Skip to main content
Endocrinology logoLink to Endocrinology
. 2018 Dec 11;160(2):343–358. doi: 10.1210/en.2018-00747

Lateral Hypothalamic Mc3R-Expressing Neurons Modulate Locomotor Activity, Energy Expenditure, and Adiposity in Male Mice

Hongjuan Pei 1, Christa M Patterson 2, Amy K Sutton 3, Korri H Burnett 1, Martin G Myers Jr 2,3, David P Olson 1,3,
PMCID: PMC6937456  PMID: 30541071

Abstract

The central melanocortin system plays a crucial role in the control of energy balance. Although the decreased energy expenditure and increased adiposity of melanocortin-3 receptor (Mc3R)–null mice suggest the importance of Mc3R-regulated neurons in energy homeostasis, the roles for specific subsets of Mc3R neurons in energy balance have yet to be determined. Because the lateral hypothalamic area (LHA) contributes to the control of energy expenditure and feeding, we generated Mc3rcre mice to determine the roles of LHA Mc3R (Mc3RLHA) neurons in energy homeostasis. We found that Mc3RLHA neurons overlap extensively with LHA neuron markers that contribute to the control of energy balance (neurotensin, galanin, and leptin receptor) and project to brain areas involved in the control of feeding, locomotion, and energy expenditure, consistent with potential roles for Mc3RLHA neurons in these processes. Indeed, selective chemogenetic activation of Mc3RLHA neurons increased locomotor activity and augmented refeeding after a fast. Although the ablation of Mc3RLHA neurons did not alter food intake, mice lacking Mc3RLHA neurons displayed decreased energy expenditure and locomotor activity, along with increased body mass and adiposity. Thus, Mc3R neurons lie within LHA neurocircuitry that modulates locomotor activity and energy expenditure and contribute to energy balance control.


Obesity is a major global public health concern and is associated with pathological disorders, including type 2 diabetes, hypertension, cardiovascular disease, and cancer (1). Development of effective antiobesity therapies requires an understanding of the physiologic systems that regulate energy homeostasis. In this regard the melanocortin system is critical for energy balance via its regulation of food intake and energy expenditure (2). The central melanocortin system consists of the endogenous melanocortin receptor agonist proopiomelanocortin (POMC), the endogenous melanocortin receptor antagonist agouti-related protein (AgRP), and a family of five seven-transmembrane G protein–coupled melanocortin receptors (Mc1R to Mc5R) (2, 3). Mc3R and Mc4R are the predominant melanocortin receptors in the central nervous system (CNS). Mc4Rs are expressed widely in the CNS, and Mc4R mutations are associated with severe hyperphagic obesity in both rodents and humans (4–6). Mc3Rs are expressed in the hypothalamus and limbic structures of the brain that regulate feeding behaviors and autonomic function (7, 8), and Mc3r gene polymorphisms have been linked to childhood obesity (9–11). Deletion of Mc3r in mice increases fat mass, reduces lean mass, and increases susceptibility to diet-induced obesity. Loss of Mc3r also results in reduced locomotor activity and voluntary wheel running, implicating Mc3R-expressing cells in the regulation of locomotion (12–17). Selective re-expression of Mc3Rs in the CNS of an Mc3r-null background partially rescues the obesity and activity defects accompanying global Mc3r inactivation (16); although this indicates an important contribution of Mc3R-bearing neurons in energy balance control, the neural substrates and physiologic mechanisms by which these effects are mediated are not fully known.

Within the hypothalamus, Mc3R is expressed in the arcuate nucleus (ARC), the ventromedial nucleus of the hypothalamus (VMH), the lateral hypothalamic area (LHA), and the preoptic nucleus (7). The LHA is of particular interest, as it has long been appreciated for its crucial roles in the regulation of feeding, body weight maintenance, and metabolism. LHA lesions lead to reduced feeding, drinking, and body weight (18). Alternatively, electrical stimulation or glutamate receptor activation within the LHA potently stimulates food intake (19–21). LHA neurons are heterogeneous in regard to neuropeptide and neurotransmitter content and project widely throughout the CNS (22). Genetic inactivation of LHA glutamatergic neurons is associated with increased feeding and subsequent weight gain on a high-fat diet (HFD) (23), whereas activation of LHA γ-aminobutyric acid (GABA)ergic neurons drives consummatory behaviors with a positive valence (24). Multiple LHA cell types, including neurons expressing melanin-concentrating hormone (MCH), orexin (OX)/hypocretin, galanin, leptin receptors (LepRb), and neurotensin (Nts), have been implicated in energy balance control (25). MCH stimulates feeding and contributes to the long-term regulation of body weight, as MCH knockout mice are hypophagic and lean relative to controls (26). A subset of Nts-expressing LHA cells also expresses the LepRb, and leptin action in NtsLHA neurons contributes to body weight regulation and reward behaviors (27). NtsLHA neurons project to the ventral tegmental area (VTA) and modulate locomotor activity through the mesolimbic dopamine system (28–30). Additionally, LepRb/Nts-expressing LHA cells indirectly regulate local OX neurons, which may also contribute to feeding and locomotor activity control (25, 31). Subsets of galanin cells in the LHA (GalLHA) express LepRb and Nts, and GalLHA neurons contribute to energy balance control by modulating noncompulsive locomotion, energy expenditure, and food reward, but not consummatory behaviors per se (32, 33). GalLHA cells do not project to the VTA (34), but galanin has been shown to directly regulate the activity of OX neurons, presumably through local release in the LHA (31). Thus, LHA neurons marked by Nts, LepRb, and galanin contribute to energy balance control by modifying feeding behavior, food reward, and locomotor activity at least in part by modulation of VTA cells and OX neurons.

In the context of the melanocortin system, the LHA receives projections from both POMC and AGRP/NPY neurons (35), and overexpression of a melanocortin receptor antagonist in the LHA promotes obesity in response to an HFD (36). Additionally, optogenetic activation of AgRP terminals in the LHA stimulates feeding (37), suggesting a role for melanocortin receptor–bearing LHA cells in feeding behavior. Although re-expression of Mc4Rs only in the LHA of an otherwise null Mc4r background alters glucose homeostasis and sympathetic output, it does not alter feeding (38). The physiological importance of Mc3RLHA neurons in the regulation of feeding, locomotor activity, and energy expenditure is largely unknown. To reveal the roles of Mc3RLHA neurons in energy homeostasis, we generated a knock-in transgenic line that expresses Cre recombinase in Mc3R-containing cells (Mc3rcre) and applied genetic and viral tools to identify and manipulate Mc3RLHA neurons and determine their contributions to energy balance.

Materials and Methods

Experimental animals

Mc3R-T2A-Cre mice (Mc3rcre) were generated using recombineering techniques as previously described (39). Briefly, the stop codon of Mc3r was replaced with a T2A-Cre fusion transgene that effectively tethers Cre recombinase to the Mc3r gene product via an 18–amino acid self-cleaving peptide sequence from the Thosea asigna virus. The final targeting construct containing the Mc3R-T2A-Cre fusion and 4 kb of flanking genomic sequence was electroporated into embryonic stem cells followed by neomycin selection. Appropriately targeted clones were identified by quantitative PCR and confirmed by Southern blot analysis. Targeted clones were expanded and injected into blastocysts by the University of Michigan Transgenic Core. Chimeric offspring were then bred to confirm germline transmission of the Mc3rcre allele; the neomycin selection cassette was removed by breeding to a Flpe deleter strain (The Jackson Laboratory stock no. 003946). Offspring were genotyped using primers that spanned the stop codon of the Mc3r locus and internal to the Cre transgene sequence. Mc3rcre mice were then bred to the Cre-dependent Ai9 Tomato reporter strain (The Jackson Laboratory stock no. 007909) to visualize Cre recombinase activity by fluorescent immunohistochemistry. Mc3rcre × Ai9-TdTomato mice (Mc3rcre-Tomato) were bred to the Gad1-GFP and Galanin-GFP backgrounds (33, 40) to generate animals expressing tdTomato in Mc3R neurons and GFP in Gad1- or Galanin-expressing cells, respectively. All mice were housed in a 12-hour light/12-hour dark cycle and cared for by the Unit for Laboratory Animal Medicine at the University of Michigan. Animals had ad libitum access to food and water, except in experiments when mice were fasted before perfusion or for timed feeding studies. All animal care and procedures were in accordance with the guidelines of, and approved by, the University of Michigan’s Committee on the Care and Use of Animals.

RNAscope in situ hybridization assays

Following rapid decapitation of mice under anesthesia, brains were flash frozen on 2-methylbutane at −20°C and stored at −80°C. The brains were cut to thickness of 16 μm on a cryostat and thaw mounted onto Superfrost Plus slides (Fisher Scientific). Slides were stored at −80°C until further processing. In situ hybridization was performed according to the RNAscope® 2.5 HD duplex detection kit user manual for fresh frozen tissue (Advanced Cell Diagnostics). Probes used are as follows: RNAscope® Probe-Mm-Mc3r-C1 (catalog no. 412541), RNAscope® Probe-Mm-Mc4r-C2 (catalog no. 319181-C2), and RNAscope® Probe-Cre-C2 (catalog no. 312281-C2).

Perfusion and immunohistochemistry

For some experiments, peptide colocalization required treatment with ICV colchicine (10 μg) 2 days prior to perfusion to trap neuropeptides in the cell soma. For perfusion, adult mice were euthanized with an overdose of IP pentobarbital injection and transcardially perfused with sterile PBS, followed by neutral buffered 10% formalin. Brains were removed, postfixed, and dehydrated in 30% sucrose before sectioning as 30-μm coronal slices using a freezing microtome (Leica). Coronal brain sections were collected as four representative series and stored at −20°C until further use.

For Fos immunohistochemistry (IHC), sections were pretreated with 0.3% H2O2, blocked in 3% normal donkey or goat serum and then incubated with primary antibodies for c-Fos (41) (1:10,000). Detection of primary Fos antibody was performed by the avidin-biotin/diaminobenzidine (DAB) method [Biotin-SP–conjugated donkey anti-rabbit; 1:200; Jackson ImmunoResearch Laboratories (42); ABC kit, Vector Laboratories; DAB reagents, Thermo Scientific). For hM3Dq–designer receptors exclusively activated by designer drug (DREADD)–mCherry and tract-tracing experiments, immunostaining was performed using primary antibody anti-dsRed (43) (Living Colors, rabbit, 1:1000), followed by secondary immunofluorescence detection with donkey anti-rabbit Alexa Fluor 568 (44) (1:200; Invitrogen). Peptide colocalization was performed using primary antibodies for anti-GFP (45) (Invitrogen A6455; rabbit; 1:4000), anti-Nts (46) (Millipore; rabbit; 1:1000), anti-OX (47) (Santa Cruz Biotechnology; goat; 1:1000), and anti-MCH (48) (Santa Cruz Biotechnology, goat, 1:1000) followed by secondary immunofluorescent detection with species-specific Alexa Fluor 488 or 568 antibodies (49, 50) (Invitrogen; 1:200) and processed for imaging. To identify neurons responsive to leptin, fasted mice were IP injected with sterile PBS or leptin (5 mg/kg) 2 hours before perfusion. For phosphorylated signal transducer and activator of transcription-3 (p-Stat3) IHC, sections were pretreated with 1% NaOH and 1% H2O2, 0.3% glycine, and 0.03% SDS, and then blocked with normal donkey or goat serum and incubated with primary antibody (51) (anti–p-Stat3; Cell Signaling Technology; rabbit; 1:1000). Detection of primary p-Stat3 antibody was done using the DAB method.

Stereotaxic virus injections

Adult (>8-week-old) Mc3rcre mice were anesthetized with vaporized isoflurane and fixed in a stereotaxic apparatus. Coordinates used to target the LHA were A/P, −1.34 mm; M/L, ±1.13 mm; and D/V, −5.2 mm relative to bregma. Viral injections (200 nL) were performed using a pressurized Picospritzer system coupled to a pulled glass micropipette. After 5 minutes to allow for tissue absorption of virus, the glass micropipette was removed, the cranial access was filled with bone wax, and the surgical incision was closed with sutures. Mice were allowed to recover for at least 10 days before they were used in physiologic experiments. At the conclusion of all experiments, mice were perfused and brains were harvested for IHC to confirm appropriate viral targeting. Adenovirus Ad-in synaptophysin-mCherry was prepared by the University of Michigan Viral Vector Core with a titer of 2.7 × 1012 genome copies/mL. Adeno-associated viruses (AAVs) used in these studies were purchased from the University of North Carolina Viral Vector Core with viral titers ranging from 2 to 5.9 × 1012 genome copies/mL and included (i) Cre-inducible AAV8-Syn-DIO-hM3Dq-mCherry, (ii) Cre-inducible rAAV2-Flex-taCasp3-TEVP, and (iii) AAV8-CAG-flexGFP.

Energy balance studies

Acute neuronal activation studies

After recovery from bilateral stereotaxic delivery of Cre-dependent hM3Dq-DREADD-mCherry virus to the LHA, Mc3rcre mice were habituated to daily IP injections for 5 days with sterile PBS prior to food intake assessments following DREADD-mediated activation of Mc3RLHA neurons. To assess refeeding after a fast, food was removed at the onset of the dark cycle (6:00 pm) the day before the test. On the morning of the second day, mice were injected IP with 0.5 mL of vehicle or clozapine N-oxide (CNO; 0.3 mg/kg) and food was returned to the cage. Cumulative food intake was measured 2, 4, and 8 hours following injection. Each mouse was used as its own control using a cross-over experimental design to exclude batch effects.

Chronic neuronal inhibition studies

Cohorts of male Mc3rcre mice were injected bilaterally in the LHA with 200 nL of AAV2-flex-taCasp3-TEVp virus (52). Weekly body weight and food intake were subsequently measured during the ensuing 12 weeks. For acute refeeding studies, food was removed at the onset of the dark cycle (6:00 pm) the night before the test; the next day, food was given back and cumulative food intake was measured at 2, 4, and 8 hours. At the end of the study, fat depots were collected and weighed at the time of perfusion. Brains were harvested following perfusion and processed by IHC to confirm appropriate targeting and cell ablation.

Metabolic and behavioral profiling

Energy expenditure and body composition

Energy expenditure and X-ambulatory activity were measured using a comprehensive laboratory monitoring system (Columbus Instruments) in the University of Michigan Small Animal Phenotyping Core. The comprehensive laboratory monitoring system housing maintained 12-hour dark/12-hour light cycles and an ambient temperature of 20 to 23°C. Prior to the start of data acquisition, mice were acclimatized to the sealed chambers for 2 days with free access to food and water before data were collected. Body fat and lean mass were determined using an nuclear magnetic resonance–based analyzer (Minispec LF90II; Bruker Optics) in the University of Michigan Small Animal Phenotyping Core.

Open-field activity

Separate cohorts of Mc3rcre mice injected bilaterally in the LHA with the Cre-dependent stimulatory hM3Dq-DREADD-mCherry virus were used in an open-field activity test. Mice were removed from their home cages during the light cycle and acclimated for 2 hours in an open-field arena (ENV-017M; Med Associates) without access to food or water. After acclimation, open-field activity was recorded for 30 minutes at baseline, 30 minutes after saline injection, and 90 minutes after CNO injection (0.3 mg/kg, IP). To determine the importance of the mesolimbic dopamine system in these responses, additional animals were pretreated with the dopamine receptor-1 antagonist SCH23390 (0.1 mg/kg, IP) 30 minutes before CNO administration. Data are pooled for each genotype and treatment and expressed as total ambulatory movements. At the conclusion of the experiment, mice were perfused and brains were harvested and processed by IHC to confirm appropriate targeting of the AAV- hM3Dq-DREADD-mCherry in the LHA. Animals with hM3Dq-DREADD-mCherry expression outside the LHA were excluded from analysis.

Statistical analysis

Data were analyzed and graphs were generated using GraphPad Prism. Paired t tests and two-way ANOVAs were used to determine significance of the data collected and are noted in the figure legends. Results were considered significant with a P value <0.05 (*P < 0.05, **P < 0.01, ***P < 0.001).

Results

Mc3rcre knock-in mice allow manipulation of Mc3R-expressing cells

To genetically identify and selectively manipulate Mc3R neurons, we generated knock-in mice that express Cre recombinase in Mc3R neurons (Mc3rcre) [Fig. 1(a)]. To confirm the specific expression of Cre in Mc3R neurons, we crossed Mc3rcre mice with the Ai9 Td-tomato reporter line and found tomato reporter expression in all brain regions known to express Mc3Rs [Fig. 1(b)]. To confirm that the expression of Cre activity recapitulates endogenous Mc3r expression in vivo, we used an RNAscope dual chromogen assay to detect Mc3r and Cre mRNAs. RNAscope in situ hybridization demonstrated nearly complete colocalization of Mc3R and Cre mRNAs in the VMH, ARC, and LHA [Fig. 1(c)–1(e)]. Melanocortin agonists and antagonists (POMC and AGRP peptides, respectively) are produced by neurons in the ARC and these neurons project to the LHA. To determine whether melanocortin peptides might target Mc3RLHA neurons, we stained Mc3rcre-Tomato brains for both AGRP and POMC immunoreactivities. Immunostaining revealed dense AgRP fibers in the LHA with AGRP boutons lying in close proximity to Mc3RLHA neurons [Fig. 1(f)]. We also found dense POMC fiber boutons in apposition to Mc3RLHA cell bodies [Fig. 1(g)], indicating that Mc3RLHA neurons are positioned to receive and transmit melanocortin signals through LHA neurocircuitry that modulates energy balance and behavior.

Figure 1.

Figure 1.

(a) Schematic of Mc3rcre allele and cre-mediated Td-Tomato expression in Mc3R-expressing cells of Mc3rcre-Tomato mice. (b) Immunofluorescent detection of Td-tomato (red) in the LHA, VMH, and ARC of Mc3rcre-Tomato mice. (c–e) RNAscope dual chromogen staining shows colocalization of cre transcripts (red) with Mc3R- expressing neurons (blue) in the (c) VMH, (d) ARC, and (e) LHA from Mc3Rcre mice. (f) AgRP (green) immunostaining in the LHA of Mc3rcre-Tomato (red) mice. (g) POMC (green) immunostaining in the LHA of Mc3rcre-Tomato (red) mice. Scale bars: (b) 1 mm, (c–e) 20 μm, and (f and g) 100 μm.

Neurochemical characterization of LHA Mc3R-expressing neurons

As multiple LHA cell types have been shown to contribute to energy balance regulation (22, 53), we used Mc3rcre × Ai9-TdTomato mice (Mc3rcre-Tomato) to examine the expression of known neuropeptides in Mc3RLHA neurons. Similar to LepRb-, Nts-, and galanin-expressing LHA neurons, we found little colocalization of Mc3RLHA neurons with OX or MCH [Fig. 2(a) and 2(b)]. However, ∼45% of Mc3RLHA neurons coexpressed the neurotransmitter Nts [Fig. 2(c)], whereas ∼28% of Mc3RLHA neurons contain galanin [Fig. 2(d)]. To determine whether Mc3RLHA neurons contain leptin receptors (LepRb), we treated fasted Mc3rcre-Tomato mice with leptin and examined the leptin-induction of p-STAT3; we found that ∼42% of Mc3RLHA neurons contained leptin-stimulated p-STAT3 [Fig. 2(e)]. Most Mc3RLHA neurons appear to be GABAergic based on the extensive colocalization of Mc3rcre-Tomato LHA neurons with GFP in a GAD1-GFP background, which labels GAD1-expressing GABAergic neurons (40) [Fig. 2(f)]. The overlap of Mc3RLHA neurons with Nts, galanin, and LepRb suggests that Mc3RLHA neurons lie within LHA neural circuits that modulate feeding, energy expenditure, and locomotor activity and that their activity may contribute to these energy balance parameters.

Figure 2.

Figure 2.

(a and b) Immunofluorescent detection of Td-tomato (red) and OX [(a) green] or MCH [(b) green] in the LHA of Mc3rcre-Tomato mice. (c) Immunofluorescent detection of Td-tomato (red) and Nts (green) in the LHA Mc3rcre-Tomato mice 48 h after colchicine treatment. (d) Immunofluorescent detection of Td-tomato and GFP in Mc3rcre-Tomato, GalaninGFP mice. (e) Immunohistochemical detection of p-STAT3 (green) and Td-tomato (red) in the LHA of Mc3rcre-Tomato mice 1 h after IP injection of leptin. (f) Immunofluorescent detection of Td-tomato (red) and GFP (green) in Mc3rcre-Tomato, GAD1GFP mice. Scale bars, 100 μm. Quantification of Mc3R colocalization shown in pie chart inset: red indicates percentage Mc3R neuron not colocalized; yellow indicates percentage Mc3R neuron colocalized with peptide of interest.

Determining the circuitry in which Mc3RLHA neurons reside is essential for understanding how these neurons might contribute to metabolic function. To trace efferent projections from Mc3RLHA neurons, we injected a Cre-dependent adenoviral synaptophysin:mCherry fusion construct unilaterally into the LHA of Mc3rcre mice. The synaptophysin:mCherry fusion protein preferentially accumulates in neuron terminals, allowing identification of projection target areas (54) [Fig. 3(a)]. Unilateral synaptophysin:mCherry injection revealed dense terminal projections within the LHA itself [Fig. 3(b)]. Additionally, adenoviral synaptophysin:mCherry transduction of Mc3RLHA neurons clearly labeled neural projection terminals in the paraventricular nucleus of the hypothalamus (PVH) and parabrachial nuclei (PBN) [Fig. 3(d) and 3(f)], brain regions known to regulate feeding (55). Mc3RLHA cells also sent dense projections to the bed nucleus of the stria terminalis (BNST) and VTA [Fig. 3(c) and 3(e)], which play roles in reward seeking and addiction (56, 57). VTA neurons also play crucial roles in the control of locomotor activity and energy expenditure (29, 58, 59). [See Fig. 3(g) for injection site summary and Table 1 for relative projection densities to brain areas targeted by Mc3RLHA neurons.] Activation of GABAergic-, Nts-, and Galanin-expressing LHA neurons have all been associated with alterations in locomotor activity, although galanin neurons do not project to the VTA. Although these populations do not overlap with OX neurons, LHALepRb neurons regulate OX neurons indirectly, and galanin has been shown to modulate OX neuron activity directly (31). These data suggest that local modulation of OX neuron activity by neighboring LHA cell types may also modify motivated behaviors and locomotor activity. Thus, the neurotransmitter content and projections of Mc3RLHA neurons led us to hypothesize a role for these cells in the regulation of activity, energy expenditure, and energy homeostasis in part through the control of OX neurons and the VTA.

Figure 3.

Figure 3.

(a) Schematic of cre-mediated expression of synaptophysin (syn)-mCherry in Mc3Rcre mice. (b) Immunofluorescent detection of mCherry in the LHA of Mc3Rcre mice following intra-LHA injection of Ad-syn-mCherry virus. (c–f) Immunofluorescent detection of syn-mCherry–containing projections in the (c) BNST, (d) PVH, (e) VTA, and (f) PBN after intra-LHA injection of Ad-syn-mCherry virus. Scale bars, 100 μm. (g) Summary of Ad-IN Syn-mCherry injection sites of Mc3RCre mice included in the study (n = 3). 3V, third ventricle; BNST, bed nucleus of the stria terminalis; lPBN, lateral parabrachial nucleus; PBN, parabrachial nuclei; PVH, paraventricular nucleus of the hypothalamus; scp, superior cerebellar peduncle; SN, substantia nigra.

Table 1.

Mc3Rcre LHA Projection Sites Using Cre-Dependent Viral Tracing (Ad-IN-syn-mCherry) in the LHA

Brain Area Intensity
BNST ++++
Parabrachial nucleus, medial division, medial medial part ++++
PVH +++
VTA +++
Substantia nigra, reticular part ++
Periaqueductal gray ++
Nucleus of the solitary tract +

Terminal projection sites and the relative intensity of projections in the different brain areas are shown.

Activation of Mc3RLHA neurons increases locomotor activity and augments refeeding

To establish a role for Mc3RLHA neurons in energy balance control, we used DREADDs technology to enable remote neuronal activation by the pharmacologically inert ligand CNO (60). The stimulatory DREADD (hM3Dq-DREADD-mCherry) couples through the Gq pathway to depolarize neurons and increase their firing rate in response to CNO. Mc3rcre mice were injected stereotaxically in the LHA with the Cre-dependent, stimulatory AAV-hM3Dq-DREADD-mCherry vector (Mc3RLHA-hM3Dq mice). Using Fos staining as an indicator for neuronal activation, we found that CNO treatment increased nuclear Fos immunoreactivity in Mc3RLHA-hM3Dq mice, compared with vehicle controls [Fig. 4(a) and 4(b)], demonstrating that cell-specific DREADD expression permits the CNO-dependent activation of Mc3RLHA neurons. A representation of LHA injection sites and viral transduction is found in Fig. 4(i). As we hypothesized that Mc3RLHA cells may modulate the activity of OX neurons and cells of the VTA to exert their physiologic effects, we also examined Fos in OX neurons and the VTA. We found increased nuclear Fos staining [Fig. 4(c), 4(d), and 4(g)] in OX cells and in the VTA following Mc3RLHA neuron activation [Fig. 4(e), 4(f), and 4(h)]. Because the activation of OX cells and the VTA is associated with alterations in locomotor activity and food intake under some circumstances (29, 61, 62), the increase in nuclear Fos expression seen following Mc3RLHA activation suggests the potential modulation of these behaviors by Mc3RLHA neuron activity.

Figure 4.

Figure 4.

(a and b) Fos expression in the LHA of Mc3Rcre-DREADD mice after (a) vehicle or (b) CNO (0.3 mg/kg, IP) treatment; representative images of mCherry-IR (red) and Fos-IR (green). Scale bars, 100 μm. (c and d) Immunodetection of Fos (red) and OX (green) in the LHA of Mc3Rcre-DREADD mice injected with (c) vehicle or (d) CNO. Scale bars, 100 μm. (e and f) Immunodetection of Fos in the VTA of Mc3Rcre-DREADD mice injected with (e) vehicle or (f) CNO. Scale bars, 100 μm. (g) Quantification of OX and Fos colocalization in Mc3Rcre-DREADD mice treated with vehicle or CNO (SEM, n = 7; ****P < 0.0001). (h) Number of Fos-positive cells in the VTA of Mc3Rcre-DREADD mice treated with vehicle or CNO (SEM, n = 4; **P < 0.01). (i) Summary of AAV-3Dq-DREADD LHA injection sites of Mc3RCre mice included in the study (n = 9); injection sites are represented as two groups for clarity.

To determine the effect of pharmacologic Mc3RLHA neuron activation on feeding, we analyzed the feeding responses of Mc3RLHA-hM3Dq mice following Mc3RLHA activation in both the fed state and following a fast. For the fasting–refeeding paradigm, mice were fasted overnight and then pretreated with vehicle or CNO before food was given back during the light phase. As shown in Fig. 5(a), the CNO-induced activation of Mc3RLHA neurons in the fed state had little impact of feeding during an 8-hour period (vehicle, 0.52 ± 0.13 g; CNO, 0.49 ± 0.12 g). However, food intake following a fast was augmented with Mc3RLHA activation within the first 2 hours of refeeding [Fig. 5(b)], indicating that Mc3RLHA neuron activation can enhance feeding depending on metabolic state (fasted vs fed).

Figure 5.

Figure 5.

(a) Food intake of Mc3Rcre-DREADD mice following vehicle or CNO treatment in the fed state (SEM, n = 9; **P < 0.01). (b) Food intake of Mc3Rcre-DREADD mice following vehicle or CNO treatment after a fast (SEM, n = 9; ***P < 0.001). (c) Locomotor activity following hM3Dq-mediated activation of Mc3RLHA neurons in the absence of presence of by peripheral administration of a D1R antagonist, SCH23390 (0.1 mg/kg, IP). Activity (travel distance per 30 min) is binned for the 30 min after vehicle injection and averaged per 30 min for CNO treatment (SEM, n = 7; **P < 0.01). NS, not significant.

Activation of the mesolimbic dopamine system increases locomotor activity (63) and Nts action in the VTA is associated with increased locomotor activity and dopamine release in the nucleus accumbens (29). Because our viral tracing studies revealed Mc3RLHA neuron projections into the VTA, we hypothesized that Mc3RLHA neurons may directly modulate locomotor activity. To test this hypothesis, we determined changes in locomotor activity in Mc3RLHA-hM3Dq mice in response to treatment with vehicle or CNO (0.3 mg/kg). CNO-mediated Mc3RLHA neuron activation greatly increased the locomotor activity of these mice [Fig. 5(c)], suggesting a potentially important role for Mc3RLHA cells in the control of activity. This increased locomotor activity was dopamine-dependent, as pretreatment with the dopamine receptor-1 antagonist SCH23390 significantly attenuated the locomotor response to Mc3RLHA activation [Fig. 5(c)].

Mc3RLHA neurons are required for energy homeostasis

To determine the contributions of Mc3RLHA neurons to long-term energy homeostasis, we used the Cre-dependent virus AAV-flex-taCasp3-TEVp to conditionally express caspase and induce Mc3RLHA cell death in adult animals (52). Injection of AAV-flex-taCasp3-TEVp into Mc3rcre-Tomato mice selectively ablated Mc3R neurons in the LHA while leaving Mc3R neurons in the VMH and ARC essentially unperturbed [Fig. 6(a) and 6(b)]. Importantly, caspase ablation in the LHA was limited to Mc3RLHA neurons, as cell counts within the LHA showed greatly decreased numbers of Mc3RLHA neurons, whereas neighboring OX neurons were unaffected [Fig. 6(c)].

Figure 6.

Figure 6.

(a and b) Immunodetection of Td-tomato in the Mc3rcre-Tomato mice injected with (a) control or (b) AAV-flex-taCasp3-TEVp virus. (c) Quantification of Mc3rcre-Tomato neurons and OX neurons in the LHA from Mc3rcre-Tomato mice injected with control virus or AAV-flex-taCasp3-TEVp virus (AAV-Control, n = 5; AAV-Casp, n = 5). (d) Weekly food intake in Mc3rcre-Tomato mice injected with control virus or AAV-flex-taCasp3-TEVp virus. (e) Food intake at 2, 4, and 8 h after a fast. (f) Weekly body weight change of Mc3rcre-Tomato mice injected with control virus or AAV-flex-taCasp3-TEVp virus. (g) Percentage fat and lean mass of Mc3rcre-Tomato mice injected with control virus and AAV-flex-taCasp3-TEVp virus. (h) Fat pad weights of Mc3rcre-Tomato mice injected with control virus or AAV-flex-taCasp3-TEVp virus. Average values ± SEM are shown (AAV-Control, n = 8; AAV-Casp, n = 6). *P < 0.05; **P < 0.01; ****P < 0.0001. NS, not significant.

After establishing the effectiveness of Cre-dependent caspase ablation, a cohort of Mc3rcre mice was stereotaxically injected with AAV-flex-GFP control virus or AAV-flex-taCasp3-TEVp virus to determine the necessity of Mc3RLHA neurons in energy homeostasis. After recovery and allowing time for caspase expression to induce cell death, we measured food intake and body weight weekly for both groups of mice. We found no significant difference in either weekly food intake or acute fast–refeeding food intake between control and Mc3RLHA-ablated mice [Fig. 6(d) and 6(e)]. Although the mean body weights of Mc3RLHA-ablated mice were no different than the control mice 12 weeks after virus injection (Mc3RLHA-ablated, 31.59 ± 5.5 g vs controls, 29.35 ± 4.9 g) owing to differences in weight at the time of injection (Mc3RLHA-ablated, 27.61 ± 3.8 g vs controls, 28.85 ± 2.9 g), Mc3RLHA-deleted mice gained weight more quickly than did the controls [Fig. 5(f)]. Furthermore, body composition analysis revealed an increased percentage of body fat in Mc3RLHA-ablated mice, indicating a propensity for fat mass accrual [Fig. 6(g)]. Indeed, fat pad dissection at the time of perfusion revealed increased gonadal fat in Mc3RLHA-deleted mice [Fig. 6(h)].

Because the accrual of excess fat mass in the setting of equivalent food intake suggested a defect in energy expenditure following Mc3RLHA ablation, control and Mc3RLHA-ablated mice were subjected to indirect calorimetry to assess parameters of energy expenditure and nutrient usage. Mc3RLHA-ablated mice had a lower respiratory exchange ratio in the dark phase relative to controls, consistent with a shift toward fat utilization [Fig. 7(a)]. Energy expenditure normalized to body weight was lower in Mc3R-ablated mice [Fig. 7(b)]. To avoid confounding energy expenditure assessments by body weight normalization, we also analyzed absolute energy expenditure per animal as a function of body weight. Regression analysis of 24-hour energy expenditure as a function of body weight also showed a trend of lower energy expenditure in Mc3R-ablated mice [Fig. 7(c)]. Interestingly, mice with Mc3RLHA neuron ablation demonstrated decreased horizontal movement (X-total activity) [Fig. 7(d)]. These data reveal that ablating Mc3RLHA neurons decreases both locomotor activity and energy expenditure. Reductions in activity and energy expenditure would be predicted to enhance susceptibility to obesity when exposed to an HFD. Indeed, HFD-fed Mc3RLHA-ablated mice gained weight more quickly than did controls despite similar food intake at baseline and with refeeding after a fast (Fig. 8).

Figure 7.

Figure 7.

Comprehensive laboratory animal monitoring of Mc3RLHA-ablated mice. (a) Respiratory exchange ratio (RER) and (b) energy expenditure normalized to total body weight measured by indirect calorimetry. (c) Regression analysis of total 24-h energy expenditure as a function of body weight. (d) X-total activity. Average values ± SEM are shown (AAV-Control, n = 8; AAV-Casp, n = 6). *P < 0.05. NS, not significant.

Figure 8.

Figure 8.

Diet-induced obesity in Mc3RLHA-ablated and control animals. (a) Body weight change and (b) absolute body weight of Mc3rcre-Tomato mice injected with control virus or AAV-flex-taCasp3-TEVp virus. (c) Weekly HFD intake in Mc3rcre-Tomato mice injected with control virus or AAV-flex-taCasp3-TEVp virus. (b) High-fat food intake at 2, 4, and 8 h of refeeding after a fast. Average values ± SEM are shown (AAV-Control, n = 6; AAV-Casp, n = 6). *P < 0.05; **P < 0.01.

Discussion

To selectively access and manipulate Mc3R-expressing neurons, we generated a novel Mc3rcre transgenic line and used it in combination with viral vectors to probe the neurochemical composition, connectivity, and function of Mc3RLHA neurons. We show that Mc3RLHA neurons project to brain regions that regulate energy balance and express factors known to be involved in energy balance control. Indeed, acute activation of Mc3RLHA neurons is sufficient to promote locomotor activity and augment refeeding after a fast and this is associated with activation of OX and VTA neurons. Conversely, ablation of Mc3RLHA neurons results in decreased energy expenditure and activity, with increased body weight and fat mass, revealing that Mc3RLHA neurons are required for energy homeostasis. Although Mc3RLHA neurons are engaged by AgRP and POMC neurons anatomically [Fig. 1(f) and 1(g)], our studies do not test the role of Mc3R signaling in the LHA. We use Mc3R as a neuronal marker to genetically control the activity of the Mc3RLHA neuronal subset and evaluate its role in energy balance control. Mc3r and Mc4r mRNAs overlap somewhat in the LHA (Fig. 9) with ∼18% of Mc3RLHA neurons coexpressing Mc4r mRNA. Although MC4R signaling in the LHA contributes to glucose regulation and sympathetic output, it does not impact feeding (38). Whether the coexpression of Mc3r and Mc4r in the LHA has physiologic implications is not clear. Determining the role of LHA Mc3R signaling in energy balance control will require selective deletion or reactivation of Mc3R in the LHA.

Figure 9.

Figure 9.

Colocalization of Mc3r and Mc4r mRNA in the LHA. (a) RNAscope dual chromogen staining of Mc3r transcripts (blue) with Mc4r transcripts (red) in the LHA from Mc3Rcre mice. Two representative LHA images are shown. Scale bars, 20 μm. (b) Quantification of colocalization of Mc3r and Mc4r transcripts in the LHA (n = 2 mice).

The LHA is a heterogeneous structure that contains a number of genetically distinct cell populations involved in energy homeostasis. Early lesion studies identified the LHA as an important feeding center (64), and cell-specific manipulations within the LHA profoundly alter feeding behaviors (24, 27, 33, 65, 66). Activation of GABAergic LHA neurons induces voracious feeding and increases the rewarding value of food (65, 67). Our studies reveal that Mc3RLHA neurons are part of this GABAergic LHA population, express neuropeptides and receptors that regulate energy balance (Fig. 2), and project to multiple brain regions known to be involved in feeding control (Fig. 3). Direct chemogenetic activation of Mc3RLHA cells (which are largely GABAergic) can promote feeding, but this effect is relatively small and is only observed in the fasted state. Activation of GalLHA neurons using DREADD technology enhances the motivation to work for food but does not alter overall food consumption (33). Activation of NtsLHA neurons, alternatively, restrains feeding (68), suggesting that increased feeding following LHA GABAergic stimulation is not mediated by Nts. Thus subsets of Mc3RLHA neurons that do not express either galanin or Nts are likely responsible for the feeding effects following Mc3RLHA activation. Whether Mc3RLHA activation modifies the willingness to work for food or is associated with positive valence will require additional studies.

Our viral tracing studies reveal dense Mc3RLHA neuron projections to the BNST, VTA, PBN, and PVH (Table 1), brain regions associated with feeding, reward, locomotor activity, and energy expenditure. Selective activation of GABAergic LHA projections to the PVH stimulates feeding while suppressing grooming behaviors (69, 70). Our current studies do not determine the circuitry engaged by Mc3RLHA neurons to augment refeeding. Selective activation of Mc3RLHA terminals by optogenetics would be required to delineate the contribution of each projection pathway. It is of interest that Mc3RLHA neuron activation induces nuclear Fos accumulation in OX neurons in the LHA. Although central administration of OX stimulates feeding acutely (71–74), direct activation of OX neurons using chemogenetics has given mixed results, with one group demonstrating robust feeding (75) whereas another showed no effect on feeding (76). These different outcomes may reflect the total number and/or location of OX neurons activated. Whether OX feeding effects reflect modulation of satiety pathways or a more generalized augmentation of foraging behaviors is not clear. The contribution of OX neuron activity in the physiologic responses to Mc3RLHA activation remains to be determined and will be an important area of future investigation.

To determine the contribution of Mc3RLHA neurons to body weight homeostasis, we selectively ablated these neurons using site-specific injection of Cre-conditional viral vectors. Ablation of Mc3RLHA neurons was associated with more rapid weight gain with increased fat mass independent of changes in food intake. That Mc3RLHA neuron ablation had no effect on feeding or refeeding after a fast indicates that Mc3RLHA neurons are not required for normal feeding control. However, the observed increase in fat mass independent of food intake suggested a defect in energy expenditure. Indeed, indirect calorimetry of Mc3RLHA neuron-ablated mice revealed differences in energy expenditure relative to controls either normalized to total body weight or plotted as a function of body weight. Consistent with an underlying energy expenditure defect, Mc3RLHA neuron-ablated mice challenged with an HFD gained fat mass more rapidly than did controls with matched food intake. In contrast to these findings, a study by Begriche et al. (16) showed that the peripheral Mc3R, instead of central Mc3R, is important in diet-induced obesity. Again, it is important to recognize that our approach ablates the entire Mc3R-expressing neuron rather than just removing the Mc3R. Moreover, our manipulations were performed in adult animals and sidestepped issues of developmental compensation that may occur with germline gene deletion studies. Given the alterations in energy expenditure after Mc3RLHA neuron ablation, we did expect a more pronounced weight gain with an HFD. The relatively modest weight gain seen in Mc3RLHA- ablated animals following HFD exposure may reflect the redundancy of both central and peripheral systems (including peripheral Mc3R actions) that contribute to energy utilization and weight maintenance in the face of caloric challenge.

Ablation of Mc3RLHA neurons is associated with decreased locomotor activity, whereas chemogenetic activation of Mc3RLHA neurons drives locomotor activity. Alterations in locomotor activity following LHA cell manipulations have been previously reported. Activation of GABAergic-, Nts-, and galanin-expressing LHA neurons each alter locomotor activity. As highlighted by Qualls-Creekmore et al. (33), however, LHA cell-specific alterations in locomotor activity are not qualitatively equivalent. GABAergic LHA activation is associated with repetitive or compulsive behaviors, whereas activation of GalLHA neurons is associated with alterations across a range of behaviors that are not compulsive. A detailed characterization of the induced locomotor activity changes following Mc3RLHA neuron activation will be necessary to determine whether these locomotor activity changes can be ascribed to defined subsets of Mc3RLHA cells (e.g., galanin- vs Nts-expressing cells).

Chemogenetic activation of Mc3RLHA neurons is associated with changes in Fos expression in OX neurons and neurons in the VTA, implicating these cells and brain region as possible effectors of Mc3RLHA actions. As Mc3RLHA neurons are predominantly GABAergic, the induction of Fos expression following Mc3RLHA activation is likely to be indirect or mediated by other secreted factors. Immunohistochemical analysis of Mc3RLHA neurons revealed colocalization with Nts, a neuromodulator that directly engages the dopamine system (77). Increased locomotor activity following Mc3RLHA neuron activation was blunted by pretreatment with dopamine antagonists (Fig. 5), suggesting that Mc3RLHA neurons might regulate the mesolimbic dopamine system through Nts release. NtsLHA neurons project to the VTA, and Nts release in the VTA promotes physical activity (27, 30, 59). That a subset of Mc3RLHA neurons express Nts [Fig. 2(c)] and that Mc3RLHA neurons project to the VTA [Fig. 3(e)] suggest potential molecular and neuroanatomical mechanisms by which Mc3RLHA neurons may regulate activity and link melanocortin action to the mesolimbic dopamine system. Importantly, note that a subset of Mc3RLHA neurons expresses galanin and that GalLHA neurons modulate locomotor activity without directly targeting the VTA (33, 34). Thus, the changes in locomotor activity seen upon Mc3RLHA activation may be a composite of effects driven through both NtsLHA and GalLHA pathways, which target the VTA and other brain regions. Future studies designed to selectively delete Nts (or galanin) from Mc3R-expressing neurons are needed to definitively test these hypotheses.

In summary, Mc3RLHA neurons play a crucial role in the regulation of energy expenditure and locomotor activity. Our studies demonstrate the necessity of Mc3RLHA neurons in energy homeostasis, and they provide insight into cellular and anatomical pathways by which the LHA regulates feeding, energy expenditure, locomotor activity, and fat mass.

Acknowledgments

We thank members of the Olson and Myers laboratories for helpful discussions.

Financial Support: This work was supported by the Michigan Medicine Department of Pediatrics, the Woodson Accelerator Award in Pediatrics (to D.P.O.), and by the Whitehall Foundation (to D.P.O.).

Disclosure Summary: The authors have nothing to disclose.

Glossary

Abbreviations:

AAV

adeno-associated virus

AgRP

agouti-related protein

ARC

arcuate nucleus

BNST

bed nucleus of the stria terminalis

CNO

clozapine N-oxide

CNS

central nervous system

DAB

diaminobenzidine

DREADD

designer receptors exclusively activated by designer drug

GABA

γ-aminobutyric acid

GalLHA

galanin cells in the lateral hypothalamic area

HFD

high-fat diet

IHC

immunohistochemistry

LepRb

leptin receptor

LHA

lateral hypothalamic area

MCH

melanin-concentrating hormone

Mc3R

melanocortin-3 receptor

Mc4R

melanocortin-4 receptor

Nts

neurotensin

OX

orexin

PBN

parabrachial nuclei

POMC

proopiomelanocortin

p-Stat3

phosphorylated signal transducer and activator of transcription-3

PVH

paraventricular nucleus of the hypothalamus

VMH

ventromedial nucleus of the hypothalamus

VTA

ventral tegmental area

References

  • 1. Pi-Sunyer X. The medical risks of obesity. Postgrad Med. 2009;121(6):21–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Garfield AS, Lam DD, Marston OJ, Przydzial MJ, Heisler LK. Role of central melanocortin pathways in energy homeostasis. Trends Endocrinol Metab. 2009;20(5):203–215. [DOI] [PubMed] [Google Scholar]
  • 3. Cone RD. Studies on the physiological functions of the melanocortin system. Endocr Rev. 2006;27(7):736–749. [DOI] [PubMed] [Google Scholar]
  • 4. Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell. 1997;88(1):131–141. [DOI] [PubMed] [Google Scholar]
  • 5. Vaisse C, Clement K, Guy-Grand B, Froguel P. A frameshift mutation in human MC4R is associated with a dominant form of obesity. Nat Genet. 1998;20(2):113–114. [DOI] [PubMed] [Google Scholar]
  • 6. Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG, O’Rahilly S. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet. 1998;20(2):111–112. [DOI] [PubMed] [Google Scholar]
  • 7. Roselli-Rehfuss L, Mountjoy KG, Robbins LS, Mortrud MT, Low MJ, Tatro JB, Entwistle ML, Simerly RB, Cone RD. Identification of a receptor for gamma melanotropin and other proopiomelanocortin peptides in the hypothalamus and limbic system. Proc Natl Acad Sci USA. 1993;90(19):8856–8860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Gantz I, Konda Y, Tashiro T, Shimoto Y, Miwa H, Munzert G, Watson SJ, DelValle J, Yamada T. Molecular cloning of a novel melanocortin receptor. J Biol Chem. 1993;268(11):8246–8250. [PubMed] [Google Scholar]
  • 9. Feng N, Young SF, Aguilera G, Puricelli E, Adler-Wailes DC, Sebring NG, Yanovski JA. Co-occurrence of two partially inactivating polymorphisms of MC3R is associated with pediatric-onset obesity. Diabetes. 2005;54(9):2663–2667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Savastano DM, Tanofsky-Kraff M, Han JC, Ning C, Sorg RA, Roza CA, Wolkoff LE, Anandalingam K, Jefferson-George KS, Figueroa RE, Sanford EL, Brady S, Kozlosky M, Schoeller DA, Yanovski JA. Energy intake and energy expenditure among children with polymorphisms of the melanocortin-3 receptor. Am J Clin Nutr. 2009;90(4):912–920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Zegers D, Beckers S, de Freitas F, Peeters AV, Mertens IL, Verhulst SL, Rooman RP, Timmermans JP, Desager KN, Massa G, Van Gaal LF, Van Hul W. Identification of three novel genetic variants in the melanocortin-3 receptor of obese children. Obesity (Silver Spring). 2011;19(1):152–159. [DOI] [PubMed] [Google Scholar]
  • 12. Butler AA, Kesterson RA, Khong K, Cullen MJ, Pelleymounter MA, Dekoning J, Baetscher M, Cone RD. A unique metabolic syndrome causes obesity in the melanocortin-3 receptor-deficient mouse. Endocrinology. 2000;141(9):3518–3521. [DOI] [PubMed] [Google Scholar]
  • 13. Chen AS, Marsh DJ, Trumbauer ME, Frazier EG, Guan XM, Yu H, Rosenblum CI, Vongs A, Feng Y, Cao L, Metzger JM, Strack AM, Camacho RE, Mellin TN, Nunes CN, Min W, Fisher J, Gopal-Truter S, MacIntyre DE, Chen HY, Van der Ploeg LH. Inactivation of the mouse melanocortin-3 receptor results in increased fat mass and reduced lean body mass. Nat Genet. 2000;26(1):97–102. [DOI] [PubMed] [Google Scholar]
  • 14. Sutton GM, Trevaskis JL, Hulver MW, McMillan RP, Markward NJ, Babin MJ, Meyer EA, Butler AA. Diet-genotype interactions in the development of the obese, insulin-resistant phenotype of C57BL/6J mice lacking melanocortin-3 or -4 receptors. Endocrinology. 2006;147(5):2183–2196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Ellacott KL, Murphy JG, Marks DL, Cone RD. Obesity-induced inflammation in white adipose tissue is attenuated by loss of melanocortin-3 receptor signaling. Endocrinology. 2007;148(12):6186–6194. [DOI] [PubMed] [Google Scholar]
  • 16. Begriche K, Levasseur PR, Zhang J, Rossi J, Skorupa D, Solt LA, Young B, Burris TP, Marks DL, Mynatt RL, Butler AA. Genetic dissection of the functions of the melanocortin-3 receptor, a seven-transmembrane G-protein-coupled receptor, suggests roles for central and peripheral receptors in energy homeostasis. J Biol Chem. 2011;286(47):40771–40781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Sutton GM, Perez-Tilve D, Nogueiras R, Fang J, Kim JK, Cone RD, Gimble JM, Tschöp MH, Butler AA. The melanocortin-3 receptor is required for entrainment to meal intake. J Neurosci. 2008;28(48):12946–12955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Bernardis LL, Bellinger LL. The lateral hypothalamic area revisited: neuroanatomy, body weight regulation, neuroendocrinology and metabolism. Neurosci Biobehav Rev. 1993;17(2):141–193. [DOI] [PubMed] [Google Scholar]
  • 19. Delgado JM, Anand BK. Increase of food intake induced by electrical stimulation of the lateral hypothalamus. Am J Physiol. 1953;172(1):162–168. [DOI] [PubMed] [Google Scholar]
  • 20. Stanley BG, Ha LH, Spears LC, Dee MG II. Lateral hypothalamic injections of glutamate, kainic acid, d,l-α-amino-3-hydroxy-5-methyl-isoxazole propionic acid or N-methyl-d-aspartic acid rapidly elicit intense transient eating in rats. Brain Res. 1993;613(1):88–95. [DOI] [PubMed] [Google Scholar]
  • 21. Stanley BG, Willett VL III, Donias HW, Ha LH, Spears LC. The lateral hypothalamus: a primary site mediating excitatory amino acid-elicited eating. Brain Res. 1993;630(1–2):41–49. [DOI] [PubMed] [Google Scholar]
  • 22. Stuber GD, Wise RA. Lateral hypothalamic circuits for feeding and reward. Nat Neurosci. 2016;19(2):198–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Stamatakis AM, Van Swieten M, Basiri ML, Blair GA, Kantak P, Stuber GD. Lateral hypothalamic area glutamatergic neurons and their projections to the lateral habenula regulate feeding and reward. J Neurosci. 2016;36(2):302–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Jennings JH, Ung RL, Resendez SL, Stamatakis AM, Taylor JG, Huang J, Veleta K, Kantak PA, Aita M, Shilling-Scrivo K, Ramakrishnan C, Deisseroth K, Otte S, Stuber GD. Visualizing hypothalamic network dynamics for appetitive and consummatory behaviors. Cell. 2015;160(3):516–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Brown JA, Woodworth HL, Leinninger GM. To ingest or rest? Specialized roles of lateral hypothalamic area neurons in coordinating energy balance. Front Syst Neurosci. 2015;9:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Shimada M, Tritos NA, Lowell BB, Flier JS, Maratos-Flier E. Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature. 1998;396(6712):670–674. [DOI] [PubMed] [Google Scholar]
  • 27. Leinninger GM, Opland DM, Jo YH, Faouzi M, Christensen L, Cappellucci LA, Rhodes CJ, Gnegy ME, Becker JB, Pothos EN, Seasholtz AF, Thompson RC, Myers MG Jr. Leptin action via neurotensin neurons controls orexin, the mesolimbic dopamine system and energy balance. Cell Metab. 2011;14(3):313–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kempadoo KA, Tourino C, Cho SL, Magnani F, Leinninger GM, Stuber GD, Zhang F, Myers MG, Deisseroth K, de Lecea L, Bonci A. Hypothalamic neurotensin projections promote reward by enhancing glutamate transmission in the VTA. J Neurosci. 2013;33(18):7618–7626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Patterson CM, Wong JM, Leinninger GM, Allison MB, Mabrouk OS, Kasper CL, Gonzalez IE, Mackenzie A, Jones JC, Kennedy RT, Myers MG Jr. Ventral tegmental area neurotensin signaling links the lateral hypothalamus to locomotor activity and striatal dopamine efflux in male mice. Endocrinology. 2015;156(5):1692–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Opland D, Sutton A, Woodworth H, Brown J, Bugescu R, Garcia A, Christensen L, Rhodes C, Myers M Jr, Leinninger G. Loss of neurotensin receptor-1 disrupts the control of the mesolimbic dopamine system by leptin and promotes hedonic feeding and obesity. Mol Metab. 2013;2(4):423–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Goforth PB, Leinninger GM, Patterson CM, Satin LS, Myers MG Jr. Leptin acts via lateral hypothalamic area neurotensin neurons to inhibit orexin neurons by multiple GABA-independent mechanisms. J Neurosci. 2014;34(34):11405–11415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Laque A, Zhang Y, Gettys S, Nguyen TA, Bui K, Morrison CD, Münzberg H. Leptin receptor neurons in the mouse hypothalamus are colocalized with the neuropeptide galanin and mediate anorexigenic leptin action. Am J Physiol Endocrinol Metab. 2013;304(9):E999–E1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Qualls-Creekmore E, Yu S, Francois M, Hoang J, Huesing C, Bruce-Keller A, Burk D, Berthoud HR, Morrison CD, Münzberg H. Galanin-expressing GABA neurons in the lateral hypothalamus modulate food reward and noncompulsive locomotion. J Neurosci. 2017;37(25):6053–6065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Laque A, Yu S, Qualls-Creekmore E, Gettys S, Schwartzenburg C, Bui K, Rhodes C, Berthoud HR, Morrison CD, Richards BK, Münzberg H. Leptin modulates nutrient reward via inhibitory galanin action on orexin neurons. Mol Metab. 2015;4(10):706–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Wang D, He X, Zhao Z, Feng Q, Lin R, Sun Y, Ding T, Xu F, Luo M, Zhan C. Whole-brain mapping of the direct inputs and axonal projections of POMC and AgRP neurons. Front Neuroanat. 2015;9:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kas MJ, Tiesjema B, van Dijk G, Garner KM, Barsh GS, ter Brake O, Verhaagen J, Adan RA. Induction of brain-region-specific forms of obesity by agouti. J Neurosci. 2004;24(45):10176–10181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Betley JN, Cao ZF, Ritola KD, Sternson SM. Parallel, redundant circuit organization for homeostatic control of feeding behavior. Cell. 2013;155(6):1337–1350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Morgan DA, McDaniel LN, Yin T, Khan M, Jiang J, Acevedo MR, Walsh SA, Ponto LL, Norris AW, Lutter M, Rahmouni K, Cui H. Regulation of glucose tolerance and sympathetic activity by MC4R signaling in the lateral hypothalamus. Diabetes. 2015;64(6):1976–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Garfield AS, Li C, Madara JC, Shah BP, Webber E, Steger JS, Campbell JN, Gavrilova O, Lee CE, Olson DP, Elmquist JK, Tannous BA, Krashes MJ, Lowell BB. A neural basis for melanocortin-4 receptor-regulated appetite. Nat Neurosci. 2015;18(6):863–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Abe H, Yanagawa Y, Kanbara K, Maemura K, Hayasaki H, Azuma H, Obata K, Katsuoka Y, Yabumoto M, Watanabe M. Epithelial localization of green fluorescent protein-positive cells in epididymis of the GAD67-GFP knock-in mouse. J Androl. 2005;26(5):568–577. [DOI] [PubMed] [Google Scholar]
  • 41.RRID:AB_2106755.
  • 42.RRID:AB_2340593.
  • 43.RRID:AB_10013483.
  • 44.RRID:AB_2534017.
  • 45.RRID:AB_221570.
  • 46.RRID:AB_2155582.
  • 47.RRID:AB_653611.
  • 48.RRID:AB_2237276.
  • 49.RRID:AB_141708.
  • 50.RRID:AB_2534102.
  • 51.RRID:AB_2491009.
  • 52. Yang CF, Chiang MC, Gray DC, Prabhakaran M, Alvarado M, Juntti SA, Unger EK, Wells JA, Shah NM. Sexually dimorphic neurons in the ventromedial hypothalamus govern mating in both sexes and aggression in males. Cell. 2013;153(4):896–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Qualls-Creekmore E, Münzberg H. Modulation of feeding and associated behaviors by lateral hypothalamic circuits. Endocrinology. 2018;159(11):3631–3642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Sutton AK, Pei H, Burnett KH, Myers MG Jr, Rhodes CJ, Olson DP. Control of food intake and energy expenditure by Nos1 neurons of the paraventricular hypothalamus. J Neurosci. 2014;34(46):15306–15318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Swanson LW, Sawchenko PE. Paraventricular nucleus: a site for the integration of neuroendocrine and autonomic mechanisms. Neuroendocrinology. 1980;31(6):410–417. [DOI] [PubMed] [Google Scholar]
  • 56. Dumont EC, Mark GP, Mader S, Williams JT. Self-administration enhances excitatory synaptic transmission in the bed nucleus of the stria terminalis. Nat Neurosci. 2005;8(4):413–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Russo SJ, Nestler EJ. The brain reward circuitry in mood disorders [published correction appears in Nat Rev Neurosci. 2013;14(10):736]. Nat Rev Neurosci. 2013;14(9):609–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Borgland SL, Taha SA, Sarti F, Fields HL, Bonci A. Orexin A in the VTA is critical for the induction of synaptic plasticity and behavioral sensitization to cocaine. Neuron. 2006;49(4):589–601. [DOI] [PubMed] [Google Scholar]
  • 59. Elliott PJ, Nemeroff CB. Repeated neurotensin administration in the ventral tegmental area: effects on baseline and d-amphetamine-induced locomotor activity. Neurosci Lett. 1986;68(2):239–244. [DOI] [PubMed] [Google Scholar]
  • 60. Alexander GM, Rogan SC, Abbas AI, Armbruster BN, Pei Y, Allen JA, Nonneman RJ, Hartmann J, Moy SS, Nicolelis MA, McNamara JO, Roth BL. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron. 2009;63(1):27–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Nakamura T, Uramura K, Nambu T, Yada T, Goto K, Yanagisawa M, Sakurai T. Orexin-induced hyperlocomotion and stereotypy are mediated by the dopaminergic system. Brain Res. 2000;873(1):181–187. [DOI] [PubMed] [Google Scholar]
  • 62. Sakurai T. The role of orexin in motivated behaviours [published correction appears in Nat Rev Neurosci. 2014;15:816]. Nat Rev Neurosci. 2014;15(11):719–731. [DOI] [PubMed] [Google Scholar]
  • 63. Pijnenburg AJ, Honig WM, Van der Heyden JA, Van Rossum JM. Effects of chemical stimulation of the mesolimbic dopamine system upon locomotor activity. Eur J Pharmacol. 1976;35(1):45–58. [DOI] [PubMed] [Google Scholar]
  • 64. Anand BK, Brobeck JR. Localization of a “feeding center” in the hypothalamus of the rat. Proc Soc Exp Biol Med. 1951;77(2):323–324. [DOI] [PubMed] [Google Scholar]
  • 65. Nieh EH, Matthews GA, Allsop SA, Presbrey KN, Leppla CA, Wichmann R, Neve R, Wildes CP, Tye KM. Decoding neural circuits that control compulsive sucrose seeking. Cell. 2015;160(3):528–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Kosse C, Schöne C, Bracey E, Burdakov D. Orexin-driven GAD65 network of the lateral hypothalamus sets physical activity in mice. Proc Natl Acad Sci USA. 2017;114(17):4525–4530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Jennings JH, Rizzi G, Stamatakis AM, Ung RL, Stuber GD. The inhibitory circuit architecture of the lateral hypothalamus orchestrates feeding. Science. 2013;341(6153):1517–1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Woodworth HL, Beekly BG, Batchelor HM, Bugescu R, Perez-Bonilla P, Schroeder LE, Leinninger GM. Lateral hypothalamic neurotensin neurons orchestrate dual weight loss behaviors via distinct mechanisms. Cell Reports. 2017;21(11):3116–3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Wu Z, Kim ER, Sun H, Xu Y, Mangieri LR, Li DP, Pan HL, Xu Y, Arenkiel BR, Tong Q. GABAergic projections from lateral hypothalamus to paraventricular hypothalamic nucleus promote feeding. J Neurosci. 2015;35(8):3312–3318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Mangieri LR, Lu Y, Xu Y, Cassidy RM, Xu Y, Arenkiel BR, Tong Q. A neural basis for antagonistic control of feeding and compulsive behaviors. Nat Commun. 2018;9(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Shiraishi T, Oomura Y, Sasaki K, Wayner MJ. Effects of leptin and orexin-A on food intake and feeding related hypothalamic neurons. Physiol Behav. 2000;71(3-4):251–261. [DOI] [PubMed] [Google Scholar]
  • 72. Dube MG, Kalra SP, Kalra PS. Food intake elicited by central administration of orexins/hypocretins: identification of hypothalamic sites of action. Brain Res. 1999;842(2):473–477. [DOI] [PubMed] [Google Scholar]
  • 73. Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richardson JA, Kozlowski GP, Wilson S, Arch JR, Buckingham RE, Haynes AC, Carr SA, Annan RS, McNulty DE, Liu WS, Terrett JA, Elshourbagy NA, Bergsma DJ, Yanagisawa M. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(4):573–585. [DOI] [PubMed] [Google Scholar]
  • 74. Willie JT, Chemelli RM, Sinton CM, Yanagisawa M. To eat or to sleep? Orexin in the regulation of feeding and wakefulness. Annu Rev Neurosci. 2001;24(1):429–458. [DOI] [PubMed] [Google Scholar]
  • 75. Inutsuka A, Inui A, Tabuchi S, Tsunematsu T, Lazarus M, Yamanaka A. Concurrent and robust regulation of feeding behaviors and metabolism by orexin neurons. Neuropharmacology. 2014;85:451–460. [DOI] [PubMed] [Google Scholar]
  • 76. Zink AN, Bunney PE, Holm AA, Billington CJ, Kotz CM. Neuromodulation of orexin neurons reduces diet-induced adiposity. Int J Obes. 2018;42(4):737–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Binder EB, Kinkead B, Owens MJ, Nemeroff CB. Neurotensin and dopamine interactions. Pharmacol Rev. 2001;53(4):453–486. [PubMed] [Google Scholar]

Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES