of spinal-origin and sensitive to local leptin and increases in sympathetic drive, the latter implicating lipolysis. Transsynaptic viral tract tracers revealed WAT central sympathetic and sensory circuits including SNS-sensory feedback loops that may control lipolysis.

Introduction

Despite abundant data to the contrary, many textbooks, reviews and research papers continue to assert that adrenal medullary catecholamines, especially epinephrine (EPI), is the primary stimulator of lipolysis by white adipose tissue (WAT). This dogma belies the conclusive data showing that the removal of the sole source of circulating EPI by bilateral adrenal demedullation (ADMEDx) does not block lipolysis (e.g., Nishizawa and Bray, 1978, Takahashi and Shimazu, 1981). By contrast, the sympathetic nervous system (SNS) innervation of WAT is sufficient and necessary for the initiation of WAT lipolysis and performs a commanding function in the adjustment of lipid energy stores. Thus, this review will focus on the support for the direct SNS innervation of WAT as the principal initiator of lipolysis in mammals, including, of course, humans, as well as the role of the SNS in another WAT functions – fat cell proliferation. The typical counterpart to the SNS is the parasympathetic nervous system (PSNS) and despite some suggestion to the contrary (e.g., Kreier et al., 2002), the PSNS innervation of WAT and a function to oppose lipolysis is unfounded or trivial at best (Berthoud et al., 2007, Giordano et al., 2006, Giordano et al., 2007) as will be briefly discussed below. We also will describe the sensory innervation of WAT and its potential functions in the control of lipid metabolism. Finally, we will not describe in detail the SNS innervation of brown adipose tissue (BAT) and its role in energy balance, except its role to act in concert, or separately, from the SNS innervation of WAT in response to several challenges to energy balance because we have recently reviewed the literature on this topic (for review see: Bartness et al., 2010). Because we also have reviewed the SNS innervation of WAT recently and across the years (e.g., Bartness et al., 1993, Bartness et al., 2010, Bartness et al., 2005, Bartness and Song, 2007a, Bartness and Song, 2007b), we will make every attempt not to repeat this information, although some reiteration is necessary. Instead, we will add to this growing body of knowledge embellishing previous topics with new findings, suggest new areas of research to deepen our understanding of its role in lipolysis, and to speculate about the various mysteries of this area that remain to be solved.

Before delving into the heart of the review, we feel it would be helpful to indicate the location of the WAT depots we will be discussing primarily in rodent model research, as well as to compare and contrast them with WAT depots in humans. Tchkonia et al. (2013) nicely demonstrate the location of WAT depots in rodents and humans (Fig. 1). Although there are some similarities in WAT depots [e.g., mesenteric WAT (MWAT), perirenal WAT, retroperitoneal WAT (RWAT) – the latter not clearly shown; Fig. 1], there also are striking differences including the absence of omental WAT in rodents, the presence of perigonadal WAT in rodents [epididymal WAT (EWAT) or parametrial WAT (PWAT)] but their absence in humans, and differences in the extent of the subcutaneous WAT with restriction of these depots around the haunches in rodents [inguinal WAT (IWAT) and dorsosubcutaneous WAT (DWAT)], but underlying most of the skin in humans, and finally the absence of leg WAT in rodents. We feel these differences are not trivial distinctions and that conclusions based on rodent models, whether involving functional differences between visceral and subcutaneous WAT as well as individual WAT depots, should be made with more caution that historically or currently is the practice.

Our interest in the study of the SNS innervation of WAT was driven by our initial focus on the naturally-occurring decrease in the body fat of Siberian hamsters (Phodopus sungorus) triggered by changes in daylength. During the long days (LDs) of summer, their impressive obesity (e.g., ∼50% of body mass as body fat Bartness et al., 1989, Wade and Bartness, 1984) is at its peak and is reversed to a more moderate level of adiposity (∼20% of body mass as body fat) in the short days (SDs) of winter in the field naturally (Weiner, 1987). Fortunately, these naturally-occurring changes in lipid mass can be mimicked in the laboratory by changing only the photoperiod from LDs to SDs, while holding all other environmental factors constant such as temperature and food availability (Wade and Bartness, 1984; for review see: Bartness et al., 2002, Bartness and Wade, 1985). This is because for Siberian hamsters, and many other species exhibiting seasonal changes in adiposity and reproductive status, the daylength (photoperiod) cue is translated into a neuroendocrine signal via the duration of the nocturnal secretion of melatonin (MEL) from the pineal gland that occurs in direct proportion to the length of the dark period (for review see: Bartness and Goldman, 1989, Bartness et al., 1993, Goldman, 2001) thereby stimulating the MEL1a receptor (a.k.a. MT1-R) subtype that mediates photoperiodic responses (e.g., Roca et al., 1996). Because MEL does not affect lipolysis in vivo even at ‘industrial strength’ doses (Ng and Wong, 1986), an intermediary must exist. Even though there was nearly 100 years of suggestive, indirect evidence for the SNS innervation of WAT and its role in lipid mobilization (lipolysis), we fell into the trap of most researchers of the 1980s studying lipolysis and focused on circulating factors – in the case of Siberian hamsters, those that changed with changes in the daylength that also had been implicated in altering lipolysis [e.g., epinephrine (EPI), glucocorticoids, prolactin, thyroid hormones, gonadal steroids, insulin; for review see: Bartness et al., 2002]. None of these factors could account for the photoperiod-induced reversal of obesity by Siberian hamsters; therefore, there appeared to be a non-circulating factor initiating WAT lipolysis – perhaps a neural one. In addition, another factor favoring a ‘neural hypothesis’ was that in our initial and follow-up studies of the photoperiodic reversal of seasonal obesity, the intra-abdominal WAT pads (EWAT, RWAT) had the greatest degree of lipid mobilization, with the IWAT pad showing a lesser and later degree of lipid mobilization (Bartness, 1995, Bartness, 1996, Bartness et al., 1989, Wade and Bartness, 1984), a feat that could be accomplished by a circulating factor if its receptor number/affinity/signaling cascade varied accordingly among the WAT depots, or more simply by differential SNS drive to pads via its innervation and the release of norepinephrine (NE), the principal sympathetic nerve neurotransmitter. Indeed, in vitro lipolysis increases in isolated white adipocytes incubated with physiological concentrations of NE (e.g., Fain and Garcija-Sainz, 1983, Pecquery et al., 1983).

As noted in brief above, historically, but also unfortunately presently, adrenal medullary EPI often is ascribed as the primary initiator of WAT lipolysis. Perhaps this is due to the profound lipolysis engendered by application of physiological concentrations of the monoamine to WAT fragments ex vivo or isolated adipocytes in vitro (e.g., Bukowiecki et al., 1980, Leboeuf et al., 1959, Prigge and Grande, 1971, Rochon and Bukowiecki, 1990, Rosak and Hittelman, 1977, White and Engel, 1958, Wool et al., 1954). The role of adrenal medullary EPI for in vivo lipolysis has been discredited, however, because adrenal demedullation (ADMEDx), which removes the sole source of circulating EPI, does not block fasting-, exercise-, electrical stimulation of the hypothalamus- or glucoprivation-induced lipid mobilization in laboratory rats and mice or lipid mobilization in SD-exposed Siberian hamsters (Demas and Bartness, 2001, Kumon et al., 1976, Nishizawa and Bray, 1978, Takahashi and Shimazu, 1981, Teixeira et al., 1973).

Glucagon has long been implicated in mediating WAT lipolysis (Gjedsted et al., 2007, Lefebvre et al., 1973, Lefebvre, 1975, Lefebvre and Luyckx, 1969, Luyckx et al., 1975, Vaughan and Steinberg, 1963), but its effects on lipolysis are independent of CNS action because WAT SNS denervation does not block glucagon-induced glycerol release (an index of lipolysis), although it does decrease free fatty acid (FFA) release (Lefebvre et al., 1973). The latter effect is not due to a blockade of the effects of glucagon on lipolysis, rather it is attributed to re-esterification (Lefebvre et al., 1973). Indeed, exogenously administered i.v. glucagon that results in physiological concentrations of the hormone does not stimulate lipolysis in vivo as assessed by intra-WAT microdialysis (Gravholt et al., 2001).

Section snippets

Neuroanatomical evidence for the SNS innervation of WAT

When we finally turned to the SNS as a possible mediator of lipolysis, we realized direct evidence (tract tracing) of the sympathetic innervation of WAT had not been demonstrated. Previous studies used histological approaches at the level of the WAT pad. To our knowledge the first was by Dogiel 115 years ago (Dogiel, 1898) where he reported visible nerves of unknown type/origin entering WAT depots. The sympathetic innervation of WAT was then demonstrated at the level of the fat pad using

PRV, a transneuronal viral tract tracer, can be used to define central SNS outflow circuitry to WAT

As noted in brief above, the Bartha’s K strain of PRV is a viral transneuronal tract tracer that provides the ability to define multisynaptic circuits within the same animal (for review see: Ekstrand et al., 2008, Enquist, 2002, Song et al., 2005a). Some neurotropic viruses, such as PRV, are endocytosed at axon terminal membranes after binding to viral attachment protein molecules that act as ‘viral receptors’. They then are transported retrogradely (Curanovic and Enquist, 2009) specifically

AR subtype, number and affinity involved with WAT lipid mobilization

The control of lipolysis by the SNS relies on many factors at the cellular level, not the least of which are the number, affinity and type of ARs on the adipocyte membranes (for review see: Collins et al., 2004, Langin, 2006). The classic studies of Lafontan, Langin and associates (Carpene et al., 1993, Lafontan et al., 1979, Langin et al., 1991, Mauriege et al., 1988) discovered, clarified and highlighted the role of white adipocyte receptors in catecholamine and thus principally SNS-induced

A series of lipases work together to fully hydrolyze triacylglycerol (TAG) in white adipose tissue

Lipid is stored primarily as TAG and primarily in white adipocytes. Some distinction should be made between basal and stimulated lipolysis in terms of the relative importance of the series of enzymatic steps involved in complete TAG hydrolysis into three fatty acids (FAs) and glycerol. In brief, regarding basal lipolysis (i.e., non-SNS/NE-stimulated lipolysis), adipose triglyceride lipase (ATGL; a.k.a. desnutrin) has a preferential affinity for hydrolysis of TAG compared with diacylglycerol

Preclinical and clinical measures of SNS activity

A pervading assumption that likely has done more harm than good in our understanding of the SNS control of WAT lipolysis is that the SNS activity can be generalized from measures of one of its effected tissues such as measuring suspected indices of the SNS drive to the heart, skeletal muscle, or BAT and generalizing this to WAT. Perhaps this type of thinking stems from Walter Cannon’s ‘fight-or-flight’ response (Cannon, 1915) where the SNS is turned on uniformly with stress/fear (e.g., a bear

Lipolysis is blocked by SNS denervation and increased by SNS electrical stimulation

The first functional indication that activation of WAT SNS innervation is the primary initiator of lipolysis was reported more than one hundred years ago by Mansfeld (Mansfeld and Muller, 1913) whose hemiplegic patient with cancer cachexia only mobilized lipid from their neurally intact leg. Laboratory findings functionally implicating the SNS innervation of WAT mediating lipid mobilization date back to 1926 (Wertheimer, 1926, for review see: Bartness and Bamshad, 1998), where dogs with damage

Insulin is a major SNS inhibitory factor for lipolysis

NE-induced lipolysis is countered by pancreatic insulin, a powerful and prevalent inhibitor of lipolysis (e.g., Froesch et al., 1965, Goodridge and Ball, 1965, Prigge and Grande, 1971). This insulin-induced lipolysis inhibition can promote adiposity in humans because overeating triggers large increases in circulating insulin concentrations postprandially – a situation that currently prevails during waking hours given the availability of inexpensive and calorically dense foods (Wang et al., 2008

Leptin and sympathetically-and non-sympathetically-mediated lipolysis

There a few exceptions to the conclusion that the SNS is the principal initiator of lipolysis in WAT. Leptin was initially reported to increase lipolysis in vitro by adipocytes (Fruhbeck et al., 1997) and to increase rat SNS activity to WAT when administered intravenously at very high doses yielding a relatively small lipolytic response compared with equimolar NE injections (Shen et al., 2007). The relatively small in vivo lipolytic response to both peripheral and central leptin administration

Sympathetic drive to white adipose tissue is not uniform

As noted above, it is critical to realize that the SNS drive to peripheral tissues is not uniform. Moreover, across WAT depots we find that the pattern of SNS drive, as measured by NETO, is seemingly unique for each lipolytic stimulus tested to date prompting us to term this a ‘neurochemical fingerprint’ due to this individuality and the neurochemical nature of this measure of SNS drive. As examples, cold exposure increases NETO to IWAT, EWAT, RWAT and especially IBAT, but not DWAT (Brito et

Role of SNS in fat cell proliferation

Despite being the hallmark of obesity, white adipocyte proliferation is studied relatively infrequently compared with the vast literature on white adipocyte differentiation (for review see: Hausman et al., 2001). In 1992, Roy Martin’s laboratory (Jones et al., 1992) showed that in the presence of NE, cultured rat stromovascular fraction does not exhibit the normal increases in fat cell proliferation. Moreover, application of the β-AR antagonist propranolol before NE application disinhibited the

Neuroanatomical evidence for the sensory innervation of WAT

The sensory nerve-associated peptides in neural fibers innervating WAT were the earliest suggestions of WAT sensory innervation seen as seen by immunohistochemical labeling of the sensory nerve-associated peptide substance P (Fredholm, 1985) and later another sensory-nerve associated peptide CGRP (Hill et al., 1996) in laboratory rat WAT (Giordano et al., 1996) and CGRP in Siberian hamster IWAT and EWAT (Foster and Bartness, 2006, Shi and Bartness, 2005, Shi et al., 2005). Direct evidence

Conclusions and perspectives

This review summarizes the current knowledge of the roles of the sympathetic and sensory innervation of WAT. We have attempted to be comprehensive and at the same time not reiterative of our previous reviews. For what we consider to be an interesting historical perspective on the sympathetic innervation of WAT going back more than 100 years, see our initial review on the topic from 1998 (Bartness and Bamshad, 1998).

Clearly, investigations of the SNS innervation of WAT occupy the vast majority of

Acknowledgements

The authors thank Drs. James Granneman, Dominique Langin, Ruth Harris, Susan Fried, Patricj Card, Michael Jensen and John Kral for their discussions and insights.

References (322)

  • T.J. Bartness et al.

    Photoperiod-peptide interactions in the energy intake of Siberian hamsters

    Peptides

    (1986)
  • T.J. Bartness et al.

    Sensory and sympathetic nervous system control of white adipose tissue lipolysis

    Mol. Cell. Endocrinol.

    (2010)
  • T.W. Burns et al.

    Site of free-fatty-acid inhibition of lipolysis by human adipocytes

    Metabolism

    (1975)
  • G.Y. Carmen et al.

    Signalling mechanisms regulating lipolysis

    Cell. Signal.

    (2006)
  • P. Chakrabarti et al.

    FoxO1 controls insulin-dependent adipose triglyceride lipase (ATGL) expression and lipolysis in adipocytes

    J. Biol. Chem.

    (2009)
  • J. Ciriello et al.

    Systemic administration of leptin potentiates the response of neurons in the nucleus of the solitary tract to chemoreceptor activation in the rat

    Neuroscience

    (2013)
  • A.J. de Bold et al.

    A rapid and potent natriuretic response to intravenous injection of atrial myocardial extract in rats

    Life Sci.

    (1981)
  • V.P. Dole

    Effect of nucleic acid metabolites on lipolysis in adipose tissue

    J. Biol. Chem.

    (1961)
  • N.J. Dun et al.

    Orexins: a role in medullary sympathetic outflow

    Regul. Pept.

    (2000)
  • S.L. Edwards et al.

    Distinct preganglioinic neurons innervate noradrenaline and adrenaline cells in the cat adrenal medulla

    Neurosci.

    (1996)
  • M.I. Ekstrand et al.

    The alpha-herpesviruses: molecular pathfinders in nervous system circuits

    Trends Mol. Med.

    (2008)
  • J.N. Fain et al.

    Adrenergic regulation of adipocyte metabolism

    J. Lipid Res.

    (1983)
  • J.N. Fain et al.

    Effects of adenosine nucleosides on adenylate cyclase, phosphodiesterase, cyclic adenosine monophosphate accumulation, and lipolysis in fat cells

    J. Biol. Chem.

    (1972)
  • B.B. Fredholm

    Nervous control of circulation and metabolism in white adipose tissue

  • B.B. Fredholm et al.

    Adenosine and the regulation of metabolism and body temperature

    Adv. Pharmacol.

    (2011)
  • E.S. Adler et al.

    Neurochemical characterization and sexual dimorphism of projections from the brain to abdominal and subcutaneous white adipose tissue in the rat

    J. Neurosci.

    (2012)
  • S. Akselrod et al.

    Power spectrum analysis of heart rate fluctuation: a quantitative probe of beat-to-beat cardiovascular control

    Science

    (1981)
  • M.E. Altuncu et al.

    The use of short-term analysis of heart rate variability to assess autonomic function in obese children and its relationship with metabolic syndrome

    Cardiol. J.

    (2012)
  • B. Ballantyne

    Histochemical and biochemical aspects of cholinesterase activity of adipose tissue

    Arch. Int. Pharmacodyn. Ther.

    (1968)
  • M. Bamshad et al.

    Central nervous system origins of the sympathetic nervous system outflow to white adipose tissue

    Am. J. Physiol.

    (1998)
  • B.W. Banfield et al.

    Development of pseudorabies virus strains expressing red fluorescent proteins: new tools for multisynaptic labeling applications

    J. Virol.

    (2003)
  • T.J. Bartness et al.

    Innervation of mammalian white adipose tissue: Implications for the regulation of total body fat

    Am. J. Physiol.

    (1998)
  • T.J. Bartness et al.

    Mammalian pineal melatonin: a clock for all seasons

    Experientia

    (1989)
  • T.J. Bartness et al.

    Regional differences in fat pad responses to short days in Siberian hamsters

    Am. J. Physiol.

    (1989)
  • T.J. Bartness et al.

    SCN lesions block responses to systemic melatonin infusions in Siberian hamsters

    Am. J. Physiol.

    (1991)
  • T.J. Bartness et al.

    The timed infusion paradigm for melatonin delivery: what has it taught us about the melatonin signal, its reception, and the photoperiodic control of seasonal responses?

    J. Pineal Res.

    (1993)
  • T.J. Bartness et al.

    Seasonal changes in adiposity: the roles of the photoperiod, melatonin and other hormones and the sympathetic nervous system

    Exp. Biol. Med.

    (2002)
  • T.J. Bartness et al.

    Brain-adipose tissue cross talk

    Proc. Nutr. Soc.

    (2005)
  • T.J. Bartness et al.

    Sympathetic and sensory innervation of brown adipose tissue

    Int. J. Obes. (Lond.)

    (2010)
  • H.R. Berthoud et al.

    Vagaries of adipose tissue innervation

    Am. J. Physiol. Regul. Integr.Comp. Physiol.

    (2006)
  • H.R. Berthoud et al.

    Controversial white adipose tissue innervation by the vagus nerve: seeing is believing

    Am. J. Physiol.

    (2007)
  • A.B.L. Beznak et al.

    The effect of sympathectomy on the fatty deposit in connective tissue

    Quart. J. Exp. Physiol.

    (1937)
  • M. Bordicchia et al.

    Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes

    J. Clin. Invest.

    (2012)
  • R.R. Bowers et al.

    Sympathetic innervation of white adipose tissue and its regulation of fat cell number

    Am. J. Physiol.

    (2004)
  • R.R. Bowers et al.

    Short photoperiod exposure increases adipocyte sensitivity to noradrenergic stimulation in Siberian hamsters

    Am. J. Physiol.

    (2005)
  • G.A. Bray et al.

    Ventromedial hypothalamus modulates fat mobilisation during fasting

    Nature

    (1978)
  • M.N. Brito et al.

    Differential activation of the sympathetic innervation of adipose tissues by melanocortin receptor stimulation

    Endocrinology

    (2007)
  • N.A. Brito et al.

    Differential sympathetic drive to adipose tissues after food deprivation, cold exposure or glucoprivation

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2008)
  • B.B. Brodie et al.

    Application of steady state kinetics to the estimation of synthesis rate and turnover time of tissue catecholamines

    J. Pharmacol. Exp. Ther.

    (1966)
  • C. Buettner et al.

    Leptin controls adipose tissue lipogenesis via central, STAT3-independent mechanisms

    Nat. Med.

    (2008)
  • Cited by (0)

    This research was supported, in part, by National Institutes of Health Research Grant R37 DK 35254 to TJB.

    View full text