Next Article in Journal
Impact on Isoleucine and Valine Supplementation When Decreasing Use of Medical Food in the Nutritional Management of Methylmalonic Acidemia
Previous Article in Journal
β-hydroxybutyrate Impedes the Progression of Alzheimer’s Disease and Atherosclerosis in ApoE-Deficient Mice
Previous Article in Special Issue
Leptin, Obesity, and Leptin Resistance: Where Are We 25 Years Later?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Incendiary Leptin

by
Patricia Seoane-Collazo
1,2,3,*,†,
Noelia Martínez-Sánchez
4,*,†,
Edward Milbank
2,3 and
Cristina Contreras
5,*
1
International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
2
CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain
3
CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
4
Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
5
Department of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
*
Authors to whom correspondence should be addressed.
These authors have contributed equally to this work.
Nutrients 2020, 12(2), 472; https://doi.org/10.3390/nu12020472
Submission received: 13 December 2019 / Revised: 6 February 2020 / Accepted: 8 February 2020 / Published: 13 February 2020
(This article belongs to the Special Issue Leptin)

Abstract

:
Leptin is a hormone released by adipose tissue that plays a key role in the control of energy homeostasis through its binding to leptin receptors (LepR), mainly expressed in the hypothalamus. Most scientific evidence points to leptin’s satiating effect being due to its dual capacity to promote the expression of anorexigenic neuropeptides and to reduce orexigenic expression in the hypothalamus. However, it has also been demonstrated that leptin can stimulate (i) thermogenesis in brown adipose tissue (BAT) and (ii) the browning of white adipose tissue (WAT). Since the demonstration of the importance of BAT in humans 10 years ago, its study has aroused great interest, mainly in the improvement of obesity-associated metabolic disorders through the induction of thermogenesis. Consequently, several strategies targeting BAT activation (mainly in rodent models) have demonstrated great potential to improve hyperlipidemias, hepatic steatosis, insulin resistance and weight gain, leading to an overall healthier metabolic profile. Here, we review the potential therapeutic ability of leptin to correct obesity and other metabolic disorders, not only through its satiating effect, but by also utilizing its thermogenic properties.

1. The Obesity Pandemic

The World Health Organization defines being obese and overweight as an abnormal or excessive fat accumulation that may impair health [1]. The risk increases when fat accumulation overtakes adipose tissue storage capacity, leading to an ectopic accumulation in other tissues or organs [2,3,4,5,6]—therefore, the regulation and maintenance of energy homeostasis are essential for organism survival. Since energy balance is influenced by a myriad of factors (including genetic, hormonal and nutritional), compounded by environmental and psychosocial variables, the regulation of energy homeostasis is complex. In general terms, the variation in body mass is the result of an alteration of the balance between energy intake and energy expenditure (EE), which are dynamically influencing each other [7] in such a way that individuals with a balanced body mass are protected against an excessive increase or decrease in their fat deposits by compensatory changes in EE [8,9,10].
Obesity is associated with a decrease in life expectancy of between 5 and 20 years and can engender many disabilities depending on the severity of the condition and associated comorbidities [11,12]. These include, among others, hypertension, dyslipidemia, hypertriglyceridemia, insulin resistance and inflammation; cumulatively resulting in a reduced quality of life and social disadvantages [12]. The simultaneous occurrence of several of these factors is known as “metabolic syndrome” and is an important risk factor in the development of diabetes, cardiovascular diseases and/or different types of cancer. The combination of hypercaloric overnutrition, sedentary lifestyle and the evolutionary maintenance of energy-conserving genes [13] has led to an increase of the incidence of obesity in a large portion of the population, independently of the country’s per capita income or development level [14]. Considering the substantial complications of obesity, healthcare spending represents a large budget burden in the developed countries. Due to this, a huge push has been made to identify the underlying molecular mechanisms controlling and regulating energy homeostasis.

2. Thermogenesis

Dietary interventions to reduce body weight usually fail due to compensatory changes in EE. In light of this, the role of thermogenesis (i.e., heat production not related to physical activity [15]) in the modulation of EE has been widely investigated and studied. Total EE is the sum of several components: basal metabolic rate (BMR) (approximately 80%), physical activity (10%) and thermogenesis (10%). Of particular note, in active individuals, the combination of both activity and thermogenesis accounts for nearly half (44%) of the total average EE [15,16]. Thermogenesis is a fundamental process used by homeothermic organisms to maintain their body temperature, with the main organ involved in thermogenic regulation being brown adipose tissue (BAT), a specialized adipose tissue mainly involved in heat production, a regulatory process known as “non shivering thermogenesis” (NST) [17]. BAT is especially important in small mammals, as it permits the generation of heat independently of shivering thermogenesis in which heat is produced by involuntary muscle contractions [17,18].
At a histological level adipocytes located in BAT differ from those found in white adipose tissue (WAT). BAT adipocytes possess a polygonal shape with multilocular lipid droplets surrounded by numerous mitochondria [19], containing iron-pigmented cytochromes largely responsible for the brown colour of BAT. Inside the mitochondria, the energy resulting from the movement of electrons through the respiratory chain is classically used by ATP synthase to generate ATP from ADP [20,21,22]; however, this energy-producing process can be modified by uncoupling protein 1 (UCP1). UCP1 provides an alternative way to return protons to the mitochondrial matrix, bypassing ATP synthase and producing heat via thermogenesis [17,23,24].
The adipocytes found in WAT contain few mitochondria and a single large droplet of lipids and are principally involved in fat storage; however, adipocytes displaying similar characteristics to brown adipocytes can be observed in WAT depots by a process called “beiging” or “browning” of WAT [25]. Apart from their morphological differences, brown and white adipocytes are also originating from different precursor cells [26,27,28,29,30]. While brown adipocytes differentiate from myogenic factor 5 (Myf5) expressing precursor cells (as muscle cells), white adipocytes originate from a distinct Myf5-/- cell lineage [18,31,32]. Interestingly, while possessing similar thermogenic capacities to brown adipocytes, beige adipocytes have the same cellular origin as white adipocytes, differentiating from Myf5-/- precursors [33,34,35]. It should be noted, however, that the in vivo thermogenic capacity of beige/brite (brown-in-white) cells is greatly reduced compared to brown adipocytes [36,37,38,39].

2.1. Activation of BAT Thermogenesis

Brown adipocytes are richly innervated by sympathetic nerve efferent fibres [40,41,42,43,44]. The sympathetic nervous system (SNS) is essential for the activation of thermogenesis that can be regulated both at central and peripheral levels [45,46,47,48,49,50]. The increase of sympathetic tone induces a release of norepinephrine at nerve terminals, that binds and activates β adrenergic receptors (classically β3 adrenergic receptor, β3-AR) localized to the membrane of brown adipocytes. β3-ARs are coupled to G proteins; thus, once stimulated, adenylate cyclase (AC) is activated, triggering an intracellular increase in cyclic adenosine monophosphate (cAMP), which in turn activates protein kinase A (PKA), inducing thermogenesis and subsequently the activation of p38-mitogen-activated protein kinase (MAPK) [17,35,51] (Figure 1).
Dependent on PKA activation mechanisms as well as mediating pathways, 2 models of BAT responses have been proposed: acute and chronic. During the acute effect, PKA increases lipolysis through the activation of (i) adipocyte triglyceride lipase (ATGL), (ii) hormone-sensitive lipase (HSL) and (iii) monoacylglycerol lipase (MGL), which hydrolyse the triacylglicerides (TAG) to release free non-esterified cytosolic fatty acids (NEFA). Subsequently, NEFAs-CoA are transported into the mitochondria by carnitine palmitoyltransferase 1a (CPT1a) before being oxidized, leading to the subsequent formation of NADH and FADH, which are in turn oxidized in the electron transport chain [35,52,53,54,55]. Recent studies have called into question whether this model could fully explain the mechanism supporting thermogenic activation. In the standard model, UCP1 is inactive being bound to ATP and ADP (or GDP), with this inhibition is reversed by the release of NEFAs from the lipid droplet through ATGL lipase activity [56]. Nevertheless, new evidence has shown that even in the absence of ATGL (or the subsequent ATGL-induced enzymes), thermogenesis remains inducible through BAT capturing NEFAs released by WAT [57,58], which are used as substrates and activators of UCP1. The overriding concern with this model is that the acute role of the SNS is lost/unaccounted for. Therefore, another model has been proposed, in which a norepinephrine-dependent “non-fatty acid” mechanism is required to activate UCP1, allowing its uncoupling and the use of external NEFAs for thermogenesis [56]. Aside from acute effects, the prolonged stimulation of BAT induces a chronic activation of PKA leading to an increase in UCP1 protein levels, mitochondrial biogenesis and BAT hyperplasia and hypertrophy [17,35,56,59] (Figure 2).

2.2. Thermogenesis in Humans

Until a few years ago, BAT was considered to be a relevant metabolic tissue only in rodents, hibernating mammals and newborn humans [17,43,59,60]; however, using positron emission tomography-computed tomography (PET-CT), functional BAT was also identified in human adults, and localized in dispersed, yet defined areas of variable sizes, namely: neck, supraclavicular area, perinephric area, intercostal areas and periaortic regions [41,60,61,62]. Its subsequent molecular characterization has shown however, that in the supraclavicular and/or cervical area, its expression profile is more similar to beige/brite adipocytes than to brown adipocytes [25,34,63]. In adults, BAT activity increases as the environmental temperature decreases [62]; accordingly, activated BAT was observed after a period working in a cold environment [64] or after a 2h cold exposure in controlled laboratory conditions [65]. Nonetheless, the magnitude of BAT response to cold exposure is highly variable [62,65] and depends on parameters such as age, sex, body mass index (BMI) or fat mass [65]. For instance, as has observed in aged conditions, the amount and distribution of BAT concomitantly decreases together with an increase in adiposity [66]. Hence a substantial amount of people do not exhibit any signs of BAT activation even after cold exposure [67]. Considering this, several studies have explored the possibility of BAT function recovery by repeated cold exposure showing an increase in glucose uptake by BAT (a hallmark of BAT activity) [68], as well as in BAT volume [69]. Additionally, experiments of sequential monthly acclimation to cold and warm temperatures showed reversible BAT recruitment after cold exposure [70,71].

2.3. Thermogenesis, a Therapeutic Treatment for Obesity?

Since the identification of functional BAT in adult humans, numerous studies have been performed to evaluate the therapeutic potential of BAT thermogenesis on metabolic features in humans. A caveat remains, that the relative contribution of BAT to EE is lower in humans than in rodents—while it has been estimated that in rodents thermogenesis could increase the daily EE up to 20% [35,62], maximally activated BAT in humans contributes to approximately 5% of BMR [72].
So far, the main results concerning the therapeutic role of BAT in humans are linked to a reduction in fat mass and to an improvement of glycemic and lipid metabolism. Nevertheless, the evidence gathered by cold exposure approaches have shown none [68] or, in combination with other interventions, a modest [73,74] reduction in body weight and fat mass [75]. Of note, in lean healthy individuals, glucose uptake after BAT activation is increased, improving insulin sensitivity and glucose clearance at a whole body level [76,77,78,79,80]. However, in obese patients, studies have failed to observe any increase in EE, even after recruitment of BAT, probably due to insufficient BAT activation [81]. Whereas in type 2 diabetic patients, cold acclimation was able to increase peripheral insulin sensitivity by augmenting BAT volume and activity [82], suggesting that BAT activation could be an interesting target for diabetes therapy. Interestingly, a possible role of BAT as a regulator of lipid metabolism in humans has also emerged, considering that after cold exposure activated BAT preferentially uses intracellular triglycerides as main substrate for oxidative metabolism instead of plasma glucose or NEFAs [76,83,84,85]. As the number of studies focused on cold exposure in humans has increased, new features of possible effects of BAT activation have emerged. A preliminary study (in which BAT activation was not assessed) performed on hypercholesterolemic patients submitted to prolonged cold exposure showed a reduced body mass associated to an improved total cholesterol and LDL cholesterol, independently of physical activity or changes in food intake [86]. Furthermore, it was described that individuals with cold-activated detectable BAT presented lower plasma cholesterol and LDL cholesterol compared to subjects without detectable one [65]. In any case, further studies are needed to assess if hypercholesterolemia could also be alleviated by BAT activation.
More recently, increasing efforts to target BAT in an obesity-driven context have been made, focusing mainly on the pharmacological activation of thermogenesis instead of temperature interventions [87]. These innovative BAT pharmacological interventions were developed mainly due to the fact that (i) BAT activity is inversely correlated with BMI [60,62] and (ii) because obese subjects are less prone to respond to cold-induced thermogenesis [88], probably due to greater adipose insulation. The initial pharmacological candidates were β3-AR agonists; however, almost all have been discarded following clinical trials due to the lack of effectiveness, toxicity issues and/or crossover effects with β1- and β2-AR [89]. Nevertheless, studies have found that mirabegron, a selective β3-AR agonist approved for the treatment of overactive bladder, was able to increase BAT activity and WAT lipolysis in healthy male subjects in the same range of efficiency as cold exposure [90]. In agreement with this, a recent clinical trial has found that both mirabegron and cold exposure (even if limited to a small surface skin area) induced an increase of beige adipose tissue markers in human subcutaneous WAT [91]. Of note, this effect was observed in obese insulin-resistant subjects pointing out the possible therapeutic use of mirabegron to treat obesity, although further studies are needed to determine whether these effects on WAT beiging are associated to a better clinical outcome.
Other factors inducing BAT activation have been explored in several studies such as (i) the sympathomimetic compound ephedrine that was able to increase EE (independently of BAT activation) [92], (ii) capsinoids (although providing contradictory results on EE modulation) [93,94,95,96], (iii) bile acids—inducing an increase of BAT activity [97] or (iv) leptin and leptin sensitizers which will be discussed later in this review.
In this therapeutic context, it is important to mention that mutations in the UCP1 gene could be involved in the pathology of obesity. In this sense, some nucleotide substitutions in the UCP1 gene were described in obese people suggesting that specific variations of UCP1 could promote energy storage and contribute to the development of obesity [98]. Other specific UCP1 polymorphisms, such as rs1800592 and rs3811791, are associated with obesity and abnormal values of high-density lipoprotein (HDL), low-density lipoprotein (LDL) and triglycerides levels [99]. It has also been shown that the UCP2-866 polymorphism was associated with high levels of leptin in an obese or overweight Mexican population [100]. Of note, these studies have failed to find any strong general association between other UCP1 polymorphisms and obesity, instead relying on specific parameters or obesity levels [99], highlighting the remarkable complexity of the topic.

3. Leptin History

Historically WAT has been considered as a passive energy storage organ; however insurmountable evidence has demonstrated that WAT possesses a major endocrine function through the release of several factors into the circulation. In 1987, the first description of adipose tissue being a source of sexual hormones was made [101]. The next discovered factor released by adipose tissue was adipsin, which was markedly decreased in obese rodent models [102]. In 1994, leptin was identified and characterized, cementing the endocrine function of adipose tissue [103]. Nowadays a plethora of hormones and peptides released by adipose tissue have been identified such as: interleukin (IL) 6, tumour necrosis factor-α (TNF α), monocyte chemoattractant protein (MCP) 1, plasminogen activator inhibitor (PAI)-1, resistin, adiponectin, to name a few. Collectively termed adipokines, they can act in autocrine, paracrine or endocrine manners, establishing a complex communication between adipose tissue and other organs, including the brain [104].
Before the identification of leptin, several studies had already suggested the existence of a particular circulating endocrine hormone capable of signalling information to the central nervous system (CNS) about the energy status/requirements [105]. This theory was later confirmed by Friedman and colleagues through the identification of the leptin gene (Lepob) [103] that encodes for a 16 kDa polypeptide composed of 167 amino acids, named Leptin from the Greek λεπτός (leptos), meaning “thin”. Subsequently, the leptin receptor (LepR: Lepdb in mice and Lepfa in rats) was described [106,107,108]. Leptin production is regulated by the Lepob gene in adipocytes, and therefore by lipid content and adipocyte size [109], meaning that circulating leptin levels directly correlate with adipose tissue mass (elevated numbers of subcutaneous depots compared to visceral ones) and nutritional status [110,111]. It should be noted however, that many other factors can regulate the expression and secretion of leptin.
The conservation of leptin across different species and diverse organs points to the functional relevance of this adipokine [112] and its subsequent involvement in the regulation of many biological systems including: energy homeostasis, endocrine systems, immune function, hematopoiesis, angiogenesis and bone development [104,113,114,115,116,117,118,119,120,121].
In order to understand metabolic pathologies such as obesity or type 2 diabetes, genetically modified rodent models bearing alterations in leptin signalling pathway have been developed throughout the years. Most famously, ob/ob mice completely lack functional leptin due to a single autosomal recessive mutation in Lepob [103,122], whilst db/db mice carry a single autosomal recessive mutation in the LepR gene resulting in abnormal and nonfunctional LepR. Of note, while db/db mice (lacking LepR) display high levels of leptin, they are resistant to its effect [106,107]. In addition to mice, rat models exhibiting alterations in leptin signalling have also been generated. For instance, Zucker fatty (fa) rats harbour a mutation in the LepR gene resulting in nonfunctional receptor [123]. Interestingly, all these aforementioned models develop genetic obesity and enter a prediabetic state, mimicking clinical progressions and are therefore widely used to study these pathologies and leptin signalling pathways.

4. Leptin Anorexigenic Effects

The effects of leptin on energy balance are mainly due its food intake suppressing properties and to its ability to induce thermogenesis. Regarding its primary function, leptin is considered to be the main ‘satiety hormone’ [124,125,126,127,128]. Accordingly, when injected in leptin-deficient patients, leptin is able to normalize hyperphagia through a reduction of food intake [129,130] and to decrease hunger without affecting satiety in adults [131].
The major neuronal targets of leptin are located in the hypothalamus, a brain area located under the thalamus mainly involved in the energy balance regulation, that is composed of distinct neuronal populations grouped in hypothalamic nuclei forming the: arcuate (ARC), ventromedial (VMH), paraventricular (PVH), dorsomedial (DMH), nucleus of the hypothalamus and lateral hypothalamic area (LHA) [106,119,132,133,134,135]. Interestingly, it has been demonstrated that the ARC performs a key role in mediating leptin actions [136], with two notable subpopulations of neurons expressing LepR:
(1)
Pro-opiomelanocortin (POMC) neurons that express anorexigenic neuropeptides such as POMC and cocaine- and amphetamine-regulated transcript (CART) [137,138], and are stimulated by leptin through the release of α-melanocite-stimulating hormone (α-MSH) [139,140,141] that binds the melanocortin 3 receptors (MC3R) and MC4R. Several genetic variants of POMC and MC4R genes have been associated to human obesity, suggesting that the central melanocortin system is required for the anorexigenic effect of leptin.
(2)
Agouti-related protein (AgRP) neurons [142,143] which express orexigenic neuropeptides such as neuropeptide Y (NPY) and AgRP [144]. Leptin exerts inhibitory effects on both AgRP and NPY neurons activity as well as on the release of the associated AgRP and NPY neuropeptides [142,145], resulting in a potent satiating effect.

5. Molecular Mechanisms Mediating Leptin Effect

The leptin signalling pathway is initiated when leptin binds its receptor located in central and peripheral organs. The LepR gene encodes for 6 LepR isoforms (LepRa to LepRf) through differential mRNA splicing processes, with a conserved N-terminal intracellular domain. LepRb is the only isoform that has a full-length intracellular domain and is involved in leptin signalling [106,146,147]. The hypothalamus is the main area in which leptin’s anti-obesity effects are mediated [148,149]. How other isoforms of LepR are involved in leptin signalling remains unclear; however, it is likely that they could be implicated in the transportation and clearance of leptin [145,150,151].
As LepRb does not contain any intrinsic enzymatic activity, its association to a cytoplasmic tyrosine kinase—Janus tyrosine kinase 2 (JAK2)—is needed to initiate the subsequent leptin-mediated molecular mechanisms. The binding of leptin to dimerized LepRb stimulates JAK2, inducing its activation by auto-phosphorylation allowing the phosphorylation of LepRb on three tyrosine residues—Tyr985, Tyr1077 and Tyr1138. When phosphorylated, these tyrosine residues act as binding sites for Src homology 2 (SH2) domain containing molecules triggering the corresponding downstream signalling pathways (Figure 3).
On one hand, in response to leptin, JAK2 phosphorylates LepR on Tyr1138 triggering the recruitment of the Signal Transducer and Activator of Transcript 3 (STAT3) through its SH2 domain. STAT3 is subsequently phosphorylated by JAK2, resulting in its dimerization and nuclear translocation in order to regulate the expression of STAT3-target genes, including Suppressor Of Cytokine Signaling 3 (SOCS3) [152,153,154,155]. STAT3 acts as a transcription factor essential for feeding regulation [149,156,157,158,159]. Moreover, leptin can activate STAT5 through the phosphorylation of LepRb on its Tyr1077 and Tyr 1138 (partial phosphorylation) residues. Interestingly, both STAT3 and STAT5 deletion have been associated with obesity and hyperphagia states.
On the other hand, phosphorylation of LepR on Tyr 985 provides a binding site for the SH2 domain protein tyrosine phosphatase 2 (SHP2) which regulates Extracellular Signal Regulate Kinase (ERK) pathway, that is known to be associated with both thermogenic and anorectic effects of leptin [160]. Phospho-Tyr985 can also be triggered by SOCS3 which exerts a negative feedback effect suppressing the activation of the LepRb/JAK2 pathways [161]. It is important to include here that a selective mutation in Tyr985 improved leptin signalling in lean mice, highlighting the role of Tyr985 in this negative feedback signalling [162,163].
Finally, the activation of JAK2 following leptin binding to LepRb promotes the phosphorylation of insulin receptor substrate 1 and 2 (IRS1 and 2) inducing the activation of the Phosphatidylinositol 3-Kinase (PI3K)/protein kinase B (Akt) pathway [164,165,166,167,168,169] and of its two corresponding downstream events: (1) phosphorylation of Forkhead box protein O1 (FOXO1) that is translocated from the cytosol to the nucleus, promoting the transcription of POMC and the inhibition of AgRP and NPY transcription, leading to the suppression of food intake [170,171,172]. In this sense, it has been shown that an overexpression of FoxO1 in the ARC abolishes leptin response increasing feeding and body weight, while its deletion results in an inverted phenotype [170,171,173]; (2) The other downstream event derived from IRS/PI3K/Akt pathway is the activation of the mammalian Target Of Rapamycin (mTOR)/ribosomal S6 Kinase (S6K) [142]. It has been demonstrated that the hypothalamic activation of mTOR complex1 (mTORC1) decreased food intake and body weight in rodent models, while rapamycin, its suppressor, had orexigenic effects [174], suggesting that mTOR plays a crucial role in the leptin-mediated regulation of energy homeostasis.
Besides the classical leptin-dependent signalling pathway, the Calcium Calmodulin-dependent protein Kinase Kinase (CaMKK2)/5′-AMP-activated protein kinase (AMPK)/acetyl-CoA caroboxylase (ACC) pathway has been demonstrated to be involved in leptin receptor signalling [142,175,176]. AMPK is typically activated under decreased intracellular energy levels (i.e., low ATP/ADP ratio) [177]. Thus, while leptin signalling is activated by increased levels of glucose, AMPK is activated when they are decreased. Interestingly, the pharmacological inhibition of AMPK [178] restores the leptin signalling pathway, suggesting that CaMKK2/AMPK/ACC signalling exerts a key role in the modulation of the leptin-dependent pathways.

6. Leptin Effects on Thermogenesis and Browning

In addition to its actions regulating food intake, leptin also positively regulates energy expenditure and thermogenesis. Deficiency in leptin or in LepR leads to a decrease in energy expenditure and in core body temperature [179,180]. Indeed, ob/ob mice present (i) a reduced SNS activity, (ii) lower expression of β3-AR [181,182] and (iii) decreased body temperature [126,183,184]. On the other hand, many agents (cold exposure, hormones, β3-AR agonists, dietary factors and exercise among others) have been described to be clear browning inducers [50,185,186,187] while the exact role of leptin in WAT browning still remains unclear. In the following section, the central and peripheral mechanisms of leptin involved in thermogenesis and browning processes will be reviewed.

6.1. Thermogenic Effects of Leptin

The role of leptin in BAT thermogenesis stimulation was demonstrated years ago. Of note, leptin exerts its thermogenic action mainly through the activation of the SNS. In rodent models, peripheral or central leptin injections increase sympathetic nerve activity innervating BAT (an effect that depends on the integrity of LepR) [188,189,190]. Moreover, in obesity models (diet-induced-obesity and ob/ob mice), intraperitoneal injection of leptin increases BAT UCP1 mRNA levels and activity without causing significant changes in mice locomotor activity [191,192]. As mentioned above, the hypothalamus has the highest expression of LepR and interestingly, many of the hypothalamic neurons involved in the regulation of the thermogenesis are also leptin sensitive (Figure 4).
Before diving into leptin’s actions and effects on different hypothalamic areas, it is important to mention that some authors have not observed any association between thermogenesis and leptin. Leptin-deficient ob/ob mice have been characterized as thermogenic limited and hypothermic due to atrophied BAT. However, other studies have demonstrated that the BAT of these mice were perfectly functional and that leptin treatment did not increase BAT thermogenesis. Recently, Fischer and colleagues observed that leptin administration in wild type and ob/ob mice did not induce any thermogenic response. Surprisingly, they observed that ob/ob mice displayed a decreased body temperature that was normalized to wild-type levels after leptin treatment [193]. As other authors have obtained similar results, highlighting the idea that leptin may not be considered as a thermogenic but as a pyrexic one [194,195,196,197,198,199], further studies are warranted to clarify leptin’s exact role in regulating thermogenesis.

6.2. Leptin Actions in the DMH

Some evidence suggests that many neuronal populations in the DMH are important for the regulation of BAT thermogenesis [200,201,202]. In regards to the main topic of this review, it is important to note that the neurons composing the DMH display a very high density of LepR [203] and that their activity is sufficient to promote BAT thermogenesis, increase energy expenditure and locomotor activity—cumulatively leading to a decrease in body weight [204]. Concordantly, the ablation of LepR in DMH neurons results in weight gain by decreasing EE and locomotor activity [204,205]. Specifically, some authors have shown that after intraperitoneal injection of leptin, DMH phospho-STAT3 expression increased, inducing sympathetic activation and increased BAT activity [192,206]. Accordingly, following intra-DMH leptin injection, BAT temperature also increased and that this effect could be blocked by prior infusion of β3-AR antagonist [192]. Additionally, some authors have shown that the disruption of LepR in the prolactin releasing peptide (PrRP) neurons (a specific DMH neuronal subset associated with thermogenesis), induced a decrease in the thermogenic effects of peripheral leptin [207]. DMH LepR-expressing neurons project their axons to different brain regions including the PVH [203,208] and the Raphe pallidus nucleus (RPa) [209,210], areas that are involved in the sympathetic regulation of crucial physiological parameters such as body temperature, blood pressure or heart rate [203,211]. Moreover, Zhang and co-workers demonstrated that LepR in the DMH mediates the thermoregulatory actions of leptin through the use of a retrograde trans-synaptic tracer [211]. Taken together, these findings support the idea that leptin, through its actions on leptin-sensitive neurons of the DMH, plays a role in the stimulation of the BAT thermogenesis and in the control of body temperature.

6.3. Leptin Actions in the Preoptic Area (POA)

The POA is the main region involved in temperature sensing and in the integration of peripheral and central thermal information emanating from the organism. The medial POA (mPOA) detects cold temperature signals and induces a thermogenic response in BAT through hypothalamic outputs [212,213]; in this way, the mPOA plays an important role in sympathetic/thermogenic BAT circuits. More precisely, mPOA neurons project their axons directly to neurons of the DMH to regulate sympathetic BAT inputs and the associated thermoregulatory responses [200,214,215]. As in the DMH, neurons expressing LepR have been found in the mPOA [143,211] and are involved in sympathetic circuits activating BAT and in thermoregulatory leptin actions [216]. Zhang and colleagues [216], using a retrograde transsynaptic tracer pseudorabies virus (PRV), revealed that LepR neurons of the mPOA contributed to the regulation of the sympathetic BAT outputs. These LepR BAT-related neurons project to the DMH/dorsal hypothalamic area (DHA) and to the RPa forming an interconnected circuit in which leptin could act at different levels [163]. Remarkably, mPOA neurons express MC4R and the specific mPOA injection of its agonist, melanotan II (MTII), was shown to increase thermogenesis [217]. As the mPOA modulates excitatory DMH neurons projecting to the RPa [218,219], this effect is ablated by lesioning the DMH [217].

6.4. Leptin Actions in the PVH

The PVH plays an important role in the regulation of energy homeostasis and is interconnected with other hypothalamic areas. However, the role of LepRb in the PVH remains controversial as most of the evidence is indirect and unclear [220]. However, some reports suggest that both oxytocin and also thyrotropin-releasing hormone (TRH) neurons located in the posterior pituitary gland and the PVH, respectively, express LepR and could modulate EE [221]. Therefore, it has been suggested that the PVH could act as a final common pathway in response to leptin. For example, DMH neuronal efferents project to parvicellular PVH areas [222,223,224] that directly innervate parasympathetic and sympathetic preganglionic neurons in the medulla and spinal cord [137]. Some results support the idea that a subset of leptin-sensitive cells in the DMH innervate the PVH [208,211], but the functional significance of these projections needs to be clarified with further studies.

6.5. Leptin Actions in the VMH

One of the main roles of the VMH is the regulation of thermogenesis through the integration of several peripheral signals to produce a thermogenic response in BAT and WAT. Therefore, numerous peripheral signals relaying the nutritional status (as nutrients and hormones, including leptin) are conveyed to the VMH, in which high concentrations of LepR have been described [132]. The role of leptin in the VMH on thermogenesis modulation is well accepted, indeed microinjections of leptin into the VMH have been shown to increase epinephrine and norepinephrine plasma concentrations [225]. Other studies supporting this VMH leptin-mediated sympathetic activation have shown that leptin infusion into the VMH increases glucose uptake in peripheral tissues [226], as well as blood pressure and renal sympathetic activity [227,228]. Specifically, after leptin microinjections into the VMH, changes in BAT activity were detected with increased BAT glucose uptake. Interestingly, this effect was blocked after specific sympathetic denervation of BAT [226]. The dorsomedial area of the VMH is especially enriched in LepRb and in the specific transcription factor Steroidogenic Factor-1 (SF1) [229,230]. Leptin directly activates SF1 neurons in the VMH inducing modulation of body weight. The use of mice models lacking LepR in SF1 neurons has demonstrated that these neurons mediate, at least in part, the anti-obesity effect of leptin. Specifically, when challenged with high fat diet (HFD), these SF1 LepR-/- mice develop an increase in body weight and fat storage without hyperphagia, which was explained by a defective adaptative thermogenic response accompanied by decreased BAT UCP1 expression [231,232]. Curiously, VMH specific SF1 KO mice display significantly reduced VMH LepR expression [232]. It is important to note that the SF1 neurons are mostly glutamatergic [233,234], exerting output functions involved in the regulation of sympathetic nervous system outflows [235]. Thus, the deletion of these neurons induces a decrease in sympathetic outflow and, as a result, a decrease in thermogenesis. Other studies have shown that LIM domain only 4 (LMO4), a transcription cofactor essential in CNS development, is also involved in central leptin signalling. This cofactor is expressed in specific nuclei of the hypothalamus, including the VMH [236]. Chronic intracerebroventricular (ICV) leptin infusion in mice with neuronal specific ablation of LM04 induced a decrease in thermogenesis and energy expenditure, while the leptin-induced weight and fat loss were less marked [237]. Thus, this study proposed LMO4 as a novel modulator of leptin function in selective hypothalamic areas. Moreover, recently, a link between Cannabinoid type-1 (CB1) receptors expressed in VMH neurons and the metabolic actions of leptin was established. Mice lacking CB1 receptors specifically in SF1 expressing neurons possessed an increase in sympathetic activity and a decrease in adiposity when fed with a standard diet. Conversely, under HFD conditions, these mice developed leptin resistance and increased peripheral adiposity [238], suggesting a diet-dependent role of the SF1-VMH CB1 receptors in energy balance and metabolic responses to leptin. The regulation of leptin activity in neurons also implicates the transcription factor FOXO1 that plays a central role in metabolic homeostasis, with the VMH being a key site for its action [173]. Since FOXO1 is considered to be a negative regulator of leptin (via PI3K/pAKT pathway), its specific ablation in SF1 neurons of the VMH, induces an increase of SNS activity and UCP1 expression, and consequently EE [239]. Recently, new studies were performed in rat models, demonstrating that weight loss in leptin-treated rats only occurs in combination with the simultaneous activation of LepR in the hindbrain and forebrain, with a critical role of the VMH in this interconnected network [240].

6.6. Leptin Actions in the ARC

As previously mentioned, the ARC is a hypothalamic nucleus with high levels of expression of the LepRb isoform. The ARC is closely related to the control of leptin-dependent feeding, but is also an important area for the leptin-induced increase in BAT sympathetic outflow. Several studies have demonstrated that the ARC was involved in leptin response, indeed the sympathetic activation of BAT observed after systemic administration of leptin was blunted after specific electrolytic lesions of the ARC [241]. On the other hand, direct injections of leptin into the ARC induced the increase of BAT sympathetic tone and blood pressure [242]. Moreover, the selective deletion of LepRb in the ARC reduced BAT sympathetic nerve responses to leptin [242,243].
More specifically, the two neuronal populations expressing either the orexigenic peptides AgRP/NPY or the anorexigenic peptide POMC, are the principal sites of LepR expression in the ARC and exert opposing effects on metabolism [244]. In addition to its role in the decrease in EE, ARC and NPY neurons are critical for the sympathetic control of BAT function [245]. The connection between NPY and leptin has been long recognized: on one hand, NPY is overexpressed in the hypothalamus of ob/ob mice [246], and its repetitive administration into the CNS was shown to suppress the SNS activity of BAT, decreasing energy expenditure [247,248] and inducing obesity [249]. On the other hand, ob/ob mice treated with leptin displayed lower ARC NPY expression [127,134] and the single or repetitive central administration of leptin decreases NPY mRNA levels in the ARC [250,251,252]. On the basis of these findings, NPY seems to be a strong mediator of leptin thermogenic actions. More precisely, leptin could act centrally to stimulate BAT activity and this effect may be partially mediated by the inhibition of NPY neurons of the ARC [253]. It is also important to mention that NPY neurons project to both the PVH and the VMH and that these neuronal connections are implicated in NPY-mediated thermoregulatory effects [248,254].
The second ARC main neuronal population—POMC expressing neurons—which are part of the melanocortin system, have also been linked to leptin action [141,255]. The activation of LepR in POMC neurons has hypertensive effects, increasing blood pressure and heart rate [256]. Although POMC expressing neurons are also found in the nucleus of the tract solitarius (NTS), leptin does not stimulate them [257], suggesting that leptin can differentially regulate these two POMC neurons populations. In addition, MCR3 and MCR4 are important mediators of leptin action in the neurons of the melanocortin system [258,259]. The co-administration of leptin and SHU9119 (a synthetic antagonist of both MCR3 and MCR4 receptors), attenuated leptin-induced anorexia [260] and completely inhibited leptin-induced BAT increase in UCP1 mRNA levels [225,261]. Furthermore, central administration of the MTII MC4R agonist had opposite effects, i.e., increasing BAT UCP1 mRNA levels by stimulating SNS activity [261]. Interestingly, this effect was blunted following sympathetic denervation [262]. Moreover, peripheral leptin treatment of MC4R-null mice was neither able to induce UCP1 expression in BAT [263] nor the leptin-dependent renal sympatho-excitatory response [264]. After HFD or cold exposure (external factors known to disturb leptin production), mice showed a diminished upregulation of BAT UCP1 [265]. Together, this suggests that leptin signalling in POMC neurons regulates in part the SNS outflow through an MC4R dependent mechanism.
The association between the melanocortin system and metabolic rate remains controversial. In this regard, the injection of MTII was shown to induce hypothermia/hypometabolism (achieved by decreasing BAT thermogenesis) before engendering the opposite effect (i.e., hyperthermia/hypermetabolism) [266]. In the same study, it was observed that this hypometabolic effect was preserved in MCR 1-, 3-, 4- and 5-knockout mice, indicating that these receptors did not mediate the hypothermic response of MTII. This effect was also observed using others melanocortin agonists [266]. However, MC4R seems to be essential for the induction of the thermogenic and cardiovascular effects of melanocortin system since the hyperthermic effect of MTII was lost in the MC4R knockout mice [261,264,267]. In summary, these data indicate that complex interactions in the ARC regulate leptin actions on thermogenesis, notably mediated by the melanocortin system. However, as demonstrated with the use of genetically modified models, other signalling pathways may be implicated in this regulation.

6.7. Leptin Actions in Extra-Hypothalamic Areas

Leptin also acts directly on other extra-hypothalamic areas, such as the NTS [230]. The neurons of the NTS receive inputs from vagal afferences and project locally within the brainstem and hypothalamic areas involved in the sympathetic regulation of BAT [268]. As NTS neurons express LepR, NTS specific leptin administration leads to a reduction of body weight associated with a reduction in food intake [269]. Although, it seems that leptin alone is unable to regulate BAT thermogenesis via the brainstem, a link between leptin and thyroid releasing hormone (TRH) in this region has been postulated to activate BAT thermogenesis [270,271,272,273]. Interestingly, in addition to the ARC, the NTS is the only other central region where POMC neurons are located. The possibility that POMC neurons in the NTS could be implicated in energy balance (as ARC POMC neurons) has been postulated; however, a study led by Huo and colleagues, found that leptin did not stimulate STAT3 phosphorylation or c-Fos expression in this area, concluding that POMC neurons in the ARC and the NTS were differentially regulated by leptin [257].

6.8. Browning and Leptin Central Action

Leptin participates in the regulation of the sympathetic tone of BAT and WAT, and its secretion by the adipocytes is directly controlled by the SNS [274,275,276]. The direct action of leptin in the browning process still remains incompletely understood. In the following section, we will review the different leptin-sensitive brain areas involved in this process.
Some evidence supports that central leptin stimulates gonadal WAT browning via the activation of PI3K signalling in the CNS [277]. Recent studies have shown that ICV leptin infusion resulted in a moderate increase in the expression of several browning genes in inguinal fat. Interestingly, the co-infusion of leptin and insulin induced a higher increase in WAT browning, mediated by POMC neurons located in the ARC and involving the PI3K signalling pathway [207]. The combined inactivation of protein tyrosine phosphatase 1B (PTP1B) and T-cell protein tyrosine phosphatase (TCPTP) stimulates leptin and insulin signalling in POMC neurons increasing browning and energy expenditure [207]. Additionally, the deletion of TCPTP in AGRP-expressing neurons in murine models had the same effects—increasing energy expenditure and browning, resulting in weight loss [278].
Browning is a dynamic process that is specific to different fat depots. O-linked β-N-acetylglucosamine (O-GlcNAc) is an intracellular carbohydrate implicated in different cellular processes, and its specific genetic ablation in AgRP neurons (via inhibition of neuronal excitability) promoted WAT browning. In contrast to this, the acute activation of AgRP neurons suppresses browning preferentially in retroperitoneal and inguinal WAT [279]. As mentioned above, melanocortin neurons are important targets for leptin-induced thermogenesis and accordingly, the deletion of MC4R in sympathetic preganglionic neurons revealed that they are essential factors not only for diet- and cold-induced thermogenesis but also for browning of inguinal WAT [280].
Forkhead box C2 protein (Foxc2), a transcription factor expressed in cardiac cells, mammary gland, liver and adipose tissue, has been shown to: (i) stimulate mitochondrial metabolism, (ii) promote brown adipocytes development [281] and (iii) induce browning in white adipocytes [282,283] by increasing UCP1 mRNA levels in WAT [284]. Recently, Foxc2 and leptin were shown to be functionally associated with the browning process. Foxc2 promotes the browning of WAT through LepR signalling; specifically, cAMP Response Element Binding protein (CREB), a positive transcriptional regulator of leptin, binds the leptin promoter region to potentiate the effects of Foxc2 on WAT browning through the JAK2/STAT3 signalling pathway that is necessary for the activation of the process [284].
Leptin plays a critical role in adipogenesis by regulating the Hedgehog (Hh) signalling pathway. Recently, it has been reported that leptin inhibited Hh signalling, promoting white adipocyte browning and decreasing the adipose weight of HFD-induced obese mice [285]. Since beige adipocytes have been recently identified, more studies linking leptin and browning are necessary to determine the exact role of leptin in this process.

6.9. Leptin Actions at Peripheral Level

Although central actions of leptin have been shown to play an important role in the regulation of energy expenditure, the existence of other LepR isoforms expressed widely in the periphery [108,124,286,287] suggests that leptin effects could also be mediated by peripheral tissues.
It has been demonstrated that leptin could have auto- and paracrine effects on adipocytes, potentially contributing to the weight- and fat-reducing activity of leptin [288]. Other authors have evaluated whether the leptin could modulate the cardiovascular system through a peripheral action, but seemingly the main effect remains mediated by the DMH LepR-expressing neurons [289]. Interestingly, LepRb expressing skeletal muscle is another tissue implicated in thermogenesis [108]. Dulloo and coworkers showed, that leptin could have direct thermogenic effects in skeletal muscle. By measuring oxygen consumption in ex vivo muscle samples, they observed that leptin also stimulated thermogenesis by a direct leptin-LepR interaction [290]. Although some studies have demonstrated that leptin could play a role at a peripheral level as stated by LepR expression in peripheral tissues, the main action of leptin seems to be mediated at a central level.

7. Leptin Polymorphism and Metabolic Implications

Several attempts have been made to show a possible association between leptin and LepR polymorphisms and metabolic disturbances leading to obesity. Specifically, the LEP A19G mutation in Lepob and LEPR Q223R, K109R and K656N mutations in LepR have been proposed to be associated with obesity; however, the first results obtained from a meta-analysis of published studies were not able to establish a strong link between leptin and LepR polymorphisms and obesity [291]. Interestingly, it has been found that some people with specific mutations in LepR (LEPR 109KK) tend to prefer sweet food, implying the need of a personalized medical intervention when treating obesity [292].
Additionally, single nucleotide polymorphisms (SNPs) that could influence metabolic features have been explored in several studies [293,294]. For example, research conducted on people in some specific Asia-Pacific regions, concluded that the Q223R LepR SNP could be associated with obesity [295,296,297,298] and type 2 diabetes [299]. Other interesting LepR polymorphisms such as K109R and K656N have suggested an association with childhood obesity [297]. These SNPs have also been shown to be strongly associated with obesity in a Chinese population when both were simultaneously present with a LEP 3’flanking region polymorphism [300].
SNP studies in an obesity-driven context have been performed all over the world covering in a variety of ethnic groups, but very little conclusive data have been obtained. While some studies concluded that there was a significant association between polymorphisms and obesity or other metabolic disorders [297], others subsequently failed to find any association [301,302,303]. For a more detailed summation, Lepob and LepR polymorphisms and their implications in obesity we would refer you to previously published excellent reviews on the subject [291,294]. Although more research needs to be conducted to elucidate the importance of polymorphisms in obesity development and occurrence, it remains an interesting factor to be considered for future anti-obesity therapeutic approaches.

8. Leptin as a Therapeutic Approach to Correct Obesity

Following its discovery [103], leptin was immediately described as the anti-obesity miracle cure. Since then, leptin-based therapies have been mainly developed for the treatment of congenital leptin deficiency [129,130], leptin deficiency in lipodystrophy patients [304] and hypothalamic amenorrhea [305,306] leading to the improvement of their phenotypes, including normalization of endocrine axes, decrease in insulin resistance and improvement of lipid profile and hepatic steatosis [307]. Unfortunately, the leptin resistance observed in obese rodents and humans [124,308,309,310] has dismissed the idea of leptin as a possible treatment to treat obesity. In the first clinical trial, a dose-response relationship with weight and fat loss was observed with subcutaneous recombinant leptin injections in both lean and obese subjects, even in those who were hyperleptinemic [311]. However, even with the highest doses of recombinant leptin, huge variability in the magnitude of weight loss among individuals was observed, once again raising the idea of leptin resistance. Other studies which combined leptin therapy with lifestyle management showed similar results in terms of dose response, with observable effects on body weight only with higher doses and after strict dietary interventions [312,313,314,315]. In agreement with these findings, no additional reduction of body weight was observed in overweight/obese hypoleptinemic patients after Roux-en-Y gastric bypass augmented with leptin therapy [316]. Of note, the increased circulating leptin levels after chronic therapy induces the production of anti-leptin antibodies in caloric restricted obese subjects [317].
Interestingly, recent preclinical evidence has suggested that leptin resistance associated with obesity could be overcome which course with compensatory hyperleptinaemia [318,319]. This causes reduced efficacy of leptin replacement therapies in obese patients. The concept of leptin resistance during obesity could be due to several molecular mechanisms. Firstly, leptin could induce the expression of SOCS3, which blocks the intracellular pathway downstream of leptin receptor signalling [320]. Furthermore, hypothalamic endoplasmic reticulum (ER) stress has been proposed as an important harmful mechanism for central leptin resistance [321]. On the other hand, some evidence support the idea of a central resistance to leptin during obesity which is associated to peripheral hyperleptinaemia, probably more attributable to a reduction in leptin transport across the blood-brain-barrier (BBB) than to hypothalamic leptin insensitivity which would decrease the available amount of leptin at a hypothalamic level [142,322]. However, leptin transport efficiency was not restored after caloric restriction despite hypoleptinaemia [323].
Therefore, prevention and reversion of leptin resistance present a great challenge for researchers and clinicians in the field, as well as the generation of animal models that could be extrapolated to human patients. In this regard, combinatorial therapies with different hormones regulating energy homeostasis are now being developed. Based on these developments, and as some pharmacological treatments are known to increase leptin responsiveness [318], research studies are now focusing on the development of new integrated pharmacological approaches to take advantage of hormonal and molecular synergisms to induce body weight loss. Some interesting findings have already been observed in leptin resistant diet-induced obese rats in which the co-treatment with amylin and leptin (described to have synergistic effects) induced weight loss in a fat-specific way. Of note, hypothalamic leptin signalling was partially restored with an up-regulation of basal and leptin-stimulated signalling in the hindbrain area postrema. In the same study, a similar effect was observed in obese and overweight subjects: the co-administration of recombinant human leptin and the amylin analog pramlintide elicited weight loss of a higher magnitude than both treatments alone [324]. In line with these results, obese and overweight patients subjected to hypocaloric diet and pramlintide treatment for 4 weeks followed by co-administration of metreleptin (a leptin analog) for 20 weeks showed sustained lower weight than patients treated with a monotherapy [325]. Despite these promising results, the clinical trial was halted due to safety concerns.
Remarkably, based on the finding that cholecystokinin (CCK) synergizes with amylin to inhibit food intake in lean mice [326], a triple combination of amylin/leptin/CCK was tested in diet-induced obese rats, resulting in increased body weight loss, reduction of food intake and adiposity than amylin/leptin administration alone [327].
This synergistic effect of leptin is not exclusive to amylin and/or CKK. In recent years, novel factors influencing leptin sensitivity have been explored in several animal studies. Interestingly, some studies have found that GLP-1 receptor agonists co-administered with leptin reduced food intake and body weight in a magnitude that none of the treatments separately could induce [328,329,330,331]. In this regard, when co-administered with leptin, liraglutide (a GLP-1 receptor agonist approved for the treatment of type 2 diabetes) presumably enhanced pSTAT3 after the inhibition of PTP1B [331]. Thus, it was found that the co-administration of high-potency leptin analogs with exedin-4 (glucagon-like peptide 1 receptor agonist) or fibroblast growth factor 21 (FGF21) in diet-induced obese mice (after a change to chow diet and 30% body weight loss) induced restoration of leptin responsiveness. Remarkably this effect was due to the drug co-administration, since leptin alone in animals with similar body weight loss was unable to recapitulate the observed effects [332]. However, these results were based on a change of diet, from HFD to chow diet, and taking this into account, the same group used a different approach in which the reduction in body weight was induced by the administration of a co-agonist targeting glucagon receptor and GLP-1 receptor followed by the administration of high-potency leptin analogs. This induced a greater reduction in body weight, food intake and improved glucose and lipid metabolism than the animals treated with GLP-1/glucagon molecule alone [333].
Research in this field is continuously developing new leptin sensitizers including: meta-chlorophenylpiperazine (an activator of (5-HT) 2c receptors) [334], oxytocin [335] or more recently, uroguanylin [336]. Although results have been promising, its appropriateness in terms of effectiveness and safety in humans remain to be proven. Nevertheless, it opens a promising door for the future development of an effective therapy against obesity.

9. Conclusions

The pressing need of effective anti-obesity treatments is driving research to identify new therapeutic targets. Since the discovery of BAT in adult humans [41,60,61,62], a great effort has been made to unravel the activators and molecular pathways leading to BAT activation. BAT is an important organ in the maintenance of body temperature through dissipation of heat by mitochondrial uncoupling, namely thermogenesis [17], and it has the potential capacity to reduce fat mass and regulate glycemic and lipid metabolism [73,74,75,76,77,78,79,80,83,84,85]. Several attempts have been made to target BAT activation in humans, with by far the most potent being cold-induced thermogenesis. However, the high variability of results necessitates an effective pharmacological intervention to satisfactorily activate BAT [62,65]. Due to its anti-obesogenic effect affecting both food intake and energy expenditure, leptin is one of the principal druggable targets to fight obesity and its comorbidities [129,130,318]. While leptin levels are correlated with adipose tissue mass, crucially many other factors can modulate its expression and secretion, as well as its interaction with LepR [110,111,337,338]. Leptin stimulates BAT thermogenesis through central LepRs acting mainly through SNS [188,189,190]. Several hypothalamic areas, such as the DMH, POA, PVH, VMH and ARC, as well as extra-hypothalamic areas as the NTS are involved in leptin-induced thermogenesis [172]. The effects mediated by melanocortin system neurons [141,255] and AgRP/NPY neurons [142,145] of the ARC are of great interest. The classical central regulation of BAT function involves SNS modulation, however the molecular details of that interaction remain elusive. In this regard, recent findings have challenged this view and are still under discussion [56,57,58]. It is worth noting that several lines of evidence support that the leptin/LepR–SNS axis plays a major role in the regulation of energy metabolism, however recent studies were not able to observe any thermogenic response to leptin [193,194,195,196,197,198,199], supporting the idea that the exact role of leptin on thermogenesis still remains unclear. Currently, leptin replacement therapy is being used successfully for the treatment of congenital leptin deficiency, leptin deficiency in lipodystrophy patients and hypothalamic amenorrhea only [129,130,304,305,306]. Attempts to use leptin by itself as an obesity treatment have been disregarded on account of the leptin resistance found in obese patients. Several approaches of combined therapy of leptin with other anti-obesogenic molecules are being tested in preclinical studies, such as amylin, CKK, GLP1, FGF21 and insulin, among others [324,325,326,327,328,329,331,332,333,339]. Although some promising results have been obtained, additional work will be necessary to validate if our current belief in the anti-obesogenic effect of thermogenesis will be clinically relevant for the treatment of obesity.

Author Contributions

Conceptualization, P.S.-C., N.M.-S. and C.C.; writing original draft, P.S.-C., N.M.-S. and C.C. writing review and editing, P.S.-C., N.M.-S., E.M. and C.C. All the authors have approved the final version of the article. All authors have read and agreed to the published version of the manuscript

Funding

This work was supported by grant from Santander-UCM (CC, PR87/19-22629). P.S.-C. is recipient of a fellowship from Xunta de Galicia (ED481B 2018/050). N.M.-S. is recipient of a fellowship from the European Union’s Horizon 2020 research and innovation program under the Marie Sklodowska-Curie actions. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Acknowledgments

We would like to thank Miguel Lopez for the great collaboration, as well as his support and mentoring. The authors also thank Samuel Malone (University of Oxford, United Kingdom) for his comments and English editing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Obesity Data. Available online: https://www.who.int/topics/obesity/en/ (accessed on 12 February 2020).
  2. Schneeberger, M.; Gomis, R.; Claret, M. Hypothalamic and brainstem neuronal circuits controlling homeostatic energy balance. J. Endocrinol. 2014, 220, T25–T46. [Google Scholar] [CrossRef] [Green Version]
  3. Tschop, M.H.; Finan, B.; Clemmensen, C.; Gelfanov, V.; Perez-Tilve, D.; Muller, T.D.; DiMarchi, R.D. Unimolecular Polypharmacy for Treatment of Diabetes and Obesity. Cell Metab. 2016, 24, 51–62. [Google Scholar] [CrossRef] [Green Version]
  4. Clemmensen, C.; Muller, T.D.; Woods, S.C.; Berthoud, H.R.; Seeley, R.J.; Tschop, M.H. Gut-Brain Cross-Talk in Metabolic Control. Cell 2017, 168, 758–774. [Google Scholar] [CrossRef] [Green Version]
  5. Lopez, M.; Tena-Sempere, M. Estradiol effects on hypothalamic AMPK and BAT thermogenesis: A gateway for obesity treatment? Pharmacol. Ther. 2017, 178, 109–122. [Google Scholar] [CrossRef]
  6. Cui, H.; Lopez, M.; Rahmouni, K. The cellular and molecular bases of leptin and ghrelin resistance in obesity. Nat. Rev. Endocrinol. 2017, 13, 338. [Google Scholar] [CrossRef]
  7. Hall, K.D.; Guo, J. Obesity Energetics: Body Weight Regulation and the Effects of Diet Composition. Gastroenterology 2017, 152, 1718–1727. [Google Scholar] [CrossRef] [Green Version]
  8. Sims, E.A.; Danforth, E., Jr.; Horton, E.S.; Bray, G.A.; Glennon, J.A.; Salans, L.B. Endocrine and metabolic effects of experimental obesity in man. Recent Prog. Horm. Res. 1973, 29, 457–496. [Google Scholar]
  9. Leibel, R.L.; Rosenbaum, M.; Hirsch, J. Changes in energy expenditure resulting from altered body weight. N. Engl. J. Med. 1995, 332, 621–628. [Google Scholar] [CrossRef]
  10. Ravussin, Y.; Leibel, R.L.; Ferrante, A.W., Jr. A missing link in body weight homeostasis: The catabolic signal of the overfed state. Cell Metab. 2014, 20, 565–572. [Google Scholar] [CrossRef] [Green Version]
  11. Fontaine, K.R.; Redden, D.T.; Wang, C.; Westfall, A.O.; Allison, D.B. Years of life lost due to obesity. JAMA 2003, 289, 187–193. [Google Scholar] [CrossRef]
  12. Bluher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef]
  13. Bellisari, A. Evolutionary origins of obesity. Obes. Rev. 2008, 9, 165–180. [Google Scholar] [CrossRef]
  14. Berthoud, H.R.; Morrison, C. The brain, appetite, and obesity. Annu. Rev. Psychol. 2008, 59, 55–92. [Google Scholar] [CrossRef]
  15. Landsberg, L.; Young, J.B.; Leonard, W.R.; Linsenmeier, R.A.; Turek, F.W. Is obesity associated with lower body temperatures? Core temperature: A forgotten variable in energy balance. Metabolism 2009, 58, 871–876. [Google Scholar] [CrossRef]
  16. Doucet, E.; Imbeault, P.; St-Pierre, S.; Almeras, N.; Mauriege, P.; Despres, J.P.; Bouchard, C.; Tremblay, A. Greater than predicted decrease in energy expenditure during exercise after body weight loss in obese men. Clin. Sci. 2003, 105, 89–95. [Google Scholar] [CrossRef]
  17. Cannon, B.; Nedergaard, J. Brown adipose tissue: Function and physiological significance. Physiol. Rev. 2004, 84, 277–359. [Google Scholar] [CrossRef]
  18. Labbe, S.M.; Caron, A.; Lanfray, D.; Monge-Rofarello, B.; Bartness, T.J.; Richard, D. Hypothalamic control of brown adipose tissue thermogenesis. Front. Syst. Neurosci. 2015, 9, 150. [Google Scholar] [CrossRef]
  19. Lim, S.; Honek, J.; Xue, Y.; Seki, T.; Cao, Z.; Andersson, P.; Yang, X.; Hosaka, K.; Cao, Y. Cold-induced activation of brown adipose tissue and adipose angiogenesis in mice. Nat. Protoc. 2012, 7, 606–615. [Google Scholar] [CrossRef]
  20. Boyer, P.D. The ATP synthase—A splendid molecular machine. Annu. Rev. Biochem. 1997, 66, 717–749. [Google Scholar] [CrossRef] [Green Version]
  21. Futai, M.; Noumi, T.; Maeda, M. ATP synthase (H+-ATPase): Results by combined biochemical and molecular biological approaches. Annu. Rev. Biochem. 1989, 58, 111–136. [Google Scholar] [CrossRef]
  22. von Ballmoos, C.; Wiedenmann, A.; Dimroth, P. Essentials for ATP synthesis by F1F0 ATP synthases. Annu. Rev. Biochem. 2009, 78, 649–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Nicholls, D.G.; Locke, R.M. Thermogenic mechanisms in brown fat. Physiol. Rev. 1984, 64, 1–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Silva, J.E. Thermogenic mechanisms and their hormonal regulation. Physiol. Rev. 2006, 86, 435–464. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, J.; Bostrom, P.; Sparks, L.M.; Ye, L.; Choi, J.H.; Giang, A.H.; Khandekar, M.; Virtanen, K.A.; Nuutila, P.; Schaart, G.; et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell 2012, 150, 366–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Timmons, J.A.; Wennmalm, K.; Larsson, O.; Walden, T.B.; Lassmann, T.; Petrovic, N.; Hamilton, D.L.; Gimeno, R.E.; Wahlestedt, C.; Baar, K.; et al. Myogenic gene expression signature establishes that brown and white adipocytes originate from distinct cell lineages. Proc. Natl. Acad. Sci. USA 2007, 104, 4401–4406. [Google Scholar] [CrossRef] [Green Version]
  27. Seale, P.; Bjork, B.; Yang, W.; Kajimura, S.; Chin, S.; Kuang, S.; Scime, A.; Devarakonda, S.; Conroe, H.M.; Erdjument-Bromage, H.; et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature 2008, 454, 961–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Tseng, Y.H.; Kokkotou, E.; Schulz, T.J.; Huang, T.L.; Winnay, J.N.; Taniguchi, C.M.; Tran, T.T.; Suzuki, R.; Espinoza, D.O.; Yamamoto, Y.; et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 2008, 454, 1000–1004. [Google Scholar] [CrossRef]
  29. Shan, T.; Liang, X.; Bi, P.; Zhang, P.; Liu, W.; Kuang, S. Distinct populations of adipogenic and myogenic Myf5-lineage progenitors in white adipose tissues. J. Lipid Res. 2013, 54, 2214–2224. [Google Scholar] [CrossRef] [Green Version]
  30. Sanchez-Gurmaches, J.; Guertin, D.A. Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat. Commun. 2014, 5, 4099. [Google Scholar] [CrossRef] [Green Version]
  31. Crisan, M.; Casteilla, L.; Lehr, L.; Carmona, M.; Paoloni-Giacobino, A.; Yap, S.; Sun, B.; Leger, B.; Logar, A.; Penicaud, L.; et al. A reservoir of brown adipocyte progenitors in human skeletal muscle. Stem Cells 2008, 26, 2425–2433. [Google Scholar] [CrossRef] [Green Version]
  32. Kajimura, S.; Saito, M. A new era in brown adipose tissue biology: Molecular control of brown fat development and energy homeostasis. Annu. Rev. Physiol. 2014, 76, 225–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Petrovic, N.; Walden, T.B.; Shabalina, I.G.; Timmons, J.A.; Cannon, B.; Nedergaard, J. Chronic peroxisome proliferator-activated receptor gamma (PPARgamma) 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. 2010, 285, 7153–7164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lee, P.; Werner, C.D.; Kebebew, E.; Celi, F.S. Functional thermogenic beige adipogenesis is inducible in human neck fat. Int. J. Obes. 2014, 38, 170–176. [Google Scholar] [CrossRef] [PubMed]
  35. Contreras, C.; Gonzalez, F.; Ferno, J.; Dieguez, C.; Rahmouni, K.; Nogueiras, R.; Lopez, M. The brain and brown fat. Ann. Med. 2015, 47, 150–168. [Google Scholar] [CrossRef] [Green Version]
  36. Walden, T.B.; Hansen, I.R.; Timmons, J.A.; Cannon, B.; Nedergaard, J. Recruited versus nonrecruited molecular signatures of brown, “brite” and white adipose tissues. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, E19–E31. [Google Scholar] [CrossRef] [Green Version]
  37. Shabalina, I.G.; Petrovic, N.; de Jong, J.M.; Kalinovich, A.V.; Cannon, B.; Nedergaard, J. UCP1 in brite/beige adipose tissue mitochondria is functionally thermogenic. Cell Rep. 2013, 5, 1196–1203. [Google Scholar] [CrossRef] [Green Version]
  38. Nedergaard, J.; Cannon, B. UCP1 mRNA does not produce heat. Biochim. Biophys. Acta 2013, 1831, 943–949. [Google Scholar] [CrossRef]
  39. Keipert, S.; Jastroch, M. Brite/beige fat and UCP1—Is it thermogenesis? Biochim. Biophys. Acta 2014, 1837, 1075–1082. [Google Scholar] [CrossRef] [Green Version]
  40. Lidell, M.E.; Betz, M.J.; Enerback, S. Two types of brown adipose tissue in humans. Adipocyte 2014, 3, 63–66. [Google Scholar] [CrossRef] [Green Version]
  41. Zingaretti, M.C.; Crosta, F.; Vitali, A.; Guerrieri, M.; Frontini, A.; Cannon, B.; Nedergaard, J.; Cinti, S. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. FASEB J. 2009, 23, 3113–3120. [Google Scholar] [CrossRef]
  42. Granneman, J.G. Norepinephrine infusions increase adenylate cyclase responsiveness in brown adipose tissue. J. Pharmacol. Exp. Ther. 1988, 245, 1075–1080. [Google Scholar] [PubMed]
  43. Whittle, A.J.; Lopez, M.; Vidal-Puig, A. Using brown adipose tissue to treat obesity - the central issue. Trends Mol. Med. 2011, 17, 405–411. [Google Scholar] [CrossRef] [PubMed]
  44. Villarroya, F.; Vidal-Puig, A. Beyond the sympathetic tone: The new brown fat activators. Cell Metab. 2013, 17, 638–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Lopez, M.; Varela, L.; Vazquez, M.J.; Rodriguez-Cuenca, S.; Gonzalez, C.R.; Velagapudi, V.R.; Morgan, D.A.; Schoenmakers, E.; Agassandian, K.; Lage, R.; et al. Hypothalamic AMPK and fatty acid metabolism mediate thyroid regulation of energy balance. Nat. Med. 2010, 16, 1001–1008. [Google Scholar] [CrossRef] [PubMed]
  46. Contreras, C.; Gonzalez-Garcia, I.; Martinez-Sanchez, N.; Seoane-Collazo, P.; Jacas, J.; Morgan, D.A.; Serra, D.; Gallego, R.; Gonzalez, F.; Casals, N.; et al. Central Ceramide-Induced Hypothalamic Lipotoxicity and ER Stress Regulate Energy Balance. Cell Rep. 2014, 9, 366–377. [Google Scholar] [CrossRef] [Green Version]
  47. Martinez de Morentin, P.B.; Gonzalez-Garcia, I.; Martins, L.; Lage, R.; Fernandez-Mallo, D.; Martinez-Sanchez, N.; Ruiz-Pino, F.; Liu, J.; Morgan, D.A.; Pinilla, L.; et al. Estradiol regulates brown adipose tissue thermogenesis via hypothalamic AMPK. Cell Metab. 2014, 20, 41–53. [Google Scholar] [CrossRef] [Green Version]
  48. Martinez-Sanchez, N.; Seoane-Collazo, P.; Contreras, C.; Varela, L.; Villarroya, J.; Rial-Pensado, E.; Buque, X.; Aurrekoetxea, I.; Delgado, T.C.; Vazquez-Martinez, R.; et al. Hypothalamic AMPK-ER Stress-JNK1 Axis Mediates the Central Actions of Thyroid Hormones on Energy Balance. Cell Metab. 2017, 26, 212–229. [Google Scholar] [CrossRef]
  49. Seoane-Collazo, P.; Roa, J.; Rial-Pensado, E.; Linares-Pose, L.; Beiroa, D.; Ruiz-Pino, F.; Lopez-Gonzalez, T.; Morgan, D.A.; Pardavila, J.A.; Sanchez-Tapia, M.J.; et al. SF1-Specific AMPKalpha1 Deletion Protects Against Diet-Induced Obesity. Diabetes 2018, 67, 2213–2226. [Google Scholar] [CrossRef] [Green Version]
  50. Seoane-Collazo, P.; Linares-Pose, L.; Rial-Pensado, E.; Romero-Pico, A.; Moreno-Navarrete, J.M.; Martinez-Sanchez, N.; Garrido-Gil, P.; Iglesias-Rey, R.; Morgan, D.A.; Tomasini, N.; et al. Central nicotine induces browning through hypothalamic kappa opioid receptor. Nat. Commun. 2019, 10, 4037. [Google Scholar] [CrossRef] [Green Version]
  51. Cao, W.; Medvedev, A.V.; Daniel, K.W.; Collins, S. beta-Adrenergic activation of p38 MAP kinase in adipocytes: cAMP induction of the uncoupling protein 1 (UCP1) gene requires p38 MAP kinase. J. Biol. Chem. 2001, 276, 27077–27082. [Google Scholar] [CrossRef] [Green Version]
  52. Holm, C.; Fredrikson, G.; Cannon, B.; Belfrage, P. Hormone-sensitive lipase in brown adipose tissue: identification and effect of cold exposure. Biosci. Rep. 1987, 7, 897–904. [Google Scholar] [CrossRef] [PubMed]
  53. Shih, M.F.; Taberner, P.V. Selective activation of brown adipocyte hormone-sensitive lipase and cAMP production in the mouse by beta 3-adrenoceptor agonists. Biochem. Pharmacol. 1995, 50, 601–608. [Google Scholar] [CrossRef]
  54. Clifford, G.M.; Londos, C.; Kraemer, F.B.; Vernon, R.G.; Yeaman, S.J. Translocation of hormone-sensitive lipase and perilipin upon lipolytic stimulation of rat adipocytes. J. Biol. Chem. 2000, 275, 5011–5015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Morimoto, C.; Kameda, K.; Tsujita, T.; Okuda, H. Relationships between lipolysis induced by various lipolytic agents and hormone-sensitive lipase in rat fat cells. J. Lipid Res. 2001, 42, 120–127. [Google Scholar] [PubMed]
  56. Cannon, B.; Nedergaard, J. What Ignites UCP1? Cell Metab. 2017, 26, 697–698. [Google Scholar] [CrossRef] [PubMed]
  57. Schreiber, R.; Diwoky, C.; Schoiswohl, G.; Feiler, U.; Wongsiriroj, N.; Abdellatif, M.; Kolb, D.; Hoeks, J.; Kershaw, E.E.; Sedej, S.; et al. Cold-Induced Thermogenesis Depends on ATGL-Mediated Lipolysis in Cardiac Muscle, but Not Brown Adipose Tissue. Cell Metab. 2017, 26, 753–763. [Google Scholar] [CrossRef]
  58. Shin, H.; Ma, Y.; Chanturiya, T.; Cao, Q.; Wang, Y.; Kadegowda, A.K.G.; Jackson, R.; Rumore, D.; Xue, B.; Shi, H.; et al. Lipolysis in Brown Adipocytes Is Not Essential for Cold-Induced Thermogenesis in Mice. Cell Metab. 2017, 26, 764–777. [Google Scholar] [CrossRef] [Green Version]
  59. Lowell, B.B.; Spiegelman, B.M. Towards a molecular understanding of adaptive thermogenesis. Nature 2000, 404, 652–660. [Google Scholar] [CrossRef]
  60. van Marken Lichtenbelt, W.D.; Vanhommerig, J.W.; Smulders, N.M.; Drossaerts, J.M.; Kemerink, G.J.; Bouvy, N.D.; Schrauwen, P.; Teule, G.J. Cold-activated brown adipose tissue in healthy men. N. Engl. J. Med. 2009, 360, 1500–1508. [Google Scholar] [CrossRef] [Green Version]
  61. Nedergaard, J.; Bengtsson, T.; Cannon, B. Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol. Endocrinol. Metab. 2007, 293, E444–E452. [Google Scholar] [CrossRef]
  62. Cypess, A.M.; Lehman, S.; Williams, G.; Tal, I.; Rodman, D.; Goldfine, A.B.; Kuo, F.C.; Palmer, E.L.; Tseng, Y.H.; Doria, A.; et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 2009, 360, 1509–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Jespersen, N.Z.; Larsen, T.J.; Peijs, L.; Daugaard, S.; Homoe, P.; Loft, A.; de Jong, J.; Mathur, N.; Cannon, B.; Nedergaard, J.; et al. A classical brown adipose tissue mRNA signature partly overlaps with brite in the supraclavicular region of adult humans. Cell Metab. 2013, 17, 798–805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Huttunen, P.; Hirvonen, J.; Kinnula, V. The occurrence of brown adipose tissue in outdoor workers. Eur. J. Appl. Physiol. Occup. Physiol. 1981, 46, 339–345. [Google Scholar] [CrossRef]
  65. Matsushita, M.; Yoneshiro, T.; Aita, S.; Kameya, T.; Sugie, H.; Saito, M. Impact of brown adipose tissue on body fatness and glucose metabolism in healthy humans. Int. J. Obes. 2014, 38, 812–817. [Google Scholar] [CrossRef] [PubMed]
  66. Heaton, J.M. The distribution of brown adipose tissue in the human. J. Anat. 1972, 112, 35–39. [Google Scholar] [PubMed]
  67. Yoneshiro, T.; Aita, S.; Matsushita, M.; Okamatsu-Ogura, Y.; Kameya, T.; Kawai, Y.; Miyagawa, M.; Tsujisaki, M.; Saito, M. Age-related decrease in cold-activated brown adipose tissue and accumulation of body fat in healthy humans. Obesity 2011, 19, 1755–1760. [Google Scholar] [CrossRef] [PubMed]
  68. Yoneshiro, T.; Aita, S.; Matsushita, M.; Kayahara, T.; Kameya, T.; Kawai, Y.; Iwanaga, T.; Saito, M. Recruited brown adipose tissue as an antiobesity agent in humans. J. Clin. Investig. 2013, 123, 3404–3408. [Google Scholar] [CrossRef] [Green Version]
  69. van der Lans, A.A.; Hoeks, J.; Brans, B.; Vijgen, G.H.; Visser, M.G.; Vosselman, M.J.; Hansen, J.; Jorgensen, J.A.; Wu, J.; Mottaghy, F.M.; et al. Cold acclimation recruits human brown fat and increases nonshivering thermogenesis. J. Clin. Investig. 2013, 123, 3395–3403. [Google Scholar] [CrossRef]
  70. Lee, J.H.; Wei, L.; Gu, X.; Wei, Z.; Dix, T.A.; Yu, S.P. Therapeutic effects of pharmacologically induced hypothermia against traumatic brain injury in mice. J. Neurotrauma 2014, 31, 1417–1430. [Google Scholar] [CrossRef] [Green Version]
  71. Saito, M.; Yoneshiro, T.; Matsushita, M. Activation and recruitment of brown adipose tissue by cold exposure and food ingredients in humans. Best Pract. Res. Clin. Endocrinol. Metab. 2016, 30, 537–547. [Google Scholar] [CrossRef]
  72. van Marken Lichtenbelt, W.D.; Schrauwen, P. Implications of nonshivering thermogenesis for energy balance regulation in humans. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 301, R285–R296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Vijgen, G.H.; Bouvy, N.D.; Leenen, L.; Rijkers, K.; Cornips, E.; Majoie, M.; Brans, B.; van Marken Lichtenbelt, W.D. Vagus nerve stimulation increases energy expenditure: Relation to brown adipose tissue activity. PLoS ONE 2013, 8, e77221. [Google Scholar] [CrossRef] [Green Version]
  74. Orava, J.; Nuutila, P.; Noponen, T.; Parkkola, R.; Viljanen, T.; Enerback, S.; Rissanen, A.; Pietilainen, K.H.; Virtanen, K.A. Blunted metabolic responses to cold and insulin stimulation in brown adipose tissue of obese humans. Obesity 2013, 21, 2279–2287. [Google Scholar] [CrossRef] [PubMed]
  75. Loh, R.K.C.; Kingwell, B.A.; Carey, A.L. Human brown adipose tissue as a target for obesity management; beyond cold-induced thermogenesis. Obes. Rev. 2017, 18, 1227–1242. [Google Scholar] [CrossRef] [PubMed]
  76. Ouellet, V.; Labbe, S.M.; Blondin, D.P.; Phoenix, S.; Guerin, B.; Haman, F.; Turcotte, E.E.; Richard, D.; Carpentier, A.C. Brown adipose tissue oxidative metabolism contributes to energy expenditure during acute cold exposure in humans. J. Clin. Investig. 2012, 122, 545–552. [Google Scholar] [CrossRef] [PubMed]
  77. Orava, J.; Nuutila, P.; Lidell, M.E.; Oikonen, V.; Noponen, T.; Viljanen, T.; Scheinin, M.; Taittonen, M.; Niemi, T.; Enerback, S.; et al. Different metabolic responses of human brown adipose tissue to activation by cold and insulin. Cell Metab. 2011, 14, 272–279. [Google Scholar] [CrossRef] [Green Version]
  78. Chondronikola, M.; Volpi, E.; Børsheim, E.; Porter, C.; Annamalai, P.; Enerbäck, S.; Lidell, M.E.; Saraf, M.K.; Labbe, S.M.; Hurren, N.M.; et al. Brown Adipose Tissue Improves Whole-Body Glucose Homeostasis and Insulin Sensitivity in Humans. Diabetes 2014, 63, 4089–4099. [Google Scholar] [CrossRef] [Green Version]
  79. Lee, P.; Smith, S.; Linderman, J.; Courville, A.B.; Brychta, R.J.; Dieckmann, W.; Werner, C.D.; Chen, K.Y.; Celi, F.S. Temperature-acclimated brown adipose tissue modulates insulin sensitivity in humans. Diabetes 2014, 63, 3686–3698. [Google Scholar] [CrossRef] [Green Version]
  80. Lee, P.; Bova, R.; Schofield, L.; Bryant, W.; Dieckmann, W.; Slattery, A.; Govendir, M.A.; Emmett, L.; Greenfield, J.R. Brown Adipose Tissue Exhibits a Glucose-Responsive Thermogenic Biorhythm in Humans. Cell Metab. 2016, 23, 602–609. [Google Scholar] [CrossRef]
  81. Hanssen, M.J.; van der Lans, A.A.; Brans, B.; Hoeks, J.; Jardon, K.M.; Schaart, G.; Mottaghy, F.M.; Schrauwen, P.; van Marken Lichtenbelt, W.D. Short-term Cold Acclimation Recruits Brown Adipose Tissue in Obese Humans. Diabetes 2016, 65, 1179–1189. [Google Scholar] [CrossRef] [Green Version]
  82. Hanssen, M.J.; Hoeks, J.; Brans, B.; van der Lans, A.A.; Schaart, G.; van den Driessche, J.J.; Jorgensen, J.A.; Boekschoten, M.V.; Hesselink, M.K.; Havekes, B.; et al. Short-term cold acclimation improves insulin sensitivity in patients with type 2 diabetes mellitus. Nat. Med. 2015, 21, 863–865. [Google Scholar] [CrossRef] [PubMed]
  83. Blondin, D.P.; Labbe, S.M.; Noll, C.; Kunach, M.; Phoenix, S.; Guerin, B.; Turcotte, E.E.; Haman, F.; Richard, D.; Carpentier, A.C. Selective Impairment of Glucose but Not Fatty Acid or Oxidative Metabolism in Brown Adipose Tissue of Subjects With Type 2 Diabetes. Diabetes 2015, 64, 2388–2397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Blondin, D.P.; Frisch, F.; Phoenix, S.; Guerin, B.; Turcotte, E.E.; Haman, F.; Richard, D.; Carpentier, A.C. Inhibition of Intracellular Triglyceride Lipolysis Suppresses Cold-Induced Brown Adipose Tissue Metabolism and Increases Shivering in Humans. Cell Metab. 2017, 25, 438–447. [Google Scholar] [CrossRef] [Green Version]
  85. Chondronikola, M.; Volpi, E.; Børsheim, E.; Porter, C.; Saraf, M.K.; Annamalai, P.; Yfanti, C.; Chao, T.; Wong, D.; Shinoda, K.; et al. Brown Adipose Tissue Activation Is Linked to Distinct Systemic Effects on Lipid Metabolism in Humans. Cell Metab. 2016, 23, 1200–1206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. De Lorenzo, F.; Mukherjee, M.; Kadziola, Z.; Sherwood, R.; Kakkar, V.V. Central cooling effects in patients with hypercholesterolaemia. Clin. Sci. 1998, 95, 213–217. [Google Scholar] [CrossRef] [PubMed]
  87. Thyagarajan, B.; Foster, M.T. Beiging of white adipose tissue as a therapeutic strategy for weight loss in humans. Horm. Mol. Biol. Clin. Investig. 2017, 31. [Google Scholar] [CrossRef] [PubMed]
  88. Wijers, S.L.; Saris, W.H.; van Marken Lichtenbelt, W.D. Cold-induced adaptive thermogenesis in lean and obese. Obesity 2010, 18, 1092–1099. [Google Scholar] [CrossRef]
  89. Arch, J.R. The beta 3-adrenergic system and beta 3-adrenergic agonists. Rev. Endocr. Metab. Disord. 2001, 2, 385–393. [Google Scholar] [CrossRef]
  90. Cypess, A.M.; Weiner, L.S.; Roberts-Toler, C.; Franquet Elia, E.; Kessler, S.H.; Kahn, P.A.; English, J.; Chatman, K.; Trauger, S.A.; Doria, A.; et al. Activation of human brown adipose tissue by a beta3-adrenergic receptor agonist. Cell Metab. 2015, 21, 33–38. [Google Scholar] [CrossRef] [Green Version]
  91. Finlin, B.S.; Memetimin, H.; Confides, A.L.; Kasza, I.; Zhu, B.; Vekaria, H.J.; Harfmann, B.; Jones, K.A.; Johnson, Z.R.; Westgate, P.M.; et al. Human adipose beiging in response to cold and mirabegron. JCI Insight 2018, 3. [Google Scholar] [CrossRef]
  92. Cypess, A.M.; Chen, Y.C.; Sze, C.; Wang, K.; English, J.; Chan, O.; Holman, A.R.; Tal, I.; Palmer, M.R.; Kolodny, G.M.; et al. Cold but not sympathomimetics activates human brown adipose tissue in vivo. Proc. Natl. Acad. Sci. USA 2012, 109, 10001–10005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yoneshiro, T.; Aita, S.; Kawai, Y.; Iwanaga, T.; Saito, M. Nonpungent capsaicin analogs (capsinoids) increase energy expenditure through the activation of brown adipose tissue in humans. Am. J. Clin. Nutr. 2012, 95, 845–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Galgani, J.E.; Ryan, D.H.; Ravussin, E. Effect of capsinoids on energy metabolism in human subjects. Br. J. Nutr. 2010, 103, 38–42. [Google Scholar] [CrossRef] [PubMed]
  95. Sun, L.; Camps, S.G.; Goh, H.J.; Govindharajulu, P.; Schaefferkoetter, J.D.; Townsend, D.W.; Verma, S.K.; Velan, S.S.; Sun, L.; Sze, S.K.; et al. Capsinoids activate brown adipose tissue (BAT) with increased energy expenditure associated with subthreshold 18-fluorine fluorodeoxyglucose uptake in BAT-positive humans confirmed by positron emission tomography scan. Am. J. Clin. Nutr. 2018, 107, 62–70. [Google Scholar] [CrossRef] [PubMed]
  96. Nirengi, S.; Homma, T.; Inoue, N.; Sato, H.; Yoneshiro, T.; Matsushita, M.; Kameya, T.; Sugie, H.; Tsuzaki, K.; Saito, M.; et al. Assessment of human brown adipose tissue density during daily ingestion of thermogenic capsinoids using near-infrared time-resolved spectroscopy. J. Biomed. Opt. 2016, 21, 091305. [Google Scholar] [CrossRef] [Green Version]
  97. Broeders, E.P.; Nascimento, E.B.; Havekes, B.; Brans, B.; Roumans, K.H.; Tailleux, A.; Schaart, G.; Kouach, M.; Charton, J.; Deprez, B.; et al. The Bile Acid Chenodeoxycholic Acid Increases Human Brown Adipose Tissue Activity. Cell Metab. 2015, 22, 418–426. [Google Scholar] [CrossRef] [Green Version]
  98. Labruna, G.; Pasanisi, F.; Fortunato, G.; Nardelli, C.; Finelli, C.; Farinaro, E.; Contaldo, F.; Sacchetti, L. Sequence Analysis of the UCP1 Gene in a Severe Obese Population from Southern Italy. J. Obes. 2011, 2011, 269043. [Google Scholar] [CrossRef]
  99. Chathoth, S.; Ismail, M.H.; Vatte, C.; Cyrus, C.; Al Ali, Z.; Ahmed, K.A.; Acharya, S.; Al Barqi, A.M.; Al Ali, A. Association of Uncoupling Protein 1 (UCP1) gene polymorphism with obesity: A case-control study. BMC Med. Genet. 2018, 19, 203. [Google Scholar] [CrossRef]
  100. Samano, R.; Huesca-Gomez, C.; Lopez-Marure, R.; Hernandez-Cabrera, A.K.; Rodriguez-Ventura, A.; Tolentino, M.; Morales, R.M.; Gamboa, R. Association between UCP polymorphisms and adipokines with obesity in Mexican adolescents. J. Pediatr. Endocrinol. Metab. 2018, 31, 561–568. [Google Scholar] [CrossRef]
  101. Siiteri, P.K. Adipose tissue as a source of hormones. Am. J. Clin. Nutr. 1987, 45, 277–282. [Google Scholar] [CrossRef]
  102. Flier, J.S.; Cook, K.S.; Usher, P.; Spiegelman, B.M. Severely impaired adipsin expression in genetic and acquired obesity. Science 1987, 237, 405–408. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, Y.; Proenca, R.; Maffei, M.; Barone, M.; Leopold, L.; Friedman, J.M. Positional cloning of the mouse obese gene and its human homologue. Nature 1994, 372, 425–432. [Google Scholar] [CrossRef] [PubMed]
  104. Kershaw, E.E.; Flier, J.S. Adipose tissue as an endocrine organ. J. Clin. Endocrinol. Metab. 2004, 89, 2548–2556. [Google Scholar] [CrossRef] [PubMed]
  105. Coleman, D.L. Effects of parabiosis of obese with diabetes and normal mice. Diabetologia 1973, 9, 294–298. [Google Scholar] [CrossRef]
  106. Chen, H.; Charlat, O.; Tartaglia, L.A.; Woolf, E.A.; Weng, X.; Ellis, S.J.; Lakey, N.D.; Culpepper, J.; Moore, K.J.; Breitbart, R.E.; et al. Evidence that the diabetes gene encodes the leptin receptor: Identification of a mutation in the leptin receptor gene in db/db mice. Cell 1996, 84, 491–495. [Google Scholar] [CrossRef] [Green Version]
  107. Lee, G.H.; Proenca, R.; Montez, J.M.; Carroll, K.M.; Darvishzadeh, J.G.; Lee, J.I.; Friedman, J.M. Abnormal splicing of the leptin receptor in diabetic mice. Nature 1996, 379, 632–635. [Google Scholar] [CrossRef]
  108. Tartaglia, L.A.; Dembski, M.; Weng, X.; Deng, N.; Culpepper, J.; Devos, R.; Richards, G.J.; Campfield, L.A.; Clark, F.T.; Deeds, J.; et al. Identification and expression cloning of a leptin receptor, OB-R. Cell 1995, 83, 1263–1271. [Google Scholar] [CrossRef] [Green Version]
  109. Maffei, M.; Fei, H.; Lee, G.H.; Dani, C.; Leroy, P.; Zhang, Y.; Proenca, R.; Negrel, R.; Ailhaud, G.; Friedman, J.M. Increased expression in adipocytes of ob RNA in mice with lesions of the hypothalamus and with mutations at the db locus. Proc. Natl. Acad. Sci. USA 1995, 92, 6957–6960. [Google Scholar] [CrossRef] [Green Version]
  110. Fain, J.N.; Madan, A.K.; Hiler, M.L.; Cheema, P.; Bahouth, S.W. Comparison of the release of adipokines by adipose tissue, adipose tissue matrix, and adipocytes from visceral and subcutaneous abdominal adipose tissues of obese humans. Endocrinology 2004, 145, 2273–2282. [Google Scholar] [CrossRef] [Green Version]
  111. Wajchenberg, B.L. Subcutaneous and visceral adipose tissue: Their relation to the metabolic syndrome. Endocr. Rev. 2000, 21, 697–738. [Google Scholar] [CrossRef]
  112. Muruzabal, F.J.; Fruhbeck, G.; Gomez-Ambrosi, J.; Archanco, M.; Burrell, M.A. Immunocytochemical detection of leptin in non-mammalian vertebrate stomach. Gen. Comp. Endocrinol. 2002, 128, 149–152. [Google Scholar] [CrossRef]
  113. Allison, M.B.; Myers, M.G., Jr. 20 years of leptin: Connecting leptin signaling to biological function. J. Endocrinol. 2014, 223, T25–T35. [Google Scholar] [CrossRef]
  114. Chehab, F.F. 20 years of leptin: Leptin and reproduction: Past milestones, present undertakings, and future endeavors. J. Endocrinol. 2014, 223, T37–T48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Chou, S.H.; Mantzoros, C. 20 years of leptin: Role of leptin in human reproductive disorders. J. Endocrinol. 2014, 223, T49–T62. [Google Scholar] [CrossRef] [PubMed]
  116. DePaoli, A.M. 20 years of leptin: Leptin in common obesity and associated disorders of metabolism. J. Endocrinol. 2014, 223, T71–T81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Farooqi, I.S.; O’Rahilly, S. 20 years of leptin: Human disorders of leptin action. J. Endocrinol. 2014, 223, T63–T70. [Google Scholar] [CrossRef] [PubMed]
  118. Friedman, J. 20 years of leptin: Leptin at 20: An overview. J. Endocrinol. 2014, 223, T1–T8. [Google Scholar] [CrossRef] [Green Version]
  119. Gautron, L.; Elmquist, J.K. Sixteen years and counting: An update on leptin in energy balance. J. Clin. Investig. 2011, 121, 2087–2093. [Google Scholar] [CrossRef] [Green Version]
  120. Peelman, F.; Zabeau, L.; Moharana, K.; Savvides, S.N.; Tavernier, J. 20 years of leptin: Insights into signaling assemblies of the leptin receptor. J. Endocrinol. 2014, 223, T9–T23. [Google Scholar] [CrossRef]
  121. Rosenbaum, M.; Leibel, R.L. 20 years of leptin: Role of leptin in energy homeostasis in humans. J. Endocrinol. 2014, 223, T83–T96. [Google Scholar] [CrossRef]
  122. Friedman, J.M.; Leibel, R.L.; Siegel, D.S.; Walsh, J.; Bahary, N. Molecular mapping of the mouse ob mutation. Genomics 1991, 11, 1054–1062. [Google Scholar] [CrossRef]
  123. Takaya, K.; Ogawa, Y.; Isse, N.; Okazaki, T.; Satoh, N.; Masuzaki, H.; Mori, K.; Tamura, N.; Hosoda, K.; Nakao, K. Molecular cloning of rat leptin receptor isoform complementary DNAs--identification of a missense mutation in Zucker fatty (fa/fa) rats. Biochem. Biophys. Res. Commun. 1996, 225, 75–83. [Google Scholar] [CrossRef] [PubMed]
  124. Halaas, J.L.; Boozer, C.; Blair-West, J.; Fidahusein, N.; Denton, D.A.; Friedman, J.M. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc. Natl. Acad. Sci. USA 1997, 94, 8878–8883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Halaas, J.L.; Gajiwala, K.S.; Maffei, M.; Cohen, S.L.; Chait, B.T.; Rabinowitz, D.; Lallone, R.L.; Burley, S.K.; Friedman, J.M. Weight-reducing effects of the plasma protein encoded by the obese gene. Science 1995, 269, 543–546. [Google Scholar] [CrossRef] [PubMed]
  126. Pelleymounter, M.A.; Cullen, M.J.; Baker, M.B.; Hecht, R.; Winters, D.; Boone, T.; Collins, F. Effects of the obese gene product on body weight regulation in ob/ob mice. Science 1995, 269, 540–543. [Google Scholar] [CrossRef]
  127. Stephens, T.W.; Basinski, M.; Bristow, P.K.; Bue-Valleskey, J.M.; Burgett, S.G.; Craft, L.; Hale, J.; Hoffmann, J.; Hsiung, H.M.; Kriauciunas, A.; et al. The role of neuropeptide Y in the antiobesity action of the obese gene product. Nature 1995, 377, 530–532. [Google Scholar] [CrossRef]
  128. Cohen, S.L.; Halaas, J.L.; Friedman, J.M.; Chait, B.T.; Bennett, L.; Chang, D.; Hecht, R.; Collins, F. Human leptin characterization. Nature 1996, 382, 589. [Google Scholar] [CrossRef]
  129. Farooqi, I.S.; Jebb, S.A.; Langmack, G.; Lawrence, E.; Cheetham, C.H.; Prentice, A.M.; Hughes, I.A.; McCamish, M.A.; O’Rahilly, S. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N. Engl. J. Med. 1999, 341, 879–884. [Google Scholar] [CrossRef]
  130. Farooqi, I.S.; Matarese, G.; Lord, G.M.; Keogh, J.M.; Lawrence, E.; Agwu, C.; Sanna, V.; Jebb, S.A.; Perna, F.; Fontana, S.; et al. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J. Clin. Investig. 2002, 110, 1093–1103. [Google Scholar] [CrossRef]
  131. Licinio, J.; Caglayan, S.; Ozata, M.; Yildiz, B.O.; de Miranda, P.B.; O’Kirwan, F.; Whitby, R.; Liang, L.; Cohen, P.; Bhasin, S.; et al. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc. Natl. Acad. Sci. USA 2004, 101, 4531–4536. [Google Scholar] [CrossRef] [Green Version]
  132. Elmquist, J.K.; Bjorbaek, C.; Ahima, R.S.; Flier, J.S.; Saper, C.B. Distributions of leptin receptor mRNA isoforms in the rat brain. J. Comp. Neurol. 1998, 395, 535–547. [Google Scholar] [CrossRef]
  133. Scott, M.M.; Lachey, J.L.; Sternson, S.M.; Lee, C.E.; Elias, C.F.; Friedman, J.M.; Elmquist, J.K. Leptin targets in the mouse brain. J. Comp. Neurol. 2009, 514, 518–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Schwartz, M.W.; Seeley, R.J.; Campfield, L.A.; Burn, P.; Baskin, D.G. Identification of targets of leptin action in rat hypothalamus. J. Clin. Investig. 1996, 98, 1101–1106. [Google Scholar] [CrossRef] [PubMed]
  135. Mercer, J.G.; Hoggard, N.; Williams, L.M.; Lawrence, C.B.; Hannah, L.T.; Trayhurn, P. Localization of leptin receptor mRNA and the long form splice variant (Ob-Rb) in mouse hypothalamus and adjacent brain regions by in situ hybridization. FEBS Lett. 1996, 387, 113–116. [Google Scholar] [CrossRef] [Green Version]
  136. Morton, G.J.; Niswender, K.D.; Rhodes, C.J.; Myers, M.G., Jr.; Blevins, J.E.; Baskin, D.G.; Schwartz, M.W. Arcuate nucleus-specific leptin receptor gene therapy attenuates the obesity phenotype of Koletsky (fa(k)/fa(k)) rats. Endocrinology 2003, 144, 2016–2024. [Google Scholar] [CrossRef]
  137. Elias, C.F.; Lee, C.; Kelly, J.; Aschkenasi, C.; Ahima, R.S.; Couceyro, P.R.; Kuhar, M.J.; Saper, C.B.; Elmquist, J.K. Leptin activates hypothalamic CART neurons projecting to the spinal cord. Neuron 1998, 21, 1375–1385. [Google Scholar] [CrossRef] [Green Version]
  138. Balthasar, N.; Coppari, R.; McMinn, J.; Liu, S.M.; Lee, C.E.; Tang, V.; Kenny, C.D.; McGovern, R.A.; Chua, S.C., Jr.; Elmquist, J.K.; et al. Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron 2004, 42, 983–991. [Google Scholar] [CrossRef] [Green Version]
  139. Cowley, M.A.; Smart, J.L.; Rubinstein, M.; Cerdan, M.G.; Diano, S.; Horvath, T.L.; Cone, R.D.; Low, M.J. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 2001, 411, 480–484. [Google Scholar] [CrossRef]
  140. Fan, W.; Boston, B.A.; Kesterson, R.A.; Hruby, V.J.; Cone, R.D. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 1997, 385, 165–168. [Google Scholar] [CrossRef]
  141. Schwartz, M.W.; Seeley, R.J.; Woods, S.C.; Weigle, D.S.; Campfield, L.A.; Burn, P.; Baskin, D.G. Leptin increases hypothalamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus. Diabetes 1997, 46, 2119–2123. [Google Scholar] [CrossRef] [Green Version]
  142. Morris, D.L.; Rui, L. Recent advances in understanding leptin signaling and leptin resistance. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E1247–E1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Myers, M.G., Jr.; Munzberg, H.; Leinninger, G.M.; Leshan, R.L. The geometry of leptin action in the brain: More complicated than a simple ARC. Cell Metab. 2009, 9, 117–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Hahn, T.M.; Breininger, J.F.; Baskin, D.G.; Schwartz, M.W. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nat. Neurosci. 1998, 1, 271–272. [Google Scholar] [CrossRef] [PubMed]
  145. Myers, M.G.; Cowley, M.A.; Munzberg, H. Mechanisms of leptin action and leptin resistance. Annu. Rev. Physiol. 2008, 70, 537–556. [Google Scholar] [CrossRef] [Green Version]
  146. Baumann, H.; Morella, K.K.; White, D.W.; Dembski, M.; Bailon, P.S.; Kim, H.; Lai, C.F.; Tartaglia, L.A. The full-length leptin receptor has signaling capabilities of interleukin 6-type cytokine receptors. Proc. Natl. Acad. Sci. USA 1996, 93, 8374–8378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Chua, S.C., Jr.; Chung, W.K.; Wu-Peng, X.S.; Zhang, Y.; Liu, S.M.; Tartaglia, L.; Leibel, R.L. Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science 1996, 271, 994–996. [Google Scholar] [CrossRef] [PubMed]
  148. Friedman, J.M.; Halaas, J.L. Leptin and the regulation of body weight in mammals. Nature 1998, 395, 763–770. [Google Scholar] [CrossRef]
  149. Bates, S.H.; Myers, M.G., Jr. The role of leptin receptor signaling in feeding and neuroendocrine function. Trends Endocrinol. Metab. 2003, 14, 447–452. [Google Scholar] [CrossRef]
  150. Uotani, S.; Bjorbaek, C.; Tornoe, J.; Flier, J.S. Functional properties of leptin receptor isoforms: Internalization and degradation of leptin and ligand-induced receptor downregulation. Diabetes 1999, 48, 279–286. [Google Scholar] [CrossRef]
  151. Schaab, M.; Kausch, H.; Klammt, J.; Nowicki, M.; Anderegg, U.; Gebhardt, R.; Rose-John, S.; Scheller, J.; Thiery, J.; Kratzsch, J. Novel regulatory mechanisms for generation of the soluble leptin receptor: Implications for leptin action. PLoS ONE 2012, 7, e34787. [Google Scholar] [CrossRef] [Green Version]
  152. Bjorbaek, C.; Uotani, S.; da Silva, B.; Flier, J.S. Divergent signaling capacities of the long and short isoforms of the leptin receptor. J. Biol. Chem. 1997, 272, 32686–32695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Ghilardi, N.; Skoda, R.C. The leptin receptor activates janus kinase 2 and signals for proliferation in a factor-dependent cell line. Mol. Endocrinol. 1997, 11, 393–399. [Google Scholar] [CrossRef] [PubMed]
  154. Li, C.; Friedman, J.M. Leptin receptor activation of SH2 domain containing protein tyrosine phosphatase 2 modulates Ob receptor signal transduction. Proc. Natl. Acad. Sci. USA 1999, 96, 9677–9682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Banks, A.S.; Davis, S.M.; Bates, S.H.; Myers, M.G., Jr. Activation of downstream signals by the long form of the leptin receptor. J. Biol. Chem. 2000, 275, 14563–14572. [Google Scholar] [CrossRef] [Green Version]
  156. Bates, S.H.; Stearns, W.H.; Dundon, T.A.; Schubert, M.; Tso, A.W.; Wang, Y.; Banks, A.S.; Lavery, H.J.; Haq, A.K.; Maratos-Flier, E.; et al. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature 2003, 421, 856–859. [Google Scholar] [CrossRef]
  157. Gao, Q.; Wolfgang, M.J.; Neschen, S.; Morino, K.; Horvath, T.L.; Shulman, G.I.; Fu, X.Y. Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation. Proc. Natl. Acad. Sci. USA 2004, 101, 4661–4666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Buettner, C.; Pocai, A.; Muse, E.D.; Etgen, A.M.; Myers, M.G., Jr.; Rossetti, L. Critical role of STAT3 in leptin’s metabolic actions. Cell Metab. 2006, 4, 49–60. [Google Scholar] [CrossRef] [Green Version]
  159. Zhang, J.; Scarpace, P.J. The soluble leptin receptor neutralizes leptin-mediated STAT3 signalling and anorexic responses in vivo. Br. J. Pharmacol. 2009, 158, 475–482. [Google Scholar] [CrossRef] [Green Version]
  160. Rahmouni, K.; Sigmund, C.D.; Haynes, W.G.; Mark, A.L. Hypothalamic ERK mediates the anorectic and thermogenic sympathetic effects of leptin. Diabetes 2009, 58, 536–542. [Google Scholar] [CrossRef] [Green Version]
  161. Bjorbak, C.; Lavery, H.J.; Bates, S.H.; Olson, R.K.; Davis, S.M.; Flier, J.S.; Myers, M.G., Jr. SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985. J. Biol. Chem. 2000, 275, 40649–40657. [Google Scholar] [CrossRef] [Green Version]
  162. Bjornholm, M.; Munzberg, H.; Leshan, R.L.; Villanueva, E.C.; Bates, S.H.; Louis, G.W.; Jones, J.C.; Ishida-Takahashi, R.; Bjorbaek, C.; Myers, M.G., Jr. Mice lacking inhibitory leptin receptor signals are lean with normal endocrine function. J. Clin. Investig. 2007, 117, 1354–1360. [Google Scholar] [CrossRef] [PubMed]
  163. Munzberg, H.; Morrison, C.D. Structure, production and signaling of leptin. Metabolism 2015, 64, 13–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Kellerer, M.; Koch, M.; Metzinger, E.; Mushack, J.; Capp, E.; Haring, H.U. Leptin activates PI-3 kinase in C2C12 myotubes via janus kinase-2 (JAK-2) and insulin receptor substrate-2 (IRS-2) dependent pathways. Diabetologia 1997, 40, 1358–1362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Kim, Y.B.; Uotani, S.; Pierroz, D.D.; Flier, J.S.; Kahn, B.B. In vivo administration of leptin activates signal transduction directly in insulin-sensitive tissues: Overlapping but distinct pathways from insulin. Endocrinology 2000, 141, 2328–2339. [Google Scholar] [CrossRef] [PubMed]
  166. Anderwald, C.; Muller, G.; Koca, G.; Furnsinn, C.; Waldhausl, W.; Roden, M. Short-term leptin-dependent inhibition of hepatic gluconeogenesis is mediated by insulin receptor substrate-2. Mol. Endocrinol. 2002, 16, 1612–1628. [Google Scholar] [CrossRef]
  167. Carvalheira, J.B.; Ribeiro, E.B.; Folli, F.; Velloso, L.A.; Saad, M.J. Interaction between leptin and insulin signaling pathways differentially affects JAK-STAT and PI 3-kinase-mediated signaling in rat liver. Biol. Chem. 2003, 384, 151–159. [Google Scholar] [CrossRef]
  168. Duan, C.; Li, M.; Rui, L. SH2-B promotes insulin receptor substrate 1 (IRS1)- and IRS2-mediated activation of the phosphatidylinositol 3-kinase pathway in response to leptin. J. Biol. Chem. 2004, 279, 43684–43691. [Google Scholar] [CrossRef] [Green Version]
  169. Wauman, J.; Zabeau, L.; Tavernier, J. The Leptin Receptor Complex: Heavier Than Expected? Front. Endocrinol. 2017, 8, 30. [Google Scholar] [CrossRef] [Green Version]
  170. Kim, M.S.; Pak, Y.K.; Jang, P.G.; Namkoong, C.; Choi, Y.S.; Won, J.C.; Kim, K.S.; Kim, S.W.; Kim, H.S.; Park, J.Y.; et al. Role of hypothalamic Foxo1 in the regulation of food intake and energy homeostasis. Nat. Neurosci. 2006, 9, 901–906. [Google Scholar] [CrossRef]
  171. Plum, L.; Lin, H.V.; Dutia, R.; Tanaka, J.; Aizawa, K.S.; Matsumoto, M.; Kim, A.J.; Cawley, N.X.; Paik, J.H.; Loh, Y.P.; et al. The obesity susceptibility gene Cpe links FoxO1 signaling in hypothalamic pro-opiomelanocortin neurons with regulation of food intake. Nat. Med. 2009, 15, 1195–1201. [Google Scholar] [CrossRef] [Green Version]
  172. Kwon, O.; Kim, K.W.; Kim, M.S. Leptin signalling pathways in hypothalamic neurons. Cell. Mol. Life Sci. 2016, 73, 1457–1477. [Google Scholar] [CrossRef] [PubMed]
  173. Kitamura, T.; Feng, Y.; Kitamura, Y.I.; Chua, S.C., Jr.; Xu, A.W.; Barsh, G.S.; Rossetti, L.; Accili, D. Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake. Nat. Med. 2006, 12, 534–540. [Google Scholar] [CrossRef] [PubMed]
  174. Blouet, C.; Ono, H.; Schwartz, G.J. Mediobasal hypothalamic p70 S6 kinase 1 modulates the control of energy homeostasis. Cell Metab. 2008, 8, 459–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Minokoshi, Y.; Alquier, T.; Furukawa, N.; Kim, Y.B.; Lee, A.; Xue, B.; Mu, J.; Foufelle, F.; Ferre, P.; Birnbaum, M.J.; et al. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 2004, 428, 569–574. [Google Scholar] [CrossRef] [PubMed]
  176. Dagon, Y.; Hur, E.; Zheng, B.; Wellenstein, K.; Cantley, L.C.; Kahn, B.B. p70S6 kinase phosphorylates AMPK on serine 491 to mediate leptin’s effect on food intake. Cell Metab. 2012, 16, 104–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Lopez, M. Hypothalamic AMPK and energy balance. Eur. J. Clin. Investig. 2018, 48, e12996. [Google Scholar] [CrossRef] [Green Version]
  178. Su, H.; Jiang, L.; Carter-Su, C.; Rui, L. Glucose enhances leptin signaling through modulation of AMPK activity. PLoS ONE 2012, 7, e31636. [Google Scholar] [CrossRef]
  179. Thurlby, P.L.; Trayhurn, P. The role of thermoregulatory thermogenesis in the development of obesity in genetically-obese (ob/ob) mice pair-fed with lean siblings. Br. J. Nutr. 1979, 42, 377–385. [Google Scholar] [CrossRef] [Green Version]
  180. Breslow, M.J.; Min-Lee, K.; Brown, D.R.; Chacko, V.P.; Palmer, D.; Berkowitz, D.E. Effect of leptin deficiency on metabolic rate in ob/ob mice. Am. J. Physiol. 1999, 276, E443–E449. [Google Scholar] [CrossRef]
  181. Breslow, M.J.; An, Y.; Berkowitz, D.E. Beta-3 adrenoceptor (beta-3AR) expression in leptin treated OB/OB mice. Life Sci. 1997, 61, 59–64. [Google Scholar] [CrossRef]
  182. Collins, S.; Daniel, K.W.; Rohlfs, E.M.; Ramkumar, V.; Taylor, I.L.; Gettys, T.W. Impaired expression and functional activity of the beta 3- and beta 1-adrenergic receptors in adipose tissue of congenitally obese (C57BL/6J ob/ob) mice. Mol. Endocrinol. 1994, 8, 518–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Trayhurn, P.; Thurlby, P.L.; James, W.P. Thermogenic defect in pre-obese ob/ob mice. Nature 1977, 266, 60–62. [Google Scholar] [CrossRef] [PubMed]
  184. Harris, R.B.; Zhou, J.; Redmann, S.M., Jr.; Smagin, G.N.; Smith, S.R.; Rodgers, E.; Zachwieja, J.J. A leptin dose-response study in obese (ob/ob) and lean (+/?) mice. Endocrinology 1998, 139, 8–19. [Google Scholar] [CrossRef]
  185. Contreras, C.; Nogueiras, R.; Dieguez, C.; Rahmouni, K.; Lopez, M. Traveling from the hypothalamus to the adipose tissue: The thermogenic pathway. Redox Biol. 2017, 12, 854–863. [Google Scholar] [CrossRef] [PubMed]
  186. Martinez-Sanchez, N.; Moreno-Navarrete, J.M.; Contreras, C.; Rial-Pensado, E.; Ferno, J.; Nogueiras, R.; Dieguez, C.; Fernandez-Real, J.M.; Lopez, M. Thyroid hormones induce browning of white fat. J. Endocrinol. 2017, 232, 351–362. [Google Scholar] [CrossRef] [Green Version]
  187. Kaisanlahti, A.; Glumoff, T. Browning of white fat: Agents and implications for beige adipose tissue to type 2 diabetes. J. Physiol. Biochem. 2019, 75, 1–10. [Google Scholar] [CrossRef] [Green Version]
  188. Haynes, W.G.; Morgan, D.A.; Walsh, S.A.; Mark, A.L.; Sivitz, W.I. Receptor-mediated regional sympathetic nerve activation by leptin. J. Clin. Investig. 1997, 100, 270–278. [Google Scholar] [CrossRef]
  189. Dunbar, J.C.; Hu, Y.; Lu, H. Intracerebroventricular leptin increases lumbar and renal sympathetic nerve activity and blood pressure in normal rats. Diabetes 1997, 46, 2040–2043. [Google Scholar] [CrossRef]
  190. Richards, R.J.; Blalock, A.; Liao, J.; Reisin, E. Leptin: Sympathetic and cardiovascular effects. Curr. Cardiol. Rep. 2003, 5, 453–458. [Google Scholar] [CrossRef]
  191. Commins, S.P.; Watson, P.M.; Levin, N.; Beiler, R.J.; Gettys, T.W. Central leptin regulates the UCP1 and ob genes in brown and white adipose tissue via different beta-adrenoceptor subtypes. J. Biol. Chem. 2000, 275, 33059–33067. [Google Scholar] [CrossRef] [Green Version]
  192. Enriori, P.J.; Sinnayah, P.; Simonds, S.E.; Garcia Rudaz, C.; Cowley, M.A. Leptin action in the dorsomedial hypothalamus increases sympathetic tone to brown adipose tissue in spite of systemic leptin resistance. J. Neurosci. 2011, 31, 12189–12197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Fischer, A.W.; Hoefig, C.S.; Abreu-Vieira, G.; de Jong, J.M.A.; Petrovic, N.; Mittag, J.; Cannon, B.; Nedergaard, J. Leptin Raises Defended Body Temperature without Activating Thermogenesis. Cell Rep. 2016, 14, 1621–1631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Doring, H.; Schwarzer, K.; Nuesslein-Hildesheim, B.; Schmidt, I. Leptin selectively increases energy expenditure of food-restricted lean mice. Int. J. Obes. 1998, 22, 83–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Mistry, A.M.; Swick, A.G.; Romsos, D.R. Leptin rapidly lowers food intake and elevates metabolic rates in lean and ob/ob mice. J. Nutr. 1997, 127, 2065–2072. [Google Scholar] [CrossRef] [PubMed]
  196. Rafael, J.; Herling, A.W. Leptin effect in ob/ob mice under thermoneutral conditions depends not necessarily on central satiation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000, 278, R790–R795. [Google Scholar] [CrossRef] [Green Version]
  197. Hogberg, H.; Engblom, L.; Ekdahl, A.; Lidell, V.; Walum, E.; Alberts, P. Temperature dependence of O2 consumption; opposite effects of leptin and etomoxir on respiratory quotient in mice. Obesity 2006, 14, 673–682. [Google Scholar] [CrossRef]
  198. Ukropec, J.; Anunciado, R.V.; Ravussin, Y.; Kozak, L.P. Leptin is required for uncoupling protein-1-independent thermogenesis during cold stress. Endocrinology 2006, 147, 2468–2480. [Google Scholar] [CrossRef] [Green Version]
  199. Kaiyala, K.J.; Ogimoto, K.; Nelson, J.T.; Muta, K.; Morton, G.J. Physiological role for leptin in the control of thermal conductance. Mol. Metab. 2016, 5, 892–902. [Google Scholar] [CrossRef]
  200. Dimicco, J.A.; Zaretsky, D.V. The dorsomedial hypothalamus: A new player in thermoregulation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 292, R47–R63. [Google Scholar] [CrossRef]
  201. Morrison, S.F.; Madden, C.J.; Tupone, D. Central control of brown adipose tissue thermogenesis. Front. Endocrinol. 2012, 3, 5. [Google Scholar] [CrossRef] [Green Version]
  202. Morrison, S.F.; Nakamura, K.; Madden, C.J. Central control of thermogenesis in mammals. Exp. Physiol. 2008, 93, 773–797. [Google Scholar] [CrossRef] [PubMed]
  203. Gautron, L.; Lazarus, M.; Scott, M.M.; Saper, C.B.; Elmquist, J.K. Identifying the efferent projections of leptin-responsive neurons in the dorsomedial hypothalamus using a novel conditional tracing approach. J. Comp. Neurol. 2010, 518, 2090–2108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Rezai-Zadeh, K.; Yu, S.; Jiang, Y.; Laque, A.; Schwartzenburg, C.; Morrison, C.D.; Derbenev, A.V.; Zsombok, A.; Munzberg, H. Leptin receptor neurons in the dorsomedial hypothalamus are key regulators of energy expenditure and body weight, but not food intake. Mol. Metab. 2014, 3, 681–693. [Google Scholar] [CrossRef] [PubMed]
  205. Cakir, I.; Diaz Martinez, M.; Lining Pan, P.; Welch, E.B.; Patel, S.; Ghamari-Langroudi, M. Leptin Receptor Signaling in Sim1-expressing Neurons Regulates Body Temperature and Adaptive Thermogenesis. Endocrinology 2019, 160, 863–879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Munzberg, H.; Flier, J.S.; Bjorbaek, C. Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology 2004, 145, 4880–4889. [Google Scholar] [CrossRef] [Green Version]
  207. Dodd, G.T.; Decherf, S.; Loh, K.; Simonds, S.E.; Wiede, F.; Balland, E.; Merry, T.L.; Munzberg, H.; Zhang, Z.Y.; Kahn, B.B.; et al. Leptin and insulin act on POMC neurons to promote the browning of white fat. Cell 2015, 160, 88–104. [Google Scholar] [CrossRef] [Green Version]
  208. Elmquist, J.K.; Ahima, R.S.; Elias, C.F.; Flier, J.S.; Saper, C.B. Leptin activates distinct projections from the dorsomedial and ventromedial hypothalamic nuclei. Proc. Natl. Acad. Sci. USA 1998, 95, 741–746. [Google Scholar] [CrossRef] [Green Version]
  209. Cao, W.H.; Morrison, S.F. Disinhibition of rostral raphe pallidus neurons increases cardiac sympathetic nerve activity and heart rate. Brain Res. 2003, 980, 1–10. [Google Scholar] [CrossRef]
  210. Simonds, S.E.; Cowley, M.A. Hypertension in obesity: Is leptin the culprit? Trends Neurosci. 2013, 36, 121–132. [Google Scholar] [CrossRef]
  211. Zhang, Y.; Kerman, I.A.; Laque, A.; Nguyen, P.; Faouzi, M.; Louis, G.W.; Jones, J.C.; Rhodes, C.; Munzberg, H. Leptin-receptor-expressing neurons in the dorsomedial hypothalamus and median preoptic area regulate sympathetic brown adipose tissue circuits. J. Neurosci. 2011, 31, 1873–1884. [Google Scholar] [CrossRef] [Green Version]
  212. Nakamura, K.; Morrison, S.F. Preoptic mechanism for cold-defensive responses to skin cooling. J. Physiol. 2008, 586, 2611–2620. [Google Scholar] [CrossRef] [PubMed]
  213. Nakamura, K. Central circuitries for body temperature regulation and fever. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 301, R1207–R1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Yoshida, K.; Li, X.; Cano, G.; Lazarus, M.; Saper, C.B. Parallel preoptic pathways for thermoregulation. J. Neurosci. 2009, 29, 11954–11964. [Google Scholar] [CrossRef] [PubMed]
  215. Nakamura, K.; Matsumura, K.; Kobayashi, S.; Kaneko, T. Sympathetic premotor neurons mediating thermoregulatory functions. Neurosci. Res. 2005, 51, 1–8. [Google Scholar] [CrossRef] [PubMed]
  216. Zhang, Z.; Liu, X.; Morgan, D.A.; Kuburas, A.; Thedens, D.R.; Russo, A.F.; Rahmouni, K. Neuronal receptor activity-modifying protein 1 promotes energy expenditure in mice. Diabetes 2011, 60, 1063–1071. [Google Scholar] [CrossRef] [Green Version]
  217. Song, C.K.; Vaughan, C.H.; Keen-Rhinehart, E.; Harris, R.B.; Richard, D.; Bartness, T.J. Melanocortin-4 receptor mRNA expressed in sympathetic outflow neurons to brown adipose tissue: Neuroanatomical and functional evidence. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 295, R417–R428. [Google Scholar] [CrossRef] [Green Version]
  218. Morrison, S.F.; Madden, C.J.; Tupone, D. An orexinergic projection from perifornical hypothalamus to raphe pallidus increases rat brown adipose tissue thermogenesis. Adipocyte 2012, 1, 116–120. [Google Scholar] [CrossRef] [Green Version]
  219. Morrison, S.F.; Madden, C.J.; Tupone, D. Central neural regulation of brown adipose tissue thermogenesis and energy expenditure. Cell Metab. 2014, 19, 741–756. [Google Scholar] [CrossRef] [Green Version]
  220. Madden, C.J.; Morrison, S.F. Neurons in the paraventricular nucleus of the hypothalamus inhibit sympathetic outflow to brown adipose tissue. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2009, 296, R831–R843. [Google Scholar] [CrossRef]
  221. Pandit, R.; Beerens, S.; Adan, R.A.H. Role of leptin in energy expenditure: The hypothalamic perspective. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2017, 312, R938–R947. [Google Scholar] [CrossRef] [Green Version]
  222. Mogenson, G.J.; Swanson, L.W.; Wu, M. Neural projections from nucleus accumbens to globus pallidus, substantia innominata, and lateral preoptic-lateral hypothalamic area: An anatomical and electrophysiological investigation in the rat. J. Neurosci. 1983, 3, 189–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. ter Horst, G.J.; Luiten, P.G. The projections of the dorsomedial hypothalamic nucleus in the rat. Brain Res. Bull 1986, 16, 231–248. [Google Scholar] [CrossRef] [Green Version]
  224. Thompson, R.H.; Canteras, N.S.; Swanson, L.W. Organization of projections from the dorsomedial nucleus of the hypothalamus: A PHA-L study in the rat. J. Comp. Neurol. 1996, 376, 143–173. [Google Scholar] [CrossRef]
  225. Satoh, N.; Ogawa, Y.; Katsuura, G.; Numata, Y.; Tsuji, T.; Hayase, M.; Ebihara, K.; Masuzaki, H.; Hosoda, K.; Yoshimasa, Y.; et al. Sympathetic activation of leptin via the ventromedial hypothalamus: Leptin-induced increase in catecholamine secretion. Diabetes 1999, 48, 1787–1793. [Google Scholar] [CrossRef] [PubMed]
  226. Minokoshi, Y.; Haque, M.S.; Shimazu, T. Microinjection of leptin into the ventromedial hypothalamus increases glucose uptake in peripheral tissues in rats. Diabetes 1999, 48, 287–291. [Google Scholar] [CrossRef] [PubMed]
  227. Marsh, A.J.; Fontes, M.A.; Killinger, S.; Pawlak, D.B.; Polson, J.W.; Dampney, R.A. Cardiovascular responses evoked by leptin acting on neurons in the ventromedial and dorsomedial hypothalamus. Hypertension 2003, 42, 488–493. [Google Scholar] [CrossRef] [Green Version]
  228. Montanaro, M.S.; Allen, A.M.; Oldfield, B.J. Structural and functional evidence supporting a role for leptin in central neural pathways influencing blood pressure in rats. Exp. Physiol. 2005, 90, 689–696. [Google Scholar] [CrossRef]
  229. Ikeda, Y.; Luo, X.; Abbud, R.; Nilson, J.H.; Parker, K.L. The nuclear receptor steroidogenic factor 1 is essential for the formation of the ventromedial hypothalamic nucleus. Mol. Endocrinol. 1995, 9, 478–486. [Google Scholar] [CrossRef] [Green Version]
  230. Elias, C.F.; Kelly, J.F.; Lee, C.E.; Ahima, R.S.; Drucker, D.J.; Saper, C.B.; Elmquist, J.K. Chemical characterization of leptin-activated neurons in the rat brain. J. Comp. Neurol. 2000, 423, 261–281. [Google Scholar] [CrossRef]
  231. Dhillon, H.; Zigman, J.M.; Ye, C.; Lee, C.E.; McGovern, R.A.; Tang, V.; Kenny, C.D.; Christiansen, L.M.; White, R.D.; Edelstein, E.A.; et al. Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron 2006, 49, 191–203. [Google Scholar] [CrossRef] [Green Version]
  232. Kim, K.W.; Zhao, L.; Donato, J., Jr.; Kohno, D.; Xu, Y.; Elias, C.F.; Lee, C.; Parker, K.L.; Elmquist, J.K. Steroidogenic factor 1 directs programs regulating diet-induced thermogenesis and leptin action in the ventral medial hypothalamic nucleus. Proc. Natl. Acad. Sci. USA 2011, 108, 10673–10678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Ovesjo, M.L.; Gamstedt, M.; Collin, M.; Meister, B. GABAergic nature of hypothalamic leptin target neurones in the ventromedial arcuate nucleus. J. Neuroendocrinol. 2001, 13, 505–516. [Google Scholar] [CrossRef] [PubMed]
  234. Tong, Q.; Ye, C.; McCrimmon, R.J.; Dhillon, H.; Choi, B.; Kramer, M.D.; Yu, J.; Yang, Z.; Christiansen, L.M.; Lee, C.E.; et al. Synaptic glutamate release by ventromedial hypothalamic neurons is part of the neurocircuitry that prevents hypoglycemia. Cell Metab. 2007, 5, 383–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Lindberg, D.; Chen, P.; Li, C. Conditional viral tracing reveals that steroidogenic factor 1-positive neurons of the dorsomedial subdivision of the ventromedial hypothalamus project to autonomic centers of the hypothalamus and hindbrain. J. Comp. Neurol. 2013, 521, 3167–3190. [Google Scholar] [CrossRef] [PubMed]
  236. Kenny, D.A.; Jurata, L.W.; Saga, Y.; Gill, G.N. Identification and characterization of LMO4, an LMO gene with a novel pattern of expression during embryogenesis. Proc. Natl. Acad. Sci. USA 1998, 95, 11257–11262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Zhou, X.; Gomez-Smith, M.; Qin, Z.; Duquette, P.M.; Cardenas-Blanco, A.; Rai, P.S.; Harper, M.E.; Tsai, E.C.; Anisman, H.; Chen, H.H. Ablation of LMO4 in glutamatergic neurons impairs leptin control of fat metabolism. Cell. Mol. Life Sci. 2012, 69, 819–828. [Google Scholar] [CrossRef] [Green Version]
  238. Cardinal, P.; Andre, C.; Quarta, C.; Bellocchio, L.; Clark, S.; Elie, M.; Leste-Lasserre, T.; Maitre, M.; Gonzales, D.; Cannich, A.; et al. CB1 cannabinoid receptor in SF1-expressing neurons of the ventromedial hypothalamus determines metabolic responses to diet and leptin. Mol. Metab. 2014, 3, 705–716. [Google Scholar] [CrossRef]
  239. Kim, K.W.; Donato, J., Jr.; Berglund, E.D.; Choi, Y.H.; Kohno, D.; Elias, C.F.; Depinho, R.A.; Elmquist, J.K. FOXO1 in the ventromedial hypothalamus regulates energy balance. J. Clin. Investig. 2012, 122, 2578–2589. [Google Scholar] [CrossRef]
  240. Seamon, M.; Ahn, W.; Li, A.J.; Ritter, S.; Harris, R.B.S. Leptin receptor-expressing neurons in ventromedial nucleus of the hypothalamus contribute to weight loss caused by fourth ventricle leptin infusions. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E586–E596. [Google Scholar] [CrossRef]
  241. Rahmouni, K.; Morgan, D.A.; Morgan, G.M.; Liu, X.; Sigmund, C.D.; Mark, A.L.; Haynes, W.G. Hypothalamic PI3K and MAPK differentially mediate regional sympathetic activation to insulin. J. Clin. Investig. 2004, 114, 652–658. [Google Scholar] [CrossRef] [Green Version]
  242. Rahmouni, K.; Morgan, D.A. Hypothalamic arcuate nucleus mediates the sympathetic and arterial pressure responses to leptin. Hypertension 2007, 49, 647–652. [Google Scholar] [CrossRef] [Green Version]
  243. Harlan, S.M.; Morgan, D.A.; Agassandian, K.; Guo, D.F.; Cassell, M.D.; Sigmund, C.D.; Mark, A.L.; Rahmouni, K. Ablation of the leptin receptor in the hypothalamic arcuate nucleus abrogates leptin-induced sympathetic activation. Circ. Res. 2011, 108, 808–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Cone, R.D. The Central Melanocortin System and Energy Homeostasis. Trends Endocrinol. Metab. 1999, 10, 211–216. [Google Scholar] [CrossRef]
  245. Shi, Y.C.; Lau, J.; Lin, Z.; Zhang, H.; Zhai, L.; Sperk, G.; Heilbronn, R.; Mietzsch, M.; Weger, S.; Huang, X.F.; et al. Arcuate NPY controls sympathetic output and BAT function via a relay of tyrosine hydroxylase neurons in the PVN. Cell Metab. 2013, 17, 236–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Erickson, J.C.; Hollopeter, G.; Palmiter, R.D. Attenuation of the obesity syndrome of ob/ob mice by the loss of neuropeptide Y. Science 1996, 274, 1704–1707. [Google Scholar] [CrossRef]
  247. Billington, C.J.; Briggs, J.E.; Grace, M.; Levine, A.S. Effects of intracerebroventricular injection of neuropeptide Y on energy metabolism. Am. J. Physiol. 1991, 260, R321–R327. [Google Scholar] [CrossRef] [PubMed]
  248. Lopez-Valpuesta, F.J.; Nyce, J.W.; Griffin-Biggs, T.A.; Ice, J.C.; Myers, R.D. Antisense to NPY-Y1 demonstrates that Y1 receptors in the hypothalamus underlie NPY hypothermia and feeding in rats. Proc. Biol. Sci. 1996, 263, 881–886. [Google Scholar] [CrossRef]
  249. Stanley, B.G.; Kyrkouli, S.E.; Lampert, S.; Leibowitz, S.F. Neuropeptide Y chronically injected into the hypothalamus: A powerful neurochemical inducer of hyperphagia and obesity. Peptides 1986, 7, 1189–1192. [Google Scholar] [CrossRef]
  250. Cusin, I.; Rohner-Jeanrenaud, F.; Stricker-Krongrad, A.; Jeanrenaud, B. The weight-reducing effect of an intracerebroventricular bolus injection of leptin in genetically obese fa/fa rats. Reduced sensitivity compared with lean animals. Diabetes 1996, 45, 1446–1450. [Google Scholar] [CrossRef] [Green Version]
  251. Sahu, A. Leptin decreases food intake induced by melanin-concentrating hormone (MCH), galanin (GAL) and neuropeptide Y (NPY) in the rat. Endocrinology 1998, 139, 4739–4742. [Google Scholar] [CrossRef]
  252. Jang, M.; Mistry, A.; Swick, A.G.; Romsos, D.R. Leptin rapidly inhibits hypothalamic neuropeptide Y secretion and stimulates corticotropin-releasing hormone secretion in adrenalectomized mice. J. Nutr. 2000, 130, 2813–2820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Wang, Q.; Bing, C.; Al-Barazanji, K.; Mossakowaska, D.E.; Wang, X.M.; McBay, D.L.; Neville, W.A.; Taddayon, M.; Pickavance, L.; Dryden, S.; et al. Interactions between leptin and hypothalamic neuropeptide Y neurons in the control of food intake and energy homeostasis in the rat. Diabetes 1997, 46, 335–341. [Google Scholar] [CrossRef] [PubMed]
  254. Lopez-Valpuesta, F.J.; Nyce, J.W.; Myers, R.D. NPY-Y1 receptor antisense injected centrally in rats causes hyperthermia and feeding. Neuroreport 1996, 7, 2781–2784. [Google Scholar] [CrossRef] [PubMed]
  255. Mizuno, T.M.; Kleopoulos, S.P.; Bergen, H.T.; Roberts, J.L.; Priest, C.A.; Mobbs, C.V. Hypothalamic pro-opiomelanocortin mRNA is reduced by fasting in ob/ob and db/db mice, but is stimulated by leptin. Diabetes 1998, 47, 294–297. [Google Scholar] [CrossRef] [Green Version]
  256. do Carmo, J.M.; da Silva, A.A.; Cai, Z.; Lin, S.; Dubinion, J.H.; Hall, J.E. Control of blood pressure, appetite, and glucose by leptin in mice lacking leptin receptors in proopiomelanocortin neurons. Hypertension 2011, 57, 918–926. [Google Scholar] [CrossRef]
  257. Huo, L.; Grill, H.J.; Bjorbaek, C. Divergent regulation of proopiomelanocortin neurons by leptin in the nucleus of the solitary tract and in the arcuate hypothalamic nucleus. Diabetes 2006, 55, 567–573. [Google Scholar] [CrossRef] [Green Version]
  258. Kishi, T.; Aschkenasi, C.J.; Lee, C.E.; Mountjoy, K.G.; Saper, C.B.; Elmquist, J.K. Expression of melanocortin 4 receptor mRNA in the central nervous system of the rat. J. Comp. Neurol. 2003, 457, 213–235. [Google Scholar] [CrossRef]
  259. Begriche, K.; Sutton, G.M.; Butler, A.A. Homeostastic and non-homeostatic functions of melanocortin-3 receptors in the control of energy balance and metabolism. Physiol. Behav. 2011, 104, 546–554. [Google Scholar] [CrossRef] [Green Version]
  260. Seeley, R.J.; Yagaloff, K.A.; Fisher, S.L.; Burn, P.; Thiele, T.E.; van Dijk, G.; Baskin, D.G.; Schwartz, M.W. Melanocortin receptors in leptin effects. Nature 1997, 390, 349. [Google Scholar] [CrossRef] [Green Version]
  261. Haynes, W.G.; Morgan, D.A.; Djalali, A.; Sivitz, W.I.; Mark, A.L. Interactions between the melanocortin system and leptin in control of sympathetic nerve traffic. Hypertension 1999, 33, 542–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Williams, D.L.; Bowers, R.R.; Bartness, T.J.; Kaplan, J.M.; Grill, H.J. Brainstem melanocortin 3/4 receptor stimulation increases uncoupling protein gene expression in brown fat. Endocrinology 2003, 144, 4692–4697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Ste Marie, L.; Miura, G.I.; Marsh, D.J.; Yagaloff, K.; Palmiter, R.D. A metabolic defect promotes obesity in mice lacking melanocortin-4 receptors. Proc. Natl. Acad. Sci. USA 2000, 97, 12339–12344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Rahmouni, K.; Haynes, W.G.; Morgan, D.A.; Mark, A.L. Role of melanocortin-4 receptors in mediating renal sympathoactivation to leptin and insulin. J. Neurosci. 2003, 23, 5998–6004. [Google Scholar] [CrossRef] [PubMed]
  265. Voss-Andreae, A.; Murphy, J.G.; Ellacott, K.L.; Stuart, R.C.; Nillni, E.A.; Cone, R.D.; Fan, W. Role of the central melanocortin circuitry in adaptive thermogenesis of brown adipose tissue. Endocrinology 2007, 148, 1550–1560. [Google Scholar] [CrossRef]
  266. Lute, B.; Jou, W.; Lateef, D.M.; Goldgof, M.; Xiao, C.; Pinol, R.A.; Kravitz, A.V.; Miller, N.R.; Huang, Y.G.; Girardet, C.; et al. Biphasic effect of melanocortin agonists on metabolic rate and body temperature. Cell Metab. 2014, 20, 333–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Baran, K.; Preston, E.; Wilks, D.; Cooney, G.J.; Kraegen, E.W.; Sainsbury, A. Chronic central melanocortin-4 receptor antagonism and central neuropeptide-Y infusion in rats produce increased adiposity by divergent pathways. Diabetes 2002, 51, 152–158. [Google Scholar] [CrossRef] [Green Version]
  268. Geerling, J.C.; Loewy, A.D. Aldosterone-sensitive neurons in the nucleus of the solitary tract: Bidirectional connections with the central nucleus of the amygdala. J. Comp. Neurol. 2006, 497, 646–657. [Google Scholar] [CrossRef] [Green Version]
  269. Grill, H.J.; Schwartz, M.W.; Kaplan, J.M.; Foxhall, J.S.; Breininger, J.; Baskin, D.G. Evidence that the caudal brainstem is a target for the inhibitory effect of leptin on food intake. Endocrinology 2002, 143, 239–246. [Google Scholar] [CrossRef]
  270. Hermann, G.E.; Barnes, M.J.; Rogers, R.C. Leptin and thyrotropin-releasing hormone: Cooperative action in the hindbrain to activate brown adipose thermogenesis. Brain Res. 2006, 1117, 118–124. [Google Scholar] [CrossRef]
  271. Mark, A.L.; Agassandian, K.; Morgan, D.A.; Liu, X.; Cassell, M.D.; Rahmouni, K. Leptin signaling in the nucleus tractus solitarii increases sympathetic nerve activity to the kidney. Hypertension 2009, 53, 375–380. [Google Scholar] [CrossRef] [Green Version]
  272. Rogers, R.C.; Barnes, M.J.; Hermann, G.E. Leptin “gates” thermogenic action of thyrotropin-releasing hormone in the hindbrain. Brain Res. 2009, 1295, 135–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Rogers, R.C.; McDougal, D.H.; Hermann, G.E. Leptin amplifies the action of thyrotropin-releasing hormone in the solitary nucleus: An in vitro calcium imaging study. Brain Res. 2011, 1385, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Li, H.; Matheny, M.; Scarpace, P.J. beta 3-Adrenergic-mediated suppression of leptin gene expression in rats. Am. J. Physiol. 1997, 272, E1031–E1036. [Google Scholar] [CrossRef] [PubMed]
  275. Trayhurn, P.; Duncan, J.S.; Rayner, D.V. Acute cold-induced suppression of ob (obese) gene expression in white adipose tissue of mice: Mediation by the sympathetic system. Biochem. J. 1995, 311, 729–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Zeng, W.; Pirzgalska, R.M.; Pereira, M.M.; Kubasova, N.; Barateiro, A.; Seixas, E.; Lu, Y.H.; Kozlova, A.; Voss, H.; Martins, G.G.; et al. Sympathetic neuro-adipose connections mediate leptin-driven lipolysis. Cell 2015, 163, 84–94. [Google Scholar] [CrossRef] [Green Version]
  277. Plum, L.; Rother, E.; Munzberg, H.; Wunderlich, F.T.; Morgan, D.A.; Hampel, B.; Shanabrough, M.; Janoschek, R.; Konner, A.C.; Alber, J.; et al. Enhanced leptin-stimulated Pi3k activation in the CNS promotes white adipose tissue transdifferentiation. Cell Metab. 2007, 6, 431–445. [Google Scholar] [CrossRef] [Green Version]
  278. Dodd, G.T.; Andrews, Z.B.; Simonds, S.E.; Michael, N.J.; DeVeer, M.; Bruning, J.C.; Spanswick, D.; Cowley, M.A.; Tiganis, T. A Hypothalamic Phosphatase Switch Coordinates Energy Expenditure with Feeding. Cell Metab. 2017, 26, 577. [Google Scholar] [CrossRef]
  279. Ruan, H.B.; Dietrich, M.O.; Liu, Z.W.; Zimmer, M.R.; Li, M.D.; Singh, J.P.; Zhang, K.; Yin, R.; Wu, J.; Horvath, T.L.; et al. O-GlcNAc transferase enables AgRP neurons to suppress browning of white fat. Cell 2014, 159, 306–317. [Google Scholar] [CrossRef] [Green Version]
  280. Berglund, E.D.; Liu, T.; Kong, X.; Sohn, J.W.; Vong, L.; Deng, Z.; Lee, C.E.; Lee, S.; Williams, K.W.; Olson, D.P.; et al. Melanocortin 4 receptors in autonomic neurons regulate thermogenesis and glycemia. Nat. Neurosci. 2014, 17, 911–913. [Google Scholar] [CrossRef] [Green Version]
  281. Lidell, M.E.; Seifert, E.L.; Westergren, R.; Heglind, M.; Gowing, A.; Sukonina, V.; Arani, Z.; Itkonen, P.; Wallin, S.; Westberg, F.; et al. The adipocyte-expressed forkhead transcription factor Foxc2 regulates metabolism through altered mitochondrial function. Diabetes 2011, 60, 427–435. [Google Scholar] [CrossRef] [Green Version]
  282. Cederberg, A.; Gronning, L.M.; Ahren, B.; Tasken, K.; Carlsson, P.; Enerback, S. FOXC2 is a winged helix gene that counteracts obesity, hypertriglyceridemia, and diet-induced insulin resistance. Cell 2001, 106, 563–573. [Google Scholar] [CrossRef] [Green Version]
  283. Rahman, S.; Lu, Y.; Czernik, P.J.; Rosen, C.J.; Enerback, S.; Lecka-Czernik, B. Inducible brown adipose tissue, or beige fat, is anabolic for the skeleton. Endocrinology 2013, 154, 2687–2701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Gan, L.; Liu, Z.; Feng, F.; Wu, T.; Luo, D.; Hu, C.; Sun, C. Foxc2 coordinates inflammation and browning of white adipose by leptin-STAT3-PRDM16 signal in mice. Int. J. Obes. 2018, 42, 252–259. [Google Scholar] [CrossRef] [PubMed]
  285. Wang, J.; Ge, J.; Cao, H.; Zhang, X.; Guo, Y.; Li, X.; Xia, B.; Yang, G.; Shi, X. Leptin Promotes White Adipocyte Browning by Inhibiting the Hh Signaling Pathway. Cells 2019, 8. [Google Scholar] [CrossRef] [Green Version]
  286. Tartaglia, L.A. The leptin receptor. J. Biol. Chem. 1997, 272, 6093–6096. [Google Scholar] [CrossRef] [Green Version]
  287. Zhang, Y.; Kilroy, G.E.; Henagan, T.M.; Prpic-Uhing, V.; Richards, W.G.; Bannon, A.W.; Mynatt, R.L.; Gettys, T.W. Targeted deletion of melanocortin receptor subtypes 3 and 4, but not CART, alters nutrient partitioning and compromises behavioral and metabolic responses to leptin. FASEB J. 2005, 19, 1482–1491. [Google Scholar] [CrossRef]
  288. Siegrist-Kaiser, C.A.; Pauli, V.; Juge-Aubry, C.E.; Boss, O.; Pernin, A.; Chin, W.W.; Cusin, I.; Rohner-Jeanrenaud, F.; Burger, A.G.; Zapf, J.; et al. Direct effects of leptin on brown and white adipose tissue. J. Clin. Investig. 1997, 100, 2858–2864. [Google Scholar] [CrossRef] [Green Version]
  289. Simonds, S.E.; Pryor, J.T.; Ravussin, E.; Greenway, F.L.; Dileone, R.; Allen, A.M.; Bassi, J.; Elmquist, J.K.; Keogh, J.M.; Henning, E.; et al. Leptin mediates the increase in blood pressure associated with obesity. Cell 2014, 159, 1404–1416. [Google Scholar] [CrossRef] [Green Version]
  290. Dulloo, A.G.; Stock, M.J.; Solinas, G.; Boss, O.; Montani, J.P.; Seydoux, J. Leptin directly stimulates thermogenesis in skeletal muscle. FEBS Lett. 2002, 515, 109–113. [Google Scholar] [CrossRef] [Green Version]
  291. Paracchini, V.; Pedotti, P.; Taioli, E. Genetics of leptin and obesity: A HuGE review. Am. J. Epidemiol. 2005, 162, 101–114. [Google Scholar] [CrossRef]
  292. Mizuta, E.; Kokubo, Y.; Yamanaka, I.; Miyamoto, Y.; Okayama, A.; Yoshimasa, Y.; Tomoike, H.; Morisaki, H.; Morisaki, T. Leptin gene and leptin receptor gene polymorphisms are associated with sweet preference and obesity. Hypertens. Res. 2008, 31, 1069–1077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Costa, V.; Casamassimi, A.; Ciccodicola, A. Nutritional genomics era: Opportunities toward a genome-tailored nutritional regimen. J. Nutr. Biochem. 2010, 21, 457–467. [Google Scholar] [CrossRef] [PubMed]
  294. Ghalandari, H.; Hosseini-Esfahani, F.; Mirmiran, P. The Association of Polymorphisms in Leptin/Leptin Receptor Genes and Ghrelin/Ghrelin Receptor Genes With Overweight/Obesity and the Related Metabolic Disturbances: A Review. Int. J. Endocrinol. Metab. 2015, 13, e19073. [Google Scholar] [CrossRef] [Green Version]
  295. Murugesan, D.; Arunachalam, T.; Ramamurthy, V.; Subramanian, S. Association of polymorphisms in leptin receptor gene with obesity and type 2 diabetes in the local population of Coimbatore. Indian J. Hum. Genet. 2010, 16, 72–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Furusawa, T.; Naka, I.; Yamauchi, T.; Natsuhara, K.; Kimura, R.; Nakazawa, M.; Ishida, T.; Inaoka, T.; Matsumura, Y.; Ataka, Y.; et al. The Q223R polymorphism in LEPR is associated with obesity in Pacific Islanders. Hum. Genet. 2010, 127, 287–294. [Google Scholar] [CrossRef] [PubMed]
  297. Tabassum, R.; Mahendran, Y.; Dwivedi, O.P.; Chauhan, G.; Ghosh, S.; Marwaha, R.K.; Tandon, N.; Bharadwaj, D. Common variants of IL6, LEPR, and PBEF1 are associated with obesity in Indian children. Diabetes 2012, 61, 626–631. [Google Scholar] [CrossRef] [Green Version]
  298. Boumaiza, I.; Omezzine, A.; Rejeb, J.; Rebhi, L.; Ben Rejeb, N.; Nabli, N.; Ben Abdelaziz, A.; Bouslama, A. Association between four resistin polymorphisms, obesity, and metabolic syndrome parameters in Tunisian volunteers. Genet. Test Mol. Biomark. 2012, 16, 1356–1362. [Google Scholar] [CrossRef]
  299. Etemad, A.; Ramachandran, V.; Pishva, S.R.; Heidari, F.; Aziz, A.F.; Yusof, A.K.; Pei, C.P.; Ismail, P. Analysis of Gln223Agr polymorphism of Leptin Receptor Gene in type II diabetic mellitus subjects among Malaysians. Int. J. Mol. Sci. 2013, 14, 19230–19244. [Google Scholar] [CrossRef] [Green Version]
  300. Lu, J.; Zou, D.; Zheng, L.; Chen, G.; Lu, J.; Feng, Z. Synergistic effect of LEP and LEPR gene polymorphism on body mass index in a Chinese population. Obes. Res. Clin. Pract. 2013, 7, e445–e449. [Google Scholar] [CrossRef]
  301. Heo, M.; Leibel, R.L.; Fontaine, K.R.; Boyer, B.B.; Chung, W.K.; Koulu, M.; Karvonen, M.K.; Pesonen, U.; Rissanen, A.; Laakso, M.; et al. A meta-analytic investigation of linkage and association of common leptin receptor (LEPR) polymorphisms with body mass index and waist circumference. Int. J. Obes. 2002, 26, 640–646. [Google Scholar] [CrossRef] [Green Version]
  302. Komsu-Ornek, Z.; Demirel, F.; Dursun, A.; Ermis, B.; Piskin, E.; Bideci, A. Leptin receptor gene Gln223Arg polymorphism is not associated with obesity and metabolic syndrome in Turkish children. Turk. J. Pediatr. 2012, 54, 20–24. [Google Scholar] [PubMed]
  303. Pyrzak, B.; Wisniewska, A.; Kucharska, A.; Wasik, M.; Demkow, U. No association of LEPR Gln223Arg polymorphism with leptin, obesity or metabolic disturbances in children. Eur. J. Med. Res. 2009, 14, 201–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Oral, E.A.; Simha, V.; Ruiz, E.; Andewelt, A.; Premkumar, A.; Snell, P.; Wagner, A.J.; DePaoli, A.M.; Reitman, M.L.; Taylor, S.I.; et al. Leptin-replacement therapy for lipodystrophy. N. Engl. J. Med. 2002, 346, 570–578. [Google Scholar] [CrossRef] [PubMed]
  305. Welt, C.K.; Chan, J.L.; Bullen, J.; Murphy, R.; Smith, P.; DePaoli, A.M.; Karalis, A.; Mantzoros, C.S. Recombinant human leptin in women with hypothalamic amenorrhea. N. Engl. J. Med. 2004, 351, 987–997. [Google Scholar] [CrossRef] [PubMed]
  306. Chou, S.H.; Chamberland, J.P.; Liu, X.; Matarese, G.; Gao, C.; Stefanakis, R.; Brinkoetter, M.T.; Gong, H.; Arampatzi, K.; Mantzoros, C.S. Leptin is an effective treatment for hypothalamic amenorrhea. Proc. Natl. Acad. Sci. USA 2011, 108, 6585–6590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Paz-Filho, G.; Mastronardi, C.A.; Licinio, J. Leptin treatment: Facts and expectations. Metabolism 2015, 64, 146–156. [Google Scholar] [CrossRef]
  308. Frederich, R.C.; Hamann, A.; Anderson, S.; Lollmann, B.; Lowell, B.B.; Flier, J.S. Leptin levels reflect body lipid content in mice: Evidence for diet-induced resistance to leptin action. Nat. Med. 1995, 1, 1311–1314. [Google Scholar] [CrossRef]
  309. Myers, M.G., Jr.; Leibel, R.L.; Seeley, R.J.; Schwartz, M.W. Obesity and leptin resistance: Distinguishing cause from effect. Trends Endocrinol. Metab. 2010, 21, 643–651. [Google Scholar] [CrossRef] [Green Version]
  310. Myers, M.G., Jr.; Heymsfield, S.B.; Haft, C.; Kahn, B.B.; Laughlin, M.; Leibel, R.L.; Tschop, M.H.; Yanovski, J.A. Challenges and opportunities of defining clinical leptin resistance. Cell Metab. 2012, 15, 150–156. [Google Scholar] [CrossRef] [Green Version]
  311. Heymsfield, S.B.; Greenberg, A.S.; Fujioka, K.; Dixon, R.M.; Kushner, R.; Hunt, T.; Lubina, J.A.; Patane, J.; Self, B.; Hunt, P.; et al. Recombinant leptin for weight loss in obese and lean adults: A randomized, controlled, dose-escalation trial. JAMA 1999, 282, 1568–1575. [Google Scholar] [CrossRef]
  312. Westerterp-Plantenga, M.S.; Saris, W.H.; Hukshorn, C.J.; Campfield, L.A. Effects of weekly administration of pegylated recombinant human OB protein on appetite profile and energy metabolism in obese men. Am. J. Clin. Nutr. 2001, 74, 426–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Hukshorn, C.J.; van Dielen, F.M.; Buurman, W.A.; Westerterp-Plantenga, M.S.; Campfield, L.A.; Saris, W.H. The effect of pegylated recombinant human leptin (PEG-OB) on weight loss and inflammatory status in obese subjects. Int. J. Obes. 2002, 26, 504–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Hukshorn, C.J.; Westerterp-Plantenga, M.S.; Saris, W.H. Pegylated human recombinant leptin (PEG-OB) causes additional weight loss in severely energy-restricted, overweight men. Am. J. Clin. Nutr. 2003, 77, 771–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  315. Zelissen, P.M.; Stenlof, K.; Lean, M.E.; Fogteloo, J.; Keulen, E.T.; Wilding, J.; Finer, N.; Rossner, S.; Lawrence, E.; Fletcher, C.; et al. Effect of three treatment schedules of recombinant methionyl human leptin on body weight in obese adults: A randomized, placebo-controlled trial. Diabetes Obes. Metab. 2005, 7, 755–761. [Google Scholar] [CrossRef] [PubMed]
  316. Korner, J.; Conroy, R.; Febres, G.; McMahon, D.J.; Conwell, I.; Karmally, W.; Aronne, L.J. Randomized double-blind placebo-controlled study of leptin administration after gastric bypass. Obesity 2013, 21, 951–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  317. Shetty, G.K.; Matarese, G.; Magkos, F.; Moon, H.S.; Liu, X.; Brennan, A.M.; Mylvaganam, G.; Sykoutri, D.; Depaoli, A.M.; Mantzoros, C.S. Leptin administration to overweight and obese subjects for 6 months increases free leptin concentrations but does not alter circulating hormones of the thyroid and IGF axes during weight loss induced by a mild hypocaloric diet. Eur. J. Endocrinol. 2011, 165, 249–254. [Google Scholar] [CrossRef]
  318. Quarta, C.; Sanchez-Garrido, M.A.; Tschop, M.H.; Clemmensen, C. Renaissance of leptin for obesity therapy. Diabetologia 2016, 59, 920–927. [Google Scholar] [CrossRef] [Green Version]
  319. Rosenbaum, M.; Murphy, E.M.; Heymsfield, S.B.; Matthews, D.E.; Leibel, R.L. Low dose leptin administration reverses effects of sustained weight-reduction on energy expenditure and circulating concentrations of thyroid hormones. J. Clin. Endocrinol. Metab. 2002, 87, 2391–2394. [Google Scholar] [CrossRef]
  320. Munzberg, H.; Myers, M.G., Jr. Molecular and anatomical determinants of central leptin resistance. Nat. Neurosci. 2005, 8, 566–570. [Google Scholar] [CrossRef]
  321. Ozcan, L.; Ergin, A.S.; Lu, A.; Chung, J.; Sarkar, S.; Nie, D.; Myers, M.G., Jr.; Ozcan, U. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009, 9, 35–51. [Google Scholar] [CrossRef] [Green Version]
  322. Banks, W.A. The blood-brain barrier as a cause of obesity. Curr. Pharm. Des. 2008, 14, 1606–1614. [Google Scholar] [CrossRef] [PubMed]
  323. Adam, C.L.; Findlay, P.A. Decreased blood-brain leptin transfer in an ovine model of obesity and weight loss: Resolving the cause of leptin resistance. Int. J. Obes. 2010, 34, 980–988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  324. Roth, J.D.; Roland, B.L.; Cole, R.L.; Trevaskis, J.L.; Weyer, C.; Koda, J.E.; Anderson, C.M.; Parkes, D.G.; Baron, A.D. Leptin responsiveness restored by amylin agonism in diet-induced obesity: Evidence from nonclinical and clinical studies. Proc. Natl. Acad. Sci. USA 2008, 105, 7257–7262. [Google Scholar] [CrossRef] [Green Version]
  325. Ravussin, E.; Smith, S.R.; Mitchell, J.A.; Shringarpure, R.; Shan, K.; Maier, H.; Koda, J.E.; Weyer, C. Enhanced weight loss with pramlintide/metreleptin: An integrated neurohormonal approach to obesity pharmacotherapy. Obesity 2009, 17, 1736–1743. [Google Scholar] [CrossRef] [PubMed]
  326. Bhavsar, S.; Watkins, J.; Young, A. Synergy between amylin and cholecystokinin for inhibition of food intake in mice. Physiol. Behav. 1998, 64, 557–561. [Google Scholar] [CrossRef]
  327. Trevaskis, J.L.; Lei, C.; Koda, J.E.; Weyer, C.; Parkes, D.G.; Roth, J.D. Interaction of leptin and amylin in the long-term maintenance of weight loss in diet-induced obese rats. Obesity 2010, 18, 21–26. [Google Scholar] [CrossRef] [PubMed]
  328. Williams, D.L.; Baskin, D.G.; Schwartz, M.W. Leptin regulation of the anorexic response to glucagon-like peptide-1 receptor stimulation. Diabetes 2006, 55, 3387–3393. [Google Scholar] [CrossRef] [Green Version]
  329. Bojanowska, E.; Nowak, A. Interactions between leptin and exendin-4, a glucagon-like peptide-1 agonist, in the regulation of food intake in the rat. J. Physiol. Pharmacol. 2007, 58, 349–360. [Google Scholar]
  330. Zhao, S.; Kanoski, S.E.; Yan, J.; Grill, H.J.; Hayes, M.R. Hindbrain leptin and glucagon-like-peptide-1 receptor signaling interact to suppress food intake in an additive manner. Int. J. Obes. 2012, 36, 1522–1528. [Google Scholar] [CrossRef] [Green Version]
  331. Kanoski, S.E.; Ong, Z.Y.; Fortin, S.M.; Schlessinger, E.S.; Grill, H.J. Liraglutide, leptin and their combined effects on feeding: Additive intake reduction through common intracellular signalling mechanisms. Diabetes Obes. Metab. 2015, 17, 285–293. [Google Scholar] [CrossRef] [Green Version]
  332. Muller, T.D.; Sullivan, L.M.; Habegger, K.; Yi, C.X.; Kabra, D.; Grant, E.; Ottaway, N.; Krishna, R.; Holland, J.; Hembree, J.; et al. Restoration of leptin responsiveness in diet-induced obese mice using an optimized leptin analog in combination with exendin-4 or FGF21. J. Pept. Sci. 2012, 18, 383–393. [Google Scholar] [CrossRef] [PubMed]
  333. Clemmensen, C.; Chabenne, J.; Finan, B.; Sullivan, L.; Fischer, K.; Kuchler, D.; Sehrer, L.; Ograjsek, T.; Hofmann, S.M.; Schriever, S.C.; et al. GLP-1/glucagon coagonism restores leptin responsiveness in obese mice chronically maintained on an obesogenic diet. Diabetes 2014, 63, 1422–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Yan, C.; Yang, Y.; Saito, K.; Xu, P.; Wang, C.; Hinton, A.O., Jr.; Yan, X.; Wu, Q.; Tong, Q.; Elmquist, J.K.; et al. Meta-chlorophenylpiperazine enhances leptin sensitivity in diet-induced obese mice. Br. J. Pharmacol. 2015, 172, 3510–3521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Labyb, M.; Chretien, C.; Caillon, A.; Rohner-Jeanrenaud, F.; Altirriba, J. Oxytocin Administration Alleviates Acute but Not Chronic Leptin Resistance of Diet-Induced Obese Mice. Int. J. Mol. Sci. 2018, 20, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  336. Folgueira, C.; Beiroa, D.; Gonzalez-Rellan, M.J.; Porteiro, B.; Milbank, E.; Castelao, C.; Garcia-Palacios, M.; Casanueva, F.F.; Lopez, M.; Dieguez, C.; et al. Uroguanylin Improves Leptin Responsiveness in Diet-Induced Obese Mice. Nutrients 2019, 11, 752. [Google Scholar] [CrossRef] [Green Version]
  337. Saladin, R.; De Vos, P.; Guerre-Millo, M.; Leturque, A.; Girard, J.; Staels, B.; Auwerx, J. Transient increase in obese gene expression after food intake or insulin administration. Nature 1995, 377, 527–529. [Google Scholar] [CrossRef]
  338. Kiess, W.; Englaro, P.; Hanitsch, S.; Rascher, W.; Attanasio, A.; Blum, W.F. High leptin concentrations in serum of very obese children are further stimulated by dexamethasone. Horm. Metab. Res. 1996, 28, 708–710. [Google Scholar] [CrossRef]
  339. Zhao, Y.; Dunbar, J.D.; Kharitonenkov, A. FGF21 as a therapeutic reagent. Adv. Exp. Med. Biol. 2012, 728, 214–228. [Google Scholar] [CrossRef]
Figure 1. Thermogenic actions of brown, beige and white fat. (1) Brown adipose tissue (BAT) is responsible for heat production to maintain cell homeostasis through a process called thermogenesis. The anatomical features of brown adipocytes are adapted for thermogenesis: many mitochondria using the surrounding multi-locular lipid droplets as fuel to dissipate energy and produce heat. Furthermore, brown adipocytes express several thermogenic markers such as UCP1, PPARγ, PGC1α and PRDM16. Finally, brown adipocytes originate from myogenic (Myf5+/+) lineage like myocytes. (2) On the other hand, white adipose tissue (WAT) is responsible for energy storage. In white adipocytes, fat accumulates in large lipid droplets (responsible of white adipocytes big diameter) that occupy the entire cytoplasm. White adipocytes do not carry any thermogenic functions nor thermogenic markers. (3) Beige fat has intermediate anatomical and functional characteristics between white and brown adipocytes. Thus, beige adipocytes emerge in white fat depots bearing thermogenic functions and expressing some thermogenic markers (at a lower proportion than BAT). Beige adipocytes can also store fat. Leptin is released by WAT and exerts its actions at central and peripheral levels. At a hypothalamic level, leptin favours anorexic effects through the overexpression of anorexigenic peptides and down-regulation orexigenic ones. Furthermore, leptin exerts its thermogenic actions in 2 different manners: (i) direct interactions with brown and beige adipocytes and (ii) through hypothalamic actions. UCP1, uncoupling protein 1; PPARγ, peroxisome proliferator-activated receptor γ; PGC1α, proliferator-activated receptor-gamma coactivator 1α; PRDM16, PR domain containing 16.
Figure 1. Thermogenic actions of brown, beige and white fat. (1) Brown adipose tissue (BAT) is responsible for heat production to maintain cell homeostasis through a process called thermogenesis. The anatomical features of brown adipocytes are adapted for thermogenesis: many mitochondria using the surrounding multi-locular lipid droplets as fuel to dissipate energy and produce heat. Furthermore, brown adipocytes express several thermogenic markers such as UCP1, PPARγ, PGC1α and PRDM16. Finally, brown adipocytes originate from myogenic (Myf5+/+) lineage like myocytes. (2) On the other hand, white adipose tissue (WAT) is responsible for energy storage. In white adipocytes, fat accumulates in large lipid droplets (responsible of white adipocytes big diameter) that occupy the entire cytoplasm. White adipocytes do not carry any thermogenic functions nor thermogenic markers. (3) Beige fat has intermediate anatomical and functional characteristics between white and brown adipocytes. Thus, beige adipocytes emerge in white fat depots bearing thermogenic functions and expressing some thermogenic markers (at a lower proportion than BAT). Beige adipocytes can also store fat. Leptin is released by WAT and exerts its actions at central and peripheral levels. At a hypothalamic level, leptin favours anorexic effects through the overexpression of anorexigenic peptides and down-regulation orexigenic ones. Furthermore, leptin exerts its thermogenic actions in 2 different manners: (i) direct interactions with brown and beige adipocytes and (ii) through hypothalamic actions. UCP1, uncoupling protein 1; PPARγ, peroxisome proliferator-activated receptor γ; PGC1α, proliferator-activated receptor-gamma coactivator 1α; PRDM16, PR domain containing 16.
Nutrients 12 00472 g001
Figure 2. Brown adipose tissue activation. Currently, 3 models of brown adipose tissue (BAT) activation have been proposed. (A) (Orange pathway) In the classical model, β adrenergic receptors (β-AR) in brown adipocytes are stimulated by sympathetic nervous system (SNS) induced-release of norepinephrine (NE). These G-protein coupled receptors then activate adenylate cyclase (AC), inducing an increase in cAMP, which in turn activates protein kinase A (PKA). PKA acute effect increase lipolysis by the activation of adipocyte triglyceride lipase (ATGL), hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL), which hydrolyse the triacylglycerides to release free fatty acids (FFA) that will enter the mitochondria and will eventually be used for heat production by uncoupling protein 1 (UCP1) in the electron transport chain. The chronic effect of PKA activation increases the expression of thermogenic related genes. (B) (Green pathway) A second model has been proposed in which thermogenesis is possible even in the absence of ATGL or associated enzymes, mainly by BAT capturing FFA released by white adipose tissue (WAT) and using them as activators and substrates of UCP1. (C) (Blue pathway) A third model includes the stimulation of brown adipocytes by NE, where a “non-fatty acid” activator activates UCP1 allowing the use of external FFA for the thermogenic process.
Figure 2. Brown adipose tissue activation. Currently, 3 models of brown adipose tissue (BAT) activation have been proposed. (A) (Orange pathway) In the classical model, β adrenergic receptors (β-AR) in brown adipocytes are stimulated by sympathetic nervous system (SNS) induced-release of norepinephrine (NE). These G-protein coupled receptors then activate adenylate cyclase (AC), inducing an increase in cAMP, which in turn activates protein kinase A (PKA). PKA acute effect increase lipolysis by the activation of adipocyte triglyceride lipase (ATGL), hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL), which hydrolyse the triacylglycerides to release free fatty acids (FFA) that will enter the mitochondria and will eventually be used for heat production by uncoupling protein 1 (UCP1) in the electron transport chain. The chronic effect of PKA activation increases the expression of thermogenic related genes. (B) (Green pathway) A second model has been proposed in which thermogenesis is possible even in the absence of ATGL or associated enzymes, mainly by BAT capturing FFA released by white adipose tissue (WAT) and using them as activators and substrates of UCP1. (C) (Blue pathway) A third model includes the stimulation of brown adipocytes by NE, where a “non-fatty acid” activator activates UCP1 allowing the use of external FFA for the thermogenic process.
Nutrients 12 00472 g002
Figure 3. Leptin-induced pathways. The binding of leptin to its receptors (LepR), mainly expressed in the hypothalamus, leads to its dimerization and recruitment and auto-phosphorylation of Janus tyrosine kinase 2 (JAK2). JAK2 subsequently phosphorylates LepR on 3 distinct tyrosine residues (Tyr985, Tyr1077 and Tyr1138), facilitating the binding of Src Homology 2 (SH2) containing molecules. Depending on the phosphorylated tyrosine, different pathways will be activated. Phosphorylation at Tyr985 allows the binding of (i) Suppressor Of Cytokine Signalling 3 (SOCS3) inducing negative feedback to inhibit JAK2 and (ii) SH2 domain protein tyrosine phosphatase 2 (SHP2) to activate Extracellular-signal Regulated Kinase (ERK) pathways. The Tyr1077 permits the phosphorylation and activation of Signal Transducer And Activator of Transcript 5 (STAT5) leading to gene expression changes. Finally, the Tyr1138 allows the phosphorylation of STAT3 altering gene expression and activating SOCS3, reinforcing the negative JAK2 feedback. Furthermore, the leptin pathway is interconnected with the insulin pathway, since JAK2 activation phosphorylates Insulin Receptor (IR) and Substrate (IRS), and subsequently, Phosphatidyl Inositol 3 Kinase (PI3K), protein kinase B (Akt) leading to the activation of Forkhead box protein O1 (FOXO1) and also mammalian Target Of Rapamycin (mTOR)/ribosomal S6 kinase (S6K). Together, all these leptin-activated signalling pathways trigger energy homeostasis gene regulation, mainly consisting in over-transcription of anorexigenic Pro-opiomelanocortin (POMC) and down-transcription of orexigenic Agouti Related Protein (Agrp) and Neuropeptide Y (NPY) neuropeptides, as well as favouring energy expenditure genes, including thermogenic markers.
Figure 3. Leptin-induced pathways. The binding of leptin to its receptors (LepR), mainly expressed in the hypothalamus, leads to its dimerization and recruitment and auto-phosphorylation of Janus tyrosine kinase 2 (JAK2). JAK2 subsequently phosphorylates LepR on 3 distinct tyrosine residues (Tyr985, Tyr1077 and Tyr1138), facilitating the binding of Src Homology 2 (SH2) containing molecules. Depending on the phosphorylated tyrosine, different pathways will be activated. Phosphorylation at Tyr985 allows the binding of (i) Suppressor Of Cytokine Signalling 3 (SOCS3) inducing negative feedback to inhibit JAK2 and (ii) SH2 domain protein tyrosine phosphatase 2 (SHP2) to activate Extracellular-signal Regulated Kinase (ERK) pathways. The Tyr1077 permits the phosphorylation and activation of Signal Transducer And Activator of Transcript 5 (STAT5) leading to gene expression changes. Finally, the Tyr1138 allows the phosphorylation of STAT3 altering gene expression and activating SOCS3, reinforcing the negative JAK2 feedback. Furthermore, the leptin pathway is interconnected with the insulin pathway, since JAK2 activation phosphorylates Insulin Receptor (IR) and Substrate (IRS), and subsequently, Phosphatidyl Inositol 3 Kinase (PI3K), protein kinase B (Akt) leading to the activation of Forkhead box protein O1 (FOXO1) and also mammalian Target Of Rapamycin (mTOR)/ribosomal S6 kinase (S6K). Together, all these leptin-activated signalling pathways trigger energy homeostasis gene regulation, mainly consisting in over-transcription of anorexigenic Pro-opiomelanocortin (POMC) and down-transcription of orexigenic Agouti Related Protein (Agrp) and Neuropeptide Y (NPY) neuropeptides, as well as favouring energy expenditure genes, including thermogenic markers.
Nutrients 12 00472 g003
Figure 4. Hypothalamic leptin-sensitive neurons regulating thermogenesis and browning. The leptin receptor (LepR) is highly expressed in several hypothalamic areas implicated in the thermogenic program. The Dorsomedial Hypothalamus (DMH) (high density of LepR), the Preoptic Area (POA) and the Steroidogenic Factor 1 (SF1) expressing neurons of the ventromedial hypothalamus (VMH) have a significant role in the control of the Sympathetic Nervous System (SNS) outflow to the brown adipose tissue (BAT) and the white adipose tissue (WAT) to control thermogenesis and browning respectively. Neurons in these areas are interconnected and can control the sympathetic activity regulating downstream effector neurons in the Raphe Pallidus (RPa). The Arcuate nucleus of the hypothalamus (ARC) also has high expression of LepR: different ARC neuronal populations exert opposing effects on the thermogenesis through the inhibition of Agouti-Related Protein (AgRP)/Neuropeptide Y (NPY) neurons and activation of Pro-opiomelanocortin (POMC) ones. These neurons can act on the Paraventricular nucleus of the hypothalamus (PVH) to decrease or induce thermogenesis respectively. LepR is also expressed in extra-hypothalamic areas, such as POMC neurons in the Nucleus of the Solitary Tract (NTS); these neurons project to different hypothalamic nuclei but their actions are distinct from the POMC expressing neurons of the ARC. Although LepR is expressed in the PVH the role of the PVH in regulating thermogenesis is not completely clear, likely via indirect connections with another nucleus. TRH: thyrotropin-releasing hormone. PrRP: prolactin releasing peptide. mPOA: medial POA.
Figure 4. Hypothalamic leptin-sensitive neurons regulating thermogenesis and browning. The leptin receptor (LepR) is highly expressed in several hypothalamic areas implicated in the thermogenic program. The Dorsomedial Hypothalamus (DMH) (high density of LepR), the Preoptic Area (POA) and the Steroidogenic Factor 1 (SF1) expressing neurons of the ventromedial hypothalamus (VMH) have a significant role in the control of the Sympathetic Nervous System (SNS) outflow to the brown adipose tissue (BAT) and the white adipose tissue (WAT) to control thermogenesis and browning respectively. Neurons in these areas are interconnected and can control the sympathetic activity regulating downstream effector neurons in the Raphe Pallidus (RPa). The Arcuate nucleus of the hypothalamus (ARC) also has high expression of LepR: different ARC neuronal populations exert opposing effects on the thermogenesis through the inhibition of Agouti-Related Protein (AgRP)/Neuropeptide Y (NPY) neurons and activation of Pro-opiomelanocortin (POMC) ones. These neurons can act on the Paraventricular nucleus of the hypothalamus (PVH) to decrease or induce thermogenesis respectively. LepR is also expressed in extra-hypothalamic areas, such as POMC neurons in the Nucleus of the Solitary Tract (NTS); these neurons project to different hypothalamic nuclei but their actions are distinct from the POMC expressing neurons of the ARC. Although LepR is expressed in the PVH the role of the PVH in regulating thermogenesis is not completely clear, likely via indirect connections with another nucleus. TRH: thyrotropin-releasing hormone. PrRP: prolactin releasing peptide. mPOA: medial POA.
Nutrients 12 00472 g004

Share and Cite

MDPI and ACS Style

Seoane-Collazo, P.; Martínez-Sánchez, N.; Milbank, E.; Contreras, C. Incendiary Leptin. Nutrients 2020, 12, 472. https://doi.org/10.3390/nu12020472

AMA Style

Seoane-Collazo P, Martínez-Sánchez N, Milbank E, Contreras C. Incendiary Leptin. Nutrients. 2020; 12(2):472. https://doi.org/10.3390/nu12020472

Chicago/Turabian Style

Seoane-Collazo, Patricia, Noelia Martínez-Sánchez, Edward Milbank, and Cristina Contreras. 2020. "Incendiary Leptin" Nutrients 12, no. 2: 472. https://doi.org/10.3390/nu12020472

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop