Abstract
Obesity and its associated complications have reached epidemic proportions in the US and worldwide, highlighting the need for new and more effective treatments. While the neuropeptide oxytocin (OXT) is well recognized for its peripheral effects on reproductive behavior, release of OXT from somatodendrites and axonal terminals within the central nervous system (CNS) is also implicated in the control of control of energy balance. In this review we summarize historical data highlighting the effects of exogenous OXT as a short-term regulator of food intake in a context specific manner and the receptor populations that may mediate these effects. We also describe what is known about the physiological role of endogenous OXT in the control of energy balance and whether serum and brain levels of OXT relate to obesity on a consistent basis across animal models and humans with obesity. We describe recent data on the effectiveness of chronic CNS administration of OXT to decrease food intake and weight gain or to elicit weight loss in diet-induced obese (DIO) and genetically obese mice and rats. Of clinical importance is the finding that both chronic central and peripheral OXT treatment evoke weight loss in obese animal models with impaired leptin signaling at doses that are not associated with visceral illness, tachyphylaxis, or adverse cardiovascular effects. Moreover, these results have been largely recapitulated following chronic subcutaneous or intranasal treatment in DIO nonhuman primates (rhesus monkeys) and obese humans, respectively. We also identify plausible mechanisms that contribute to the effects of OXT on body weight and glucose homeostasis in rodents, nonhuman primates, and humans. We conclude by describing ongoing challenges that remain in order for OXT-based therapeutics to be used as a long-term strategy to treat obesity in humans.
Keywords: Oxytocin, food intake, energy expenditure, body weight, glucose homeostasis
1. INTRODUCTION
OXT is expressed in neurons located in both parvocellular and magnocellular divisions of the paraventricular nucleus (PVN) as well as in the magnocellular division of the supraoptic nucleus (SON) in rats (77, 151, 189, 196, 197, 230, 231), mole-rats (192), mice (28, 192, 220, 228), hamsters (208), nonhuman primates (3, 55, 86, 184, 218) and humans (35, 53, 97, 167, 227). OXT is also expressed to a lesser extent in magnocellular neurons in the preoptic area (mole-rat) (192), anterior hypothalamus (mole-rat) (192), anterior commissural nuclei (mouse) (28), accessory nuclei (mouse/rat) (28, 189), and periventricular nuclei (mouse) (28). In addition, it is found in neurons (undetermined if magnocellular or parvocellular) of other forebrain areas that include the dorsal hypothalamic area (hamsters) (208), bed nucleus of the stria terminalis (BNST; mole-rat/rat) (189, 192), mediobasal preoptic area (mouse) (28), medial amygdala (mole-rat/rat) (189, 192), septal region (rat) (189) and caudal subzona incerta (hamsters) (248).
Mature OXT and the carrier neurophysin are derived from cleavage and modification of the OXT/neurophysin 1 prepropeptide during axonal transport (24). Both are stored in the axon terminals prior to release (187). The predominant function of neurophysin appears to be to target, package and store OXT within secretory granules in preparation for release (for review see (54)). OXT is released distally through axon terminals within the arcuate nucleus (ARC) (123), ventral tegmental area (VTA) (207), parabrachial nucleus (PBN) (193), nucleus of the solitary tract (NTS) (189, 196), dorsal motor nucleus of the vagus (189, 196), and spinal cord (196), and it is also released locally from somatodendrites within the hypothalamus (SON and PVN). In addition to local release from the PVN and SON (180, 260, 275, 276) (for review see (117)), the extent to which it may be released in other areas with more limited expression is not clear.
OXTR receptors (OXTR) are widely distributed and largely overlap in areas linked to the control of energy balance in both mice and rats, namely the basal ganglia [e.g. nucleus accumbens (NAcc) and central amygdala (mouse (66, 193, 269)/rat (240, 243, 246), hypothalamus [ARC, MPA, and ventromedial hypothalamus (VMH)/mouse (40, 56, 66, 193, 269)/rat (95, 243, 246)], midbrain VTA [mouse (170)/rat (243)], hindbrain PBN [mouse (193)], AP [mice (56, 193, 269)], DMV [rat (240, 243, 251), NTS [mouse (56, 269)/rat (11, 161, 162, 251)] and spinal cord [mouse (257)/rat (186)]. OXTR are also found in the SON and PVN (270) of OXT neurons in the rat (45) but it is unclear if this is the case in the mouse. Unlike the rodent model, there appears to be a more limited distribution of OXTRs in CNS areas linked to the control of energy balance in nonhuman primates [NAcc, preoptic area, VMH, DMV, and spinal cord) (20, 42, 200)] and humans (central amygdala, anterior hypothalamus, MPA, PVN, VMH, AP, NTS and spinal cord) (21, 114, 115). The overlapping patterns of expression in the basal ganglia, hypothalamus, hindbrain and spinal cord suggest potentially important roles of nuclei within these areas in controlling body weight (BW) that are well conserved across species. Less is known about the source of OXT that reaches OXTR that are expressed in brain sites that have little to no OXT fiber innervation (VMH, amygdala), but OXT has been proposed to reach these receptors by diffusion [reviewed in (250, 251)] from magnocellular neurons in the SON (reviewed in (109, 195).
In this review, we focus on the role of OXT in the control of energy balance based on data from rodents, nonhuman primates and humans. We highlight its well-characterized effects in the short-term control of food intake and the receptor populations that may mediate these effects. We also describe what is known about the role of endogenous OXT signaling in the control of energy balance and whether circulating and central levels of OXT reflect the state of obesity in animal models and humans. In addition, we 1) summarize recent data on the effectiveness of chronic CNS administration of OXT to decrease food intake and weight gain or elicit weight loss in diet-induced obese (DIO) and genetically obese rodents, nonhuman primates and humans, 2) identify potential mechanisms that may mediate these effects, and 3) discuss ongoing challenges that remain in order for OXT-based therapeutics to be used as a long-term strategy to treat obesity in humans.
2. Oxytocin and short-term control of food intake: key findings from laboratory rodent studies
Extensive evidence derived from animal studies focusing on short-term regulation of meal size indicates that OXT terminates feeding by affecting various facets of physiological responses that collectively contribute to cessation of eating behavior. This role arises from the strategic localization of OXT neurons in neural and neuroendocrine pathways that mediate, among others, gut-brain interactions, hormonal changes, and reward processing.
2.1. Receptor populations thought to contribute to the effects of OXT on short-term food and fluid intake
Well before the first reports on acute anorexigenic properties of OXT were published, it had been established that lesions of the hypothalamic PVN where the OXT cells are located and the disruption of the hindbrain-PVN connectivity (along with severing OXT fibers between the PVN and caudal brain stem) (90), promote excessive feeding and BW gain in rats (107, 209, 211). Those studies served as the cornerstone of the initial experiments examining the effects of OXT injections on food consumption. In 1989 and 1990, Arletti and colleagues showed in a comprehensive manner that intracerebroventricular (ICV) administration of OXT in rats caused short-lived hypophagia reversible by an ICV OXTR antagonist (d(CH2)5Tyr(Me)2-[Orn8]-vasotocin) (4, 5). The anorexigenic effects were also recapitulated after peripheral (intraperitoneal; IP) injections. OXT-treated freely-feeding animals reduced intake of standard laboratory chow by approximately 60% (IP OXT) and 40% (ICV OXT). OXT lowered chow intake also in schedule-fed rats given food for 3 hours per day. Latency to begin a meal was increased, whereas meal duration, decreased, by the treatment. These findings were corroborated by Olson who found that lateral ventricular OXT and OXTR agonist [e-L-beta-MePhe2]OXT reduced deprivation-induced chow consumption and time spent feeding in rats accustomed to receiving a 1-hour meal (152). Over the years, OXT has been found to be effective in generating early termination of food intake when administered subcutaneously (sc), intravenously (IV), intranasally (IN) and into the lateral, third (3V) and fourth ventricle (4V; as a strategy to more specifically target hindbrain OXTRs). Considering the debate over whether peripherally administered OXT crosses the blood-brain barrier and enters the brain in rodents (67, 76, 130, 144, 190, 214, 236), pigs (185), sheep (87), nonhuman primates (29, 33, 44, 106, 134) and humans (108, 223), both the central and peripheral pools of the OXT receptor have been implicated in mediating OXT-driven hypophagia (16, 34, 67, 75, 76, 93–95, 105, 121, 138, 147, 161, 162, 189, 191, 275–277). In addition, it has been suggested that peripheral OXT may also act, in part, via hindbrain OXTRs: 4V pretreatment with an OXTR antagonist attenuated the effects of IP OXT on short-term food intake in rats (67). Based on 1) the findings by Ho (67), Iwasaki (75, 76), Wu (258, 259), and Liu (112), and 2) reports indicating that OXTRs are expressed in the nodose ganglion (255) and GI tract (183), peripheral OXT might also promote hypophagia through these peripheral OXTR populations. Importantly, as shown in hunger discrimination studies utilizing operant methodology, while OXT is effective in producing early termination of a meal that has already begun, it does not reduce the feeling of hunger (61).
Some (4, 5) but not all (e.g. (93, 138, 152)) studies have shown a reduction in water intake in animals treated with OXT, but this peptide’s effect on food consumption is thought not to arise from changes in drinking behavior. Recent data suggest that only discrete subpopulations of the OXTR-expressing neurons, such as those located in the PBN, promote changes in water intake (193).
2.2. Use of OXTR antagonists as a tool to examine the physiological role of OXT in the control of short-term food intake
While the vast majority of studies have shown reversibility of OXT’s effects on feeding with OXTR antagonists, there is no consensus as to whether OXTR blockade by itself increases consumption. For example, one study found that lateral ventricular administration of the antagonist [(CH2)51, Phe(Me)2, Thr4, Orn8] OXT did not potentiate deprivation-induced feeding of chow in rats (152), Olszewski reported no effect of an IP BBB-penetrant antagonist, L-368,899, on chow intake in mice (156), and Klockars reported no change in chow consumption after direct intraparenchymal injection of L-368,899 (94). On the other hand, it was reported that both IP and ICV administration of the OXTR antagonist, d(CH2)5Tyr(Me)2-Orn8]-vasotocin, increased food intake and reduced latency to begin a meal in schedule-fed rats (4, 5). Others (11, 17) found that 3V administration of the OXTR antagonists, [d(CH2)5,Tyr(Me)2,Orn8]-vasotocin and [d(CH2)51,Tyr(Me)2,Orn8]-oxytocin, respectively, stimulated chow consumption in rats. An additional paper reported that 4V injection of the antagonist stimulated chow intake, and, in contrast to some of the previous studies following 3V administration, found that 3V administration failed to stimulate chow consumption in 6-h fasted rats (17), indicating that mixed results have been obtained following 3V administration of OXTR antagonists in rat models. Consistent with the effects following 4V administration in rats, Blouet and Schwartz extended these findings and found that 4V administration of the OXTR antagonist, [d(CH2)5,Tyr(Me)2,Orn8]-vasotocin, stimulated chow consumption in mice through an increase in meal size and a decrease in meal latency (19). However, considering the two studies in which ICV administration of an OTR antagonist stimulated food intake in rats that were deprived for 21 h (4, 5), Baskin also found that 3V administration of the antagonist in 16-h fasted rats, produced hyperphagia (17). It is not clear at this point if differences in timing of administration may account for these differences, as a more potent effect on daytime chow intake was found when [d(CH2)5,Tyr(Me)2,Orn8]-vasotocin was administered into the 3V at the start of the light cycle in mice (276). While the effects of 3V and 4V OXTR blockade on food intake are largely consistent between rats and mice, differences with respect to length of fast, strain, or OXTR expression may explain, in part, why the OXTR antagonist may work more optimally under some conditions (276) but not others.
In the early stages of research on OXT and feeding, concerns were raised that OXT-driven hypophagia might be only a pharmacological phenomenon secondary to select homeostatic functions of the hormone. Indeed, intravenous infusion of hypertonic saline is known to increase OXT release in rats and humans (41, 221) and, simultaneously, inhibit feeding in rats (41). Plasma toxicity that leads to termination of food intake and underlies development of conditioned taste aversion (CTA) increases the number of c-Fos (marker of neuronal activation) positive OXT neurons (68, 149, 154, 155, 238). OXTR blockade prevents CTA acquisition (159, 263, 264) and drugs that block OXT neuronal activation oftentimes alleviate aversions (158, 256), although central or peripheral administration of OXT at anorexigenic doses does not cause pica or CTA (76, 147, 275) (behavioral readouts of visceral illness in rodent models). Furthermore, OXT is rapidly released into the periphery in response to electrical stimulation of gastric afferents (242) or a large degree of stomach distension regardless of energy load (143, 188). Overall the data suggest that, while one can view OXT as being involved in protecting internal milieu in a manner completely independent from feeding, in reality it acts as an integral component of homeostatic mechanisms typically triggered by adverse effects stemming from ingestive behavior (excessive stomach distension, sodium imbalance, and toxicity).
2.3. What have we learned about the role of OXT neurons in the control of short-term food intake?
What instills confidence in defining OXT as an anorexigen is that the activity of the OXT system changes in a hunger-satiation continuum and that there is a functional link between OXT and other neural and endocrine feeding regulators. Plasma OXT levels are elevated at satiation-driven end of a meal in female (ovariectomized) and male rats (116, 252). An increase in the percentage of c-Fos immunoreactive PVN and SON OXT neurons has been detected in mice and rats that have eaten a satiating amount of a calorie-dense chow or calorie-dilute liquids compared to animals anticipating presentation of a meal (79, 156, 265). In contrast, recent studies in mice using optical fiber photometry indicate that chow diet failed to alter activity of PVN OXT neurons in mice (50) raising the possibility that macronutrient composition of the diet may be important in activation of PVN OXT neurons. Consistent with this, sated rats exhibit higher hypothalamic OXT gene expression than deprived counterparts (98, 169, 241), and OXTR mRNA levels are modified by energy deprivation as well as by macronutrient composition of food [e.g. (94, 95, 156)]. Intragastric administration of nutrients that promote satiety (e.g., select amino acids), also triggers OXT gene expression and OXT neuronal activation (51) in mice. Importantly, OXT neurons or terminals co-express other anorexigens, including corticotropin-releasing hormone (CRH) (9), nesfatin-1 (96, 124) and CCK (127), and OXT appears to contribute to and/or potentiate the hypophagic action of nesfatin-1, CCK, and leucine (15, 18, 19, 124, 153) in rats and mice. OXT neuronal activation is modified by feeding regulatory peptides either directly (via synapses formed with OXT cells) or indirectly (as an element of broader circuits). For example, terminals containing cocaine-amphetamine related transcript (CART) are found in close apposition to magnocellular and parvocellular PVN OXT neurons (253) and central administration of CART increases c-Fos in both magnocellular and parvocellular PVN OXT neurons (253). In addition, OXT cells express the receptor for leptin and administration of leptin at doses that support hypophagia activates OXT neurons (17, 59, 169, 249, 261). Anorexigenic glucagon-like peptide-1 and adrenomedullin increase OXT neuronal activity and/or promote neurohypophyseal release of OXT (60, 164, 237, 279). Another anorexigenic signal, alpha-melanocyte stimulating hormone (alpha-MSH), an endogenous melanocortin 3/4 receptor (MC3/4R) agonist, also stimulates the release of OXT from hypothalamic dendrites (194) and activates c-Fos in PVN OXT neurons (160). Similarly, the synthetic MC3/4R agonist, melanotan II (MTII), increases c-Fos in PVN OXT neurons (135, 168) where MC4Rs are co-localized with OXT neurons in rodents (52, 113) and humans (210). However, alpha-MSH also suppresses OXT release from the nerve terminals by inhibiting the electrical activity of OXT neurons (194), but the extent to which this contributes to OXT’s effects on energy balance remains to be determined. On the other hand, orexigenic peptides, including Agouti-related protein and opioids, tend to suppress activity of the OXT system, thereby likely delaying the onset of satiation (6, 157). Finally, chemo- and optogenetic manipulation of glutamate-releasing ARC neurons expressing the OXTR, promotes rapid satiety in mice (40).
2.4. Role of OXT in feeding reward
It is well accepted that OXT decreases feeding in the process of facilitating satiation and/or protecting homeostasis. This role is mediated mainly by the hypothalamic-brainstem circuit (15, 95, 147). Importantly, OXT regulates feeding also via mechanisms related to reward, consistent with the OXTR being expressed in the NAcc, amygdala, BNST and VTA (56, 78, 181, 217, 226).
Studies on eating for pleasure have revealed that centrally acting OXT reduces appetite for carbohydrates and for palatable non-carbohydrate sweeteners, such as saccharin. Peripherally administered BBB-penetrant OXTR blocker, L-368,899, elevates consumption of calorie-dilute and non-caloric solutions containing carbohydrates or saccharin, but it fails to modify intake of Intralipid in mice and rats (63, 156, 212). OXT injected directly in the VTA, NAcc core and basolateral amygdala (BLA) in rats decreases the intake of episodically presented palatable sweet solutions, and this effect is reversible by pretreatment with an OXTR antagonist, L-368,899 (94, 138). OXT knockout (KO) mice drink excessive amounts of sucrose- and saccharin-sweetened water (2, 139). In a two-bottle choice test in which water is a control tastant, KO mice prefer sucrose, Polycose and even a “bland” cornstarch emulsion, but their preference for Intralipid is unchanged (132, 205). Intake of a similar amount of calorie-matched sugar water vs. fat emulsion produces a higher number of c-Fos positive PVN OXT neurons in sucrose-fed mice, and the activation is higher at the end than at the start of consumption regardless of the macronutrient content (156). The aforementioned experiments utilizing fluid consumption strongly suggest that OXT is particularly effective in reducing intake of carbohydrates and sweet solutions. One should note, however, that data also indicate that the effect of OXT on feeding appears to be more dependent on energy density of food rather than macronutrient composition or flavor. In line with this notion, genetic ablation of OXT cells in the hypothalamus generates obesity in high-fat diet-fed animals (261). ICV treatment with an OXTR antagonist increases feeding in mice (275, 276) and rats (Blevins, unpublished) given a high-fat high-sugar diet. Finally, chronicity of palatable diet presentation may be a mitigating factor in meal-end responsiveness of OXT neurons to food load. Accordingly, occasional episodic intake of a high-sugar diet in rats engaged more PVN OXT neurons than low-sugar food in rats, however daily habitual intake of sugary food promoted a significant decline in the level of activation of OXT cells (133).
2.5. Use of chemogenetic and optogenetic approaches to understand the role of PVN OXT in the control of food intake
While the existing work highlights an important role for specific OXTR populations in the control of food intake in rats and mice, based on recent lentiviral, chemogenetic and optogenetic strategies in mice the picture is becoming less clear with respect to the role of PVN OXT neurons in the control of food intake. As mentioned earlier, Zhang and Cai found that lentiviral reduction in PVN OXT expression resulted in increased food intake in both chow-fed and high fat diet-fed mice (275). In contrast, Xi and Wu found that diphtheria-toxin elicited ablation of PVN OXT had no effect on food intake in chow-fed (262) or high fat diet-fed mice (261). In addition, chemogenetic activation of PVN OXT neurons failed to suppress baseline food intake (193) or food intake following stimulation of PVN OXT neurons at the start of the dark cycle (228). Both chemogenetic and photo-activation of PVN OXT also failed to suppress refeeding of low-fat diet or chow after a 24-h fast in mice (6, 193). On the other hand, chemogenetic or tetanus toxin-elicited silencing of PVN OXT neurons failed to stimulate food intake (chow) (50, 111). While it will be important to determine if chemogenetic or optogenetic manipulation of PVN OXT neurons is sufficient to 1) impact OXT release and/or elicit c-Fos from axonal terminals in the NTS (175, 176), these findings are at least consistent with 1) a minor role of PVN OXT neurons to suppress the refeeding response following extended fasts in rat models (212). They are also consistent with data from mice with global deficiencies in OXTR or OXT signaling that show no defects in weekly food intake (81) or daily chow intake (27, 235). Given that OXT’s effects can be optimized based on time of day (276) and composition of the diet (18, 34, 120, 121, 191, 275–277), future studies should take into consideration timing of the stimulus (early vs late light cycle), macronutrient content of diet, and whether more can be gained by manipulating either OXTR populations (193) or OXT fibers (193) that innervate specific OXTR populations before more definitive conclusions can be made with respect to the importance of endogenous OXT signaling in the mouse model.
In addition, given the heterogeneity with respect to PVN OXT populations and outgoing projections, targeting specific PVN OXT neuronal populations that project to areas with distinct roles such as the spinal cord (thermoregulation and energy expenditure) (228) or hindbrain (food intake, thermoregulation and energy expenditure) (162, 191) may contribute to different outcomes across studies. Current findings indicate there might be species differences with respect to the origin (rostral vs caudal) of the more predominant descending parvocellular PVN OXT neuronal projections to the hindbrain. In particular, parvocellular PVN OT neurons that project to the NTS are located predominantly in the caudal parvocellular PVN in rats (189). While it remains to be determined if this is the case in mice, there appear to be few OT projections from the rostral parvocellular PVN to the NTS in mice and those in the rostral PVN appear to project to spinal cord (228). These findings raise the possibility that parvocellular PVN OT neurons located more caudally project to subsets of NTS OXT receptor (OXTR)-expressing neurons in both mice and rats. It is also possible that, in the mouse model, parvocellular PVN OXT neurons do not innervate the hindbrain as densely compared to the rat model although existing studies provide indirect evidence in support of such a projection in mice (19, 128, 193, 260) and suggest that caudal hindbrain OXT receptors are relevant in the control of energy balance in both mice (19, 128) and rats (11, 15, 67, 161, 162) (covered in more detail below and in sections 2.1 and 3). Recently, projections from the 1) PVN and SON OXT neurons to the ARC (123), PVN OXT neurons to the VMH (141) and PBN (193) and ARC OXTR-expressing neurons to the PVN (40) have been described in the mouse model. While it appears that PVN OXT neurons do not project to the VMH in rats, further studies will need to determine in rats whether 1) OXT neurons project from the PVN and SON to the ARC and PBN and 2) ARC OXTR-expressing neurons project to the PVN.
3. Effects of OXT treatment on weight loss in obese animal models
Recent studies have shown that central, systemic, or IN administration of OXT decreases food intake, weight gain and/or BW in DIO rodents (34, 101, 120, 121, 137, 191, 206, 216, 275, 276), genetically obese rodents (1, 8, 76, 98, 124, 137, 177), ovariectomized rodents (74), dihydrotestosterone-induced rat model of polycystic ovary syndrome (PCOS) (73), as well as in DIO nonhuman primates (16). In sections 3.1–3.2, we will review the potential mechanisms that contribute to the effects of chronic OXT in rodent and nonhuman primate models of obesity.
3.1. Does chronic OXT treatment affect BW by increasing energy expenditure?
3.1.1. Effects of acute and chronic OXT treatment on energy expenditure
OT-elicited reductions in food intake do not appear to fully explain OT-elicited weight loss. Namely, the effectiveness of OXT to decrease energy intake in DIO or genetically obese rodents appears to wane over time despite an intact response of OT to continue to reduce weight gain or BW (18, 34, 120, 121, 191). Whether the waning of the feeding effect is associated with OXT-elicited reductions in receptor binding (43, 72, 171) following chronic administration (further discussed in 7.2) remains to be determined. This is evident when rodents receive 1× daily repeated peripheral injections (120, 121) as well as chronic peripheral (1, 18, 34, 120, 121) and central infusions (18, 34). In addition, indirect evidence from pair-feeding (food in control group is matched to that of OXT-treated animals) studies in mice and rats, where the amount of weight loss following OXT exceeds that of pair-fed control animals (1, 18, 34), suggests that other mechanisms such as increased EE may also contribute to OXT-elicited weight loss. Furthermore, recent studies have shown that single or repeated acute injections of OXT increase energy expenditure in both rodents (147, 275, 276) and DIO nonhuman primates (16). Chronic infusion of OXT may also be able to prevent the counter-regulatory mechanisms that promote weight regain in DIO rodents following sustained weight loss, in part, by maintaining the level of energy expenditure to that of vehicle-treated control rats (18) and mice (121). Thus, chronic OXT treatment (repeated injections or minipump infusions) may be able to produce prolonged weight loss, in part, by increasing energy expenditure and/or preventing the reduction in energy expenditure associated with prolonged weight loss.
3.1.2. Are OXT-elicited effects on energy expenditure produced by increases in brown adipose tissue (BAT) thermogenesis?
3.1.3. Role of endogenous OXT on energy expenditure and BAT thermogenesis
PVN OXT neurons are anatomically positioned to control energy expenditure by outgoing sympathetic projections to interscapular BAT (IBAT) (151), stellate ganglia (77) [sympathetic ganglia known to innervate IBAT (151)], as well as white adipose tissue [WAT; epididymal (208, 220) and inguinal WAT (208)]. Cold exposure activates PVN OXT neurons (81) and rostral medullary raphe (RMR) neurons that express OXTRs (82). Furthermore, data from animals with impaired or defective OXT signaling (e.g. OXTR and OXT null mice, mice with viral or diphtheria toxin-elicited reductions in PVN OXT neurons, or mice treated with an OXTR antagonist) show reductions in energy expenditure (261, 275, 276) and/or defects in cold-induced thermogenesis (80–82, 235, 262), the latter of which have been shown to be rescued by OXT treatment (262). In addition, histological assessment revealed the presence of large lipid droplets in IBAT in OXTR null mice which suggests hypo-activity of IBAT tissue (235). Furthermore, chemogenetic activation of rostral PVN OXT neurons, some of which project to the thoracic spinal cord, induces c-Fos in thoracic spinal cord cholinergic neurons, increases energy expenditure and tends to increase IBAT temperature (228), although the extent to which these effects are attributed to endogenous OXT remains to be determined. In addition, viral expression of OXTRs into the RMR (82) and hypothalamus (80) in OXTR null mice restores deficits in cold-induced thermogenesis; expression of OXTRs in RMR also normalized lipid droplet size in IBAT tissue to that of control mice (82). Together, these data suggest that OXTRs in these areas may be critical in regulating BAT thermogenesis during cold exposure. Collectively, these findings support the hypothesis that endogenous OXT signaling can control BAT thermogenesis and energy expenditure, but the extent to which its effects on energy expenditure require BAT thermogenesis and/or browning of WAT and the OXTR populations that are required to mediate these effects remain to be determined.
3.1.4. Role of exogenous OXT on BAT thermogenesis and browning of WAT
We know from recent studies that 3V or 4V administration of OXT elevates IBAT temperature in rats and mice and that these effects extend to DIO rats (191). Furthermore, chronic 3V infusion of OXT elevates IBAT temperature at a time that coincides with the onset of OXT-elicited weight loss in DIO rats (191). Consistent with these findings, 4V OXT also elevated core temperature in rats (162). These findings are particularly relevant given that changes in IBAT temperature tend to precede and contribute to changes in core temperature in cases of fever and stress (84). The effects of OXT on IBAT temperature appear to be related to non-shivering BAT thermogenesis as chronic OXT administration is not associated with elevations in locomotor activity in rats (18, 34, 74) or mice (121). Chronic subcutaneous (sc) OXT treatment was also associated with increased uncoupling protein-1 (marker of brown adipocytes) immunostaining in sc WAT (177). It will be helpful if future studies could evaluate the impact of chronic OXT treatment on additional markers of browning in WAT as well as WAT temperature as a more functional readout of WAT thermogenesis. While these collective findings indicate that exogenous OXT increases BAT thermogenesis, the extent to which BAT thermogenesis and browning of WAT is required for chronic OXT to increase energy expenditure and evoke weight loss is an ongoing area of investigation.
3.2.1. Does chronic OXT treatment impact BW by increasing lipolysis?
3.2.2. In vitro and in vivo data
OXT increased glycerol in 3T3-L1 adipocytes (268), which are known to express OXTRs (198, 268), suggesting that OXT stimulates lipolysis, in part, through a direct effect. Similarly, OXT increased glycerol release from epididymal fat pads ex vivo (34). OXT also elevated serum glycerol and reduced serum triglycerides after a 14-day treatment period in rats (34). Blevins extended these findings in a translational nonhuman primate model where chronic 2× daily administration of OXT increased both serum free fatty acids and glycerol and reduced serum triglycerides following a 4-week treatment period in DIO rhesus monkeys (16). A further study showed that chronic OXT increased epididymal WAT expression of lipoprotein lipase (Lpl) and fatty acid transporter (Fat), both of which are linked to uptake of triglycerides and fatty acids, respectively. While chronic OXT treatment did not impact enzymes linked to lipogenesis and storage of triglycerides (acetyl-coenzyme A carboxylase alpha, fatty acid synthase, and diacylglycerol O-acyltransferase homolog 1), it did increase enzymes linked to lipolysis (patatin-like phospholipase domain containing 2 and hormone-sensitive lipase) (34). Chronic central or peripheral OXT treatment is also associated with reductions in respiratory quotient in DIO mice (121) and rats (18, 34) relative to vehicle treatment (18, 34, 121) or pair-fed control animals (34). Taken together, these findings suggest that increased lipolysis and lipid oxidation may contribute to OXT-elicited weight loss.
3.3. How does chronic OXT treatment impact body composition?
Chronic central (lateral ventricle, 3V) or peripheral OXT treatment elicited a relative reduction (post vs pre-intervention) in fat mass or decreased total fat mass (post-intervention) compared to vehicle in lean (34) and DIO rats (18, 34, 137, 191), DIO C57BL/6J (191) or C57BL/6 mice (216, 276), db/db mice (177), and ob/ob mice (1) without impacting lean mass. In regards to targeting specific fat depots, OXT treatment appears to reduce visceral (specific visceral fat depot not determined) (120), sc (120), mesenteric (121) and epididymal fat (1, 121) in DIO C57Bl/6J (121) and ob/ob mice (1), as well as in female ovariectomized Wistar rats (74). It has also been shown to reduce adipocyte area from mesenteric (121), epididymal (8, 38, 121), sc (74, 177), visceral (74), perirenal (177), and epicardial (177) depots in db/db mice (177), C57Bl/6J mice (121) obese Zucker rats (8) or female ovariectomized Wistar rats (74). Chronic sc OXT treatment also reduced liver weight and fat in hepatocytes in DIO C57BL/6J mice (121) but was found to have no effect on liver triglyceride content in ob/ob mice (1). In contrast, some studies have reported that chronic systemic or central (4V) OXT administration elicited a relative reduction in lean mass compared to vehicle in lean C57Bl/6J (1) and DIO C57BL/6 mice (216) as well as in DIO CD IGS rats (191) with or without any relative reductions in fat mass. However, chronic 4V OXT infusions produced a relative reduction in fat mass without impacting lean mass compared to 4V vehicle in DIO Long-Evans rats (191) raising the possibility that differential effects may be attributed, in part, to rodent strain, dosing and/or route of administration. While there are exceptions, overall OXT treatment appears to preferentially reduce fat mass while preserving lean mass across rodent models.
3.4. Does chronic OXT treatment produce sex-specific effects on energy balance in animal models?
While an earlier study reported no sex-specific effects of acute OXT administration (central or peripheral) on food intake in male and female rats (12), the majority of studies that have tested the effects of chronic OXT on energy balance have focused predominantly on male animal models. One previous study found that chronic OXT reduced both food intake and weight gain in female Sim1 haploinsufficient mice but these effects were not observed in female wild-type mice (98). Maejima recently compared the effects of chronic sc administration of OXT on weight loss in both male and female DIO C57Bl/6J mice (120) and found that OXT reduced BW and fat mass (sc and visceral) to a similar extent in both sexes. Seelke recently reported that female DIO prairie voles may be more sensitive than males to the effects of IN OXT on BW although the sample size in this study was small (206). In addition, chronic OXT treatment was found to reduce food intake and BW in female ovariectomized rats (74). Further studies will need to confirm these effects in other male and female models of obesity, including nonhuman primates and humans.
3.4.1. Are there sex differences with respect to the effects of endogenous OXT signaling on energy balance?
There have been somewhat inconsistent results from the few studies that have examined the effects of global loss in OXT or OXTR signaling on energy balance in male and female rodents. While some have found that both male (27, 81) and female OXT null mice (27) develop adult onset obesity (27, 81), only male OXTR deficient mice become obese (235). In addition, selective ablation of PVN OXT neurons in Oxytocin-IresCre:Rosa26iDTR/+ mice predisposes male but not female mice to diet-induced obesity relative to control Rosa26iDTR/+ mice (261). Further studies will be required in order to provide more clarity on whether there are sex differences in the role of endogenous OXT in regulating energy balance.
3.5. Does OXT treatment produce adverse side effects in animal models?
3.5.1. Visceral illness
Previous studies have indicated that chronic systemic administration of OXT, at doses that are effective in reducing weight gain or evoking weight loss, does not appear to be associated with nausea or food aversion in rodent models. In particular, chronic peripheral administration of OXT (50, 100, 200 nmol/day) failed to significantly alter 2-h saccharin preference ratios in fluid choice tests (saccharin intake/water intake; a paradigm in which a reduction in saccharin preference ratios is a readout of a CTA) and OXT (50 nmol/day) also failed to increase kaolin consumption (index of pica behavior) (18). Chronic central delivery of OXT (3V, 4V) did not impact kaolin consumption in DIO mice or rats (18, 191). These findings extend previous reports showing that acute bolus injections of OXT into the CNS [3V (4 μg), VMH (0.1, 1 nmol)] or periphery (200 μg/kg, IP), at doses that reduced food intake, failed to impact kaolin consumption (275) or elicit a CTA (76, 147, 275) in mice or rats. In addition, no evidence of diarrhea or vomiting was found following daily sc OXT treatment over a 4-week period in rhesus monkeys (16). Taken together, these findings suggest that peripheral or central delivery of OXT, at doses that evoke weight loss, are not associated with visceral illness.
3.6. Effects of OXT treatment on glucose homeostasis in animal models
While the majority of studies to date largely provide support for a therapeutic role of OXT to stimulate insulin secretion, insulin sensitivity or glucose tolerance, there are also cases of conflicting data that may be related to animal model, route of administration, or dosing. Chronic systemic infusions [sc; 1.6 μg/kg/day (~0.0563 nmol/day)] or repeated 1× daily systemic administration (IP; 1 mg/kg) over a 1 to 6 week period resulted in an improvement in glucose tolerance in high-fat diet (HFD)-fed mice (121, 276, 277). Similar results were obtained even in the absence of any weight loss when HFD-fed mice received 2× central injections of OXT [3V; 4 μg] during an overnight fast (277). Others have shown that chronic central (ICV; 1.6 nmol/day) or systemic (sc; 50 nmol/day) infusions at the end of the 14-day infusion period either improved glucose tolerance or increased insulin sensitivity in HFD-fed rats (34). Acute IN (0.1, 1 or 10 μg) or central (ICV; 0.4 and 4 μg) administration failed to impact glucose tolerance but acute IP administration of OXT (IP; 40 and 400 μg/kg) improved glucose tolerance in C57BL/6J mice (122). Similarly, acute systemic administration of OXT (IP; 2 mg/kg) improved glucose tolerance in both lean and obese C57BL/6 mice (216) and these effects appear to be mediated, in part, by increased insulin secretion (216). In addition, acute injections of IN OXT over a 7-day period were able to reduce fasting levels of insulin and also appeared to reduce fasting levels of glucose (not significant) in a DIO prairie vole model (206). Finally, 2× daily sc OXT administration over a 4-week period (0.2 mg/kg for initial 2 weeks; 0.4 mg/kg for final 2 weeks) decreased fasting blood glucose in a more translational DIO rhesus monkey model (16). However, chronic ICV (1.6 nmol/day) or 3V infusion (16 nmol/day) failed to exert a significant effect on 1) blood glucose and insulin in the fed state (34) as well as 2) fasting blood glucose (18, 191) and insulin (191) at the end of the 14- to 28-day infusion periods in DIO rats and mice. In contrast, chronic systemic infusions of OXT (sc; 50 nmol/day) over 14 days appeared to worsen fasting blood glucose and fasting insulin at the end of the treatment period in C57BL6/J mice (1). In addition, the same group reported that chronic systemic infusions of OXT (sc; 50 nmol/day) over 14 days appeared to worsen both fasting blood glucose at the end of treatment and daily basal blood glucose in the fed state during the infusion period in ob/ob mice (1). Similarly, chronic systemic infusions of OXT [sc; 3.6 μg/100 g or ~8.22 (lean) or 12.5 nmol/day (obese)] over 15 days failed to impact fasting blood glucose but worsened fasting insulin in both lean and obese Zucker rats (8). In addition, this study found that OXT infusions over 13 days impaired glucose tolerance in obese Zucker rats (8). To summarize, the majority of data from animal models appear to support a promising effect of exogenous OXT to improve glucose tolerance. However, additional studies that 1) include weight-restricted controls in both genetically obese and DIO rodents and DIO nonhuman primates, and 2) explore whether differences in route of administration, strain, and/or dosing will be helpful in clarifying the role of OXT signaling on glucose homeostasis. Given that OXTRs are present on α cells and β cells in pancreatic islets (229), OXT may act, in part, to stimulate insulin and glucagon release from mouse islets (49) and rat pancreas (36, 37) through a direct effect, but may also stimulate insulin secretion through an indirect effect by activating vagal cholinergic neurons that innervate β cells (14). There is also evidence to suggest that OXT may increase insulin sensitivity, in part, by increasing insulin-sensitive glucose transporter 4 mRNA in fat (38).
4. OXT levels in animal models of obesity
4.1. Serum measurements in animal models
It is important to acknowledge that measurements of endogenous levels of OXT in serum or plasma remain controversial and assay validation continues to be an ongoing need (110, 119, 129). Many laboratories have used radioimmunoassays (RIAs), enzyme-linked immunosorbent assays (ELISAs) or mass spectrometry to measure circulating levels of OXT. Many of the newer commercially available ELISA kits have generated higher levels of OXT which may be due, in part, to interference from other substances, including OXT fragments and/or OXT degradation products (233). The extent to which these may be biologically active (and potentially impact energy balance) remains to be determined. Recent work with commercially available ELISA kits has resulted in endogenous OXT levels being up to two orders of magnitude higher than RIAs in cases where extraction was not incorporated (129). Extraction before ELISAs helps to eliminate some of the interfering substances and results in yields that are similar to those obtained with the more traditional RIAs (233). However, the discarded substances may include OXT bound to proteins (22) as well as OXT fragments and/or OXT degradation products. While absolute OXT levels cannot be compared across studies due to assay differences, measurements can be useful for comparing relative levels of peripheral OXT between groups. Future studies should also consider the impact of circadian variations that occur with respect to circulating OXT (276) and how circulating OXT levels may be impacted by 1) factors that impact central release of OXT (34), 2) feeding (116, 252, 265) and 3) obesity in DIO (276) and genetically obese animal models (48, 177, 204, 273).
4.2. “Genetic” models
Endogenous OXT levels have been measured in different genetic rodent models of obesity with divergent results.
4.2.1. Otsuka Long-Evans Tokushima Fatty (OLETF) and Zucker rats
The OLETF rat has a null-mutation in a gene that expresses a receptor (cholecystokinin 1 receptor) for the satiety signal cholecystokinin (CCK) and it presents with overeating-induced obesity, hyperleptinemia and predisposition to type 2 diabetes (85). OLETF rats showed significantly higher plasma OXT levels compared to the lean, Long-Evans Tokushima Otsuka (LETO) control strain (204, 273). Pair-feeding (diet restriction) from weaning can dramatically normalize OLETF rats to lower mean BW and plasma OXT levels, similar to those of lean LETO controls (202, 203).
Recently, levels of OXT gene expression in the PVN were measured on postnatal day 23 (PND 23) and PND 90 by in-situ hybridization in OLETF and LETO rats (89). PVN OXT expression was lower in OLETF rats both at PND 23 and PND 90, compared to lean LETO controls. This pattern is the opposite of what was found for dorsomedial hypothalamus (DMH) neuropeptide Y (Npy) gene expression. This raised the possibility that DMH NPY down-regulates OXT expression in the PVN. As discussed in the current review, PVN OXT has been postulated to mediate the effects of leptin on meal size (17). Thus, the signaling pathway of DMH NPY regulation of PVN OXT in modulating food intake merits further investigation.
In Zucker fatty rats, obesity is a consequence of spontaneous mutation (fa) in the gene encoding the leptin receptor, resulting in hyperphagia (174) and characteristics of type 2 diabetes (83). Obese Zucker rats had 40% lower plasma OXT levels, assessed by enzyme immunoassay (EIA), compared to lean Zucker rats (48).
4.2.2. ob/ob and db/db mice
The leptin deficient ob/ob mouse model shows extreme obesity together with high basal glycemia and insulinemia, thus presenting a model of an advanced stage in the development of type 2 diabetes. However, in males, their plasma OXT levels were similar to those of lean controls (1).
The leptin receptor-deficient db/db mouse is characterized by an obese phenotype with hyperglycemia and subsequent hyperinsulinemia as a result of two mutant copies of the leptin receptor gene. Male obese and diabetic db/db mice had lower (46%) serum OXT levels compared with control db+/mice (177).
In summary, circulating levels of OXT in genetic models of obesity are either increased (OLETF), decreased (Zucker rats, db/db mice) or not different (ob/ob mice) from lean controls. Whether differences in leptin signaling, genetic background, and/or OXT-degrading enzymes (48) contribute to these divergent results is an area for future research. Central expression/levels have been studied less frequently; obese OLETF rats showed lower OXT gene expression in the PVN. There are not enough data available on central expression levels to reach conclusions at this point.
4.3. High-fat/cafeteria DIO rodent models
HFD
Rats maintained on a chow or “cafeteria” diet weighed 14% more than chow-fed controls and had significantly higher fasting levels of plasma OXT measured by RIA (148). In contrast, lower levels of plasma and serum OXT have been reported at study termination in male HFD-fed DIO mice relative to chow-fed mice (275, 276). However, Maejima (120) did not see a decrease of plasma OXT in HFD-fed DIO mice at study termination relative to chow-fed mice [note that this may have been due to length of time the mice were on HFD (8 weeks (males) or 12 weeks (females) prior to study termination and blood collection relative to the other study (6 months) (275)]. In a study from our (Blevins) lab (137), we also did not see a decrease in serum OXT in male HFD-fed DIO rats (or in DIO mice (Blevins, unpublished) relative to low fat/chow diet-fed controls (though there was a non-significant tendency in the direction of a decrease in the rats). Comparing our (Blevins) null results and previous reports (275, 276), our mice were on the HFD for about the same amount of time.
Regarding brain OXT levels, in a recent, yet unpublished study in our (Weller) lab, male Wistar rats received 60% HFD from weaning (PND 21) to adulthood (PND 120). Preliminary data show that compared to rats raised on standard chow, these rats showed 3-fold higher OXT mRNA expression (measured by real time qPCR) in the hypothalamic PVN (Cohen-Or, Gerberg, Weller & Meiri, unpublished). However, using a much shorter period of exposure to this 60% HFD, 10 days, from PND 20 to PND 29 in male Sprague Dawley rats, significantly lower OXT levels (measured by ELISA) were found in the infralimbic medial prefrontal cortex, compared to chow-fed controls (267). Note that there were no significant BW differences between the two groups in this study. The two latter studies differ in many aspects, including brain area assessed, method of measurement and duration of HFD exposure, so clearly further research is warranted. Furthermore, Zhang and Cai used an ex vivo OXT release assay to determine if HFD feeding could affect OXT release in hypothalamic PVN and found that KCl-induced depolarization elicited OXT release in PVN slices of chow-fed wild type mice, but this effect was blunted by HFD feeding (276).
Summarizing this section, the overall data seem to be mixed in the cafeteria and HFD-induced obese rodent models, but this may be, at least in some cases, due to length of time on the high fat diet, time of blood collection (276), and/or species/strain differences.
5. Endogenous oxytocin levels and relationship to markers of energy homeostasis in humans
As in animal models, there is controversy regarding measurement of peripheral OXT levels in humans (119); however these assessments can be useful in comparing relative between-group levels of circulating OXT as well as the relationship between OXT and related clinical endpoints, such as appetitive behavior and markers of energy homeostasis (102). Consistent with the role of OXT as a signal of energy availability, serum OXT levels are positively associated with body mass index, leptin levels, and body fat in non-diabetic females across the weight spectrum (104, 199). Circulating OXT levels are low in states of energy deficit, for example in low-weight women with anorexia nervosa (104, 136) or normal-weight females with exercise-induced hypothalamic amenorrhea and low percent body fat (103) compared to controls. There is also evidence for higher peripheral OXT levels in individuals with excess energy stores, including overweight/obese women and men (166, 199, 222, 234, 254). However, in a number of studies of obese adults with a high prevalence of type 2 diabetes or insulin resistance (46, 125, 182) and children with insulin resistance (13), OXT levels were low; this may be directly related to impaired glucose homeostasis as individuals with type 1 or type 2 diabetes have low OXT levels (99, 182). Some studies in individuals with type 2 diabetes or insulin resistance and overweight/obesity have found that lower OXT levels are associated with increased features of metabolic syndrome (13, 46, 125, 272). However, one study of older men (mean weight 80 kg) -- a minority of whom had type 2 diabetes -- found that higher OXT levels were associated with greater weight and metabolic syndrome (234), and another of men and women across a wide spectrum of weights with insulin resistance in those with obesity reported higher OXT levels were associated with greater body mass index and insulin resistance (Homeostatic Model Assessment of Insulin Resistance HOMA-IR)] (166). Consistent with these data, a recent study of 721 adults across the weight spectrum (excluding participants with diabetes) found a positive association between OXT levels and HOMA-IR, which remained significant after adjusting for sex, age and body mass index (BMI) (254). Further research will be important to explain these divergent results and how metabolic status and BW impact OXT secretion. Whether low OXT levels in some individuals with overweight or obesity, e.g., in the context of type 2 diabetes, contribute to weight gain is an important area for future investigation.
Studies examining endogenous OXT levels in response to caloric intake in humans also show discrepant results, with no change (131, 222), increased (150), or decreased (7) postprandial levels reported, possibly related to differences in meal size, macronutrient or fiber content, meal form (liquid vs. solid) or in some cases, small sample size. A recent investigation of 55 healthy women and girls fed a balanced mixed meal showed a 20% postprandial decrease in OXT levels (7). Further, a smaller percent decrease in postprandial serum OXT was associated with a greater sense of fullness and less hunger after the 400 kcal mixed meal, suggesting that endogenous OXT secretion may modulate subjective appetite in humans (7).
6. Effects of IN oxytocin administration on energy homeostasis and metabolism in humans
Recent translational studies have shown that intranasal (IN) administration of supraphysiologic OXT decreases food intake and/or BW in obese humans (69, 105, 239, 277). A single dose of 24 IU IN OXT in men reduces caloric consumption without impacting subjective appetite (25, 105, 219, 239). Importantly OXT reduces food intake in fasted (105, 239) and fed (25, 165, 239) states with preferential effects in reducing sweet (25, 165, 239), fatty (105) and salty (25) foods, suggesting effects on both homeostatic and hedonic eating. Mechanistic studies using functional magnetic resonance imaging (fMRI) have confirmed that OXT modulates homeostatic (e.g., hypothalamus) (178, 245) and hedonic (e.g., ventral tegmental area, orbitofrontal cortex, insula) (178, 219) neurocircuitry in response to food images. In addition, fMRI studies have shown that OXT increases activation of brain regions responsible for self-control (e.g., anterior cingulate cortex, prefrontal cortex, supplementary motor area) (178, 219, 224). Therefore, OXT may reduce caloric intake in humans via effects on multiple neurobiological pathways, including homeostatic, hedonic and impulse control circuitry.
Based on data in animal models showing that OXT reduces body fat and induces weight loss in part via increased fat utilization and energy expenditure (previously discussed in section 3.1), human studies have recently examined whether IN OXT has similar effects in men. A single dose of 24 IU IN OXT reduced respiratory quotient in men, indicating increased fat utilization (105), but had no impact on resting energy expenditure (105, 239) or diet-induced thermogenesis (239). Only one small pilot study to date has investigated the chronic effects of IN OXT on BW in humans with overweight/obesity (277). In this randomized, placebo-controlled trial of 20 men and women, nine of whom completed OXT treatment, eight weeks of 24 IU IN OXT before meals and at bedtime resulted in approximately 20 lbs of weight loss and 10 cm waist circumference reduction (277). The effects of chronically administered OXT on eating behavior, fat metabolism and energy expenditure were not assessed. Currently, an NIH-funded study of 8 weeks of IN OXT for weight loss is underway in 60 adults with obesity ( NCT03043053). This study, which includes an equal number of males and females, will investigate the efficacy, safety and underlying mechanisms of chronic IN OXT (24 IU before meals and at bedtime) as a weight loss therapy.
As in animal models (see section 3.6), the effects of IN OXT on glucose homeostasis are inconsistent in human studies. A single dose of 24 IU IN OXT in men across the weight spectrum reduced fasting insulin without a change in glucose levels, consistent with improved insulin sensitivity (105); after a meal, OXT resulted in reduced glucose levels without a change in insulin, independent of caloric intake (165, 239). Interestingly, OXT attenuated the increase in insulin and glucose after a 75 gram oral glucose tolerance test in men of normal-weight (92) but not in those with obesity and insulin resistance (23), suggesting that the effects of exogenous OXT on glucose homeostasis may be impacted by an individual’s adiposity and/or glucose regulation status. This is supported by the finding that 8 weeks of IN OXT failed to improve fasting or postprandial levels of insulin or glucose after a 100 g standard meal despite weight loss in a small study of prediabetic men with overweight/obesity (277).
While the glucose regulatory effects of OXT may be reduced in the context of obesity (23), there is evidence that OXT effects in modulating homeostatic caloric intake (239) and weight (277) may be stronger in obesity. In a study of 18 men with obesity and 20 men with normal weight, a single dose of 24 IU IN OXT vs. placebo reduced caloric intake in the fasting state only in those with obesity, while reducing postprandial snacking in both groups (239). Further, in the pilot study of chronic IN OXT for weight loss in adults, those with obesity (BMI approximately 34–44 kg/m2) appeared to have greater weight reduction than those with overweight (277). Future studies will be important to improve our understanding of the impact of adiposity and metabolic status on OXT effects in humans. More investigation of women will also be critical as OXT is known to have sex-specific effects.
7. Therapeutic implications
Based on the effects of OXT in reducing caloric intake, increasing energy expenditure, decreasing metabolically unfavorable fat depots (e.g., visceral and possibly liver fat), and potentially improving glucose homeostasis, OXT-based therapeutics are under active investigation for weight loss in humans (102). Intravenous/intramuscular OXT is FDA-approved and has a long track record of clinical use in parturition (244). Risks in the setting of parturition include arrhythmias, changes in blood pressure (see section 7.1 below) and rarely hyponatremia (244). IN OXT was previously FDA-approved (at lower doses than studied for weight loss) for lactation in women and was discontinued due to business -- not safety -- concerns. It continues to be used outside of the U.S. and is generally considered to be safe and well-tolerated. However, safety of IN OXT in individuals with obesity at the doses required for weight loss will require systematic investigation. Rigorous randomized controlled trials will also be necessary to determine the efficacy and underlying mechanisms of OXT actions. Currently proof of concept trials of IN OXT in adults with obesity ( NCT03043053) and children/young adults with hypothalamic obesity due to brain tumors ( NCT02849743) are underway.
7.1. Effects of OXT treatment on blood pressure and heart rate in animals and humans
Anorexigenic agents that that are thought to activate SNS outflow, including MC3/4R ligands which act, in part, through downstream CNS OXT circuits (see section 2), may adversely affect cardiac function in rodents (100, 213), nonhuman primates (88) and humans (57). This is a potential concern given the large number of overweight and obese humans with hypertension (145). Current data suggest that PVN OXT neurons are anatomically positioned to control SNS outflow (77, 151, 208, 215, 228) and OXT null mice appear to have reductions in SNS tone (27). However, animals studies have provided mixed results demonstrating an increase, decrease or no change in heart rate (65, 118, 121, 142, 173, 177, 266, 269) and blood pressure (118, 121, 142, 173, 271) depending on species and route of OXT administration [for review see (58, 172)]. More recently, acute systemic injections of OXT at doses that suppressed food intake (3 mg/kg, IP), resulted in an increase in blood pressure in DIO Long-Evans rats (1 and 3 mg/kg, IP) while a more selective OXT analogue (OXTGLY) had no effect (216). Studies using IN OXT in humans have not reported adverse side effects on heart rate (26, 91, 105) or blood pressure (91, 105) in men (26, 91, 105, 277) and non-pregnant women (91, 277). While the data are mixed in animal models (depending on species and route of administration) and IV/IM OXT in the context of childbirth may have effects on blood pressure and/or heart rate, preliminary studies in overweight/obese humans using IN OXT, at doses shown to reduce caloric intake (105) and BW (277), have not been shown to have an effect on either. It will be important to confirm in future studies that chronic administration of OXT-based therapeutics is not associated with adverse effects on blood pressure and heart rate in obese nonhuman primates and humans.
7.2. Remaining challenges
One limitation to the use of OXT as a therapeutic target to treat obesity is the short half-life in the periphery [T1/2=3.2–6 min (39, 247)] relative to the CNS [T1/2=19 min (130)]. There is a focus on developing novel therapeutics to target the OXT system as a way to circumvent the short half-life and prolong duration of action in the periphery. In some cases, modifications have been made to the disulfide bond (140) [essential for biological activity (140, 225)] to alter its activity and stability. Recent findings indicate that when encapsulated into a nanoparticle formulation, OXT may have improved stability and passage across the blood brain barrier (163, 274). One other modification has been to replace the Pro7 of OXT with either N-(p-fluorobenzyl)glycine or N-(3-hydroxypropyl)glycine which yielded long-lasting effects in an animal model of social behavior (71). Carbetocin is another analogue with modifications that include N-terminal desamination and replacement of the 1–6 disulfide bridge with a methylene group (70) and a reported half-life of 17.2 min in horses (201) and 41 min in non-pregnant women (232). Altirriba and colleagues have demonstrated that carbetocin reduces weight gain over a 10-day period in ob/ob mice (1), but it has not yet been examined in DIO rodent models. Novel OXT analogues have recently been produced by attaching a fatty acid to the peptide (acylated OXT) or replaced proline (position 7) with glycine in the acylated OXT to generate acylated OXTGly. Chronic systemic treatment (1× daily) with acylated OXT (sc, 2 μmol/kg) and acylated OXTGly (sc, 2 μmol/kg) over a 14-day period reduced food intake and BW in DIO C57BL/6 mice. As expected, these effects were also associated with reductions in fat mass. While acylated OXTGly exerted a protective effect on lean mass, acylated OXT actually produced a relative reduction in lean mass (216) (also see section 3.3). Snider also noted improvements in glucose tolerance following acute administration of another analogue, OXTGly, in both lean and DIO C57BL/6 mice (IP, 2 mg/kg) (also see section 3.6). In contrast, they found that chronic 14-day treatment with acylated OXT or acylated OXTGly failed to improve glucose tolerance in DIO C57BL/6 mice and, in fact, acylated OXTGly may have actually worsened glucose tolerance (216). Consistent with this finding, fasting plasma glucose tended to be higher following chronic treatment with both analogues (not significant) while fasting plasma insulin was significantly higher in the group treated with acylated OXTGly. Zhang and Cai found that 2× daily 3V administration of other analogues, [Ser4, Ile8]-OT (isotocin) and [Asu1,6]-OT, resulted in an improvement in glucose tolerance and fasting blood insulin levels that were independent of changes in BW in HFD-fed C57Bl/6J mice and streptozotocin-treated mice with diabetes (277). Importantly, from a translational perspective, the improvements in glucose tolerance independent of changes in BW were also observed following peripheral administration of [Ser4,Ile8]-OT (277). It will be helpful to include weight-restricted controls and examine the effects of these analogues on glucose tolerance following a more extended treatment at doses that are not associated with adverse side effects in both genetically obese and DIO rodent and DIO nonhuman primate models.
OXT is a pulsatile hormone (10, 47, 146), but the secretory patterns in humans are not well defined. Thus far, OXT administration multiple times per day, continuously, or during the early part of the light cycle to coincide with diurnal peaks of circulating OXT in rodent models (corresponds to time when rodents do not normally eat) appears to maximize its effects (16, 276). Further research into the secretory dynamics of OXT in normal human physiology may help guide optimal drug development and dosing. It would also be useful to examine if intermittent or pulsatile administration (30–32, 62, 278) of OXT in animal models will enhance its ability to promote weight loss and potentially overcome the reductions in OXTR binding that develop with chronic CNS infusions (43, 72, 171). However, further studies will be required to determine at what point reductions in OXTR binding occur following continuous OXT infusions and whether this precedes or coincides with the waning of OT’s effects on food intake. Mechanistic studies delineating central vs. peripheral effects of OXT will also provide important information. In vitro data indicate that OXT has a 150-, 1250- and 4375-fold greater affinity for the human OXTR relative to the vasopressin (V1aR), vasopressin 1B receptor (V1bR) and vasopressin 2 receptor (V2R), respectively (126). Similar specificity has also been observed for the rat and mouse species (personal communication with Dr. Gilles Guillon). While recent findings indicate that OXT failed to reduce food intake in OXT null mice (75), previous in vitro data indicate that OXT may also cross-react with the V2R receptor (216) and others have reported that OXT is about 10-fold more selective for the OXTR relative to VP (54, 179). Given the mixed results and the possibility for off-target effects, OXT agonists that are more selective to the OXTR could reduce the potential for side effects due to cross-reactivity with VP receptors. Ultimately using OXT-based therapeutics given alone or in combination with other interventions targeting other pathways (69) may be useful in improving efficacy while minimizing side effects.
8. Conclusions
Taken together, preclinical data indicate that OXT is involved in the regulation of eating behavior, energy expenditure, lipid metabolism, and glucose homeostasis. The anorexigenic properties of OXT appear to be mediated by homeostatic and reward-related neural pathways (4, 5, 15, 17, 64, 132, 138, 152, 153, 156, 205). Studies in humans support these mechanisms, and also indicate that OXT may act via modulating impulse control circuitry (178, 219, 245). While studies in animal models show that acute or chronic administration of OXT increases energy expenditure (16, 147, 275, 276), single dose IN OXT does not appear to impact energy expenditure in men (105, 239). The relevance of proposed mechanisms for OXT effects on energy expenditure in animal models (via sympathetic activation of BAT and WAT) may be less relevant in humans, and the question of whether chronic administration of OXT increases energy expenditure in humans is an important area for future investigation. OXT increases markers of lipolysis and reduces fat mass, including metabolically unfavorable visceral fat depots, in animals (1, 18, 34, 74, 120, 121, 137, 177, 191, 198, 216, 268, 276). In humans, IN has been shown to increase fat utilization when given as a single dose (105), and reduce waist circumference (a marker for visceral fat) when given chronically over eight weeks (277). Effects of OXT on glucose homeostasis are mixed in preclinical and human studies, and may depend on dose, delivery, and/or metabolic state. In humans, several studies have shown improvements in glucose homeostasis with a single dose of IN OXT (92, 105, 165, 239), however one investigation in obese men with insulin resistance did not demonstrate OXT effects in response to an oral glucose tolerance test (23). Further, a pilot study of eight weeks of IN OXT in obese adults showed no improvement in glucose homeostasis despite significant weight loss (277).
Based on translational data demonstrating that OXT induces weight loss, OXT-based therapeutics are considered promising for the treatment of obesity. Safety, efficacy and underlying mechanisms of these drugs will be important to investigate in patients with obesity.
GRANTS & ACKNOWLEDGEMENTS
This material was based upon work supported by the Office of Research and Development, Medical Research Service, Department of Veterans Affairs (VA) and the National Institutes of Health (NIH). This work was also supported by NIH grants R01 DK109932 (EAL), R01DK115976 (JEB), and P30 DK040561 (EAL); Merit Review Award BX004102, Office of Research and Development, Medical Research Service, United States Department of Veterans Affairs Biomedical Laboratory Research and Development Service (JEB); Royal Society of New Zealand grant 1203 (PKO); Israel Science Foundation grant 1781/16 (AW) and Israel Ministry of Science, Technology & Space, grant 3-13608 (AW). This review is based on work presented in the symposium titled, “Oxytocin and vasopressin as regulators of metabolism and food consumption” during the 13th World Congress on Neurohypophyseal Hormones, April 8–April 11, 2019, at the Ein Gedi Resort at the Dead Sea, Israel.
Footnotes
The contents do not represent the views of the U.S. Department of Veterans Affairs or the United States Government.
Disclosures: EAL and JEB have a financial interest in OXT Therapeutics, Inc., a company developing an intranasal oxytocin and long-acting analogs of oxytocin to treat obesity and metabolic disease. Part of AW’s research (not related to the current manuscript) is funded by Europacific Medical, Inc. The authors’ interests were reviewed and are managed by their local institutions in accordance with their conflict of interest policies.
REFERENCES
- 1.Altirriba J, Poher AL, Caillon A, Arsenijevic D, Veyrat-Durebex C, Lyautey J, Dulloo A, and Rohner-Jeanrenaud F. Divergent effects of oxytocin treatment of obese diabetic mice on adiposity and diabetes. Endocrinology 155: 4189–4201, 2014. [DOI] [PubMed] [Google Scholar]
- 2.Amico JA, Vollmer RR, Cai HM, Miedlar JA, and Rinaman L. Enhanced initial and sustained intake of sucrose solution in mice with an oxytocin gene deletion. Am J Physiol Regul Integr Comp Physiol 289: R1798–1806, 2005. [DOI] [PubMed] [Google Scholar]
- 3.Antunes JL and Zimmerman EA. The hypothalamic magnocellular system of the rhesus monkey: an immunocytochemical study. The Journal of comparative neurology 181: 539–565, 1978. [DOI] [PubMed] [Google Scholar]
- 4.Arletti R, Benelli A, and Bertolini A. Influence of oxytocin on feeding behavior in the rat. Peptides 10: 89–93, 1989. [DOI] [PubMed] [Google Scholar]
- 5.Arletti R, Benelli A, and Bertolini A. Oxytocin inhibits food and fluid intake in rats. Physiology & behavior 48: 825–830, 1990. [DOI] [PubMed] [Google Scholar]
- 6.Atasoy D, Betley JN, Su HH, and Sternson SM. Deconstruction of a neural circuit for hunger. Nature 488: 172–177, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Aulinas A, Pulumo RL, Asanza E, Mancuso CJ, Slattery M, Tolley C, Plessow F, Thomas JJ, Eddy KT, Miller KK, Klibanski A, Misra M, and Lawson EA. Endogenous Oxytocin Levels in Relation to Food Intake, Menstrual Phase, and Age in Females. The Journal of clinical endocrinology and metabolism 104: 1348–1356, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Balazova L, Krskova K, Suski M, Sisovsky V, Hlavacova N, Olszanecki R, Jezova D, and Zorad S. Metabolic effects of subchronic peripheral oxytocin administration in lean and obese zucker rats. Journal of physiology and pharmacology : an official journal of the Polish Physiological Society 67: 531–541, 2016. [PubMed] [Google Scholar]
- 9.Barakat Y, Pape JR, Chaigniau M, El Ouezzani S, Alaoui A, Barakat L, and Magoul R. Oxytocin-containing neurons in the hypothalamic parvicellular paraventricular nucleus of the jerboa: no plasticity related to acute immobilization. Neuro endocrinology letters 29: 547–551, 2008. [PubMed] [Google Scholar]
- 10.Baskaran C, Plessow F, Silva L, Asanza E, Marengi D, Eddy KT, Sluss PM, Johnson ML, Misra M, and Lawson EA. Oxytocin secretion is pulsatile in men and is related to social-emotional functioning. Psychoneuroendocrinology 85: 28–34, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Baskin DG, Kim F, Gelling RW, Russell BJ, Schwartz MW, Morton GJ, Simhan HN, Moralejo DH, and Blevins JE. A new oxytocin-saporin cytotoxin for lesioning oxytocin-receptive neurons in the rat hindbrain. Endocrinology 151: 4207–4213, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Benelli A, Bertolini A, and Arletti R. Oxytocin-induced inhibition of feeding and drinking: no sexual dimorphism in rats. Neuropeptides 20: 57–62, 1991. [DOI] [PubMed] [Google Scholar]
- 13.Binay C, Paketci C, Guzel S, and Samanci N. Serum Irisin and Oxytocin Levels as Predictors of Metabolic Parameters in Obese Children. J Clin Res Pediatr Endocrinol 9: 124–131, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bjorkstrand E, Eriksson M, and Uvnas-Moberg K. Evidence of a peripheral and a central effect of oxytocin on pancreatic hormone release in rats. Neuroendocrinology 63: 377–383, 1996. [DOI] [PubMed] [Google Scholar]
- 15.Blevins JE, Eakin TJ, Murphy JA, Schwartz MW, and Baskin DG. Oxytocin innervation of caudal brainstem nuclei activated by cholecystokinin. Brain research 993: 30–41, 2003. [DOI] [PubMed] [Google Scholar]
- 16.Blevins JE, Graham JL, Morton GJ, Bales KL, Schwartz MW, Baskin DG, and Havel PJ. Chronic oxytocin administration inhibits food intake, increases energy expenditure, and produces weight loss in fructose-fed obese rhesus monkeys. American journal of physiology Regulatory, integrative and comparative physiology 308: R431–438, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Blevins JE, Schwartz MW, and Baskin DG. Evidence that paraventricular nucleus oxytocin neurons link hypothalamic leptin action to caudal brain stem nuclei controlling meal size. American journal of physiology Regulatory, integrative and comparative physiology 287: R87–96, 2004. [DOI] [PubMed] [Google Scholar]
- 18.Blevins JE, Thompson BW, Anekonda VT, Ho JM, Graham JL, Roberts ZS, Hwang BH, Ogimoto K, Wolden-hanson TH, Nelson JO, Kaiyala KJ, Havel PJ, Bales KL, Morton GJ, Schwartz MW, and Baskin DG. Chronic CNS oxytocin signaling preferentially induces fat loss in high fat diet-fed rats by enhancing satiety responses and increasing lipid utilization. American Journal of Physiology-Regulatory Integrative and Comparative Physiology, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Blouet C, Jo YH, Li X, and Schwartz GJ. Mediobasal hypothalamic leucine sensing regulates food intake through activation of a hypothalamus-brainstem circuit. The Journal of neuroscience : the official journal of the Society for Neuroscience 29: 8302–8311, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Boccia ML, Panicker AK, Pedersen C, and Petrusz P. Oxytocin receptors in non-human primate brain visualized with monoclonal antibody. Neuroreport 12: 1723–1726, 2001. [DOI] [PubMed] [Google Scholar]
- 21.Boccia ML, Petrusz P, Suzuki K, Marson L, and Pedersen CA. Immunohistochemical localization of oxytocin receptors in human brain. Neuroscience 253: 155–164, 2013. [DOI] [PubMed] [Google Scholar]
- 22.Brandtzaeg OK, Johnsen E, Roberg-Larsen H, Seip KF, MacLean EL, Gesquiere LR, Leknes S, Lundanes E, and Wilson SR. Proteomics tools reveal startlingly high amounts of oxytocin in plasma and serum. Scientific reports 6: 31693, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Brede S, Fehr S, Dalla-Man C, Cobelli C, Lehnert H, Hallschmid M, and Klement J. Intranasal oxytocin fails to acutely improve glucose metabolism in obese men. Diabetes, obesity & metabolism 21: 424–428, 2019. [DOI] [PubMed] [Google Scholar]
- 24.Brownstein MJ, Russell JT, and Gainer H. Synthesis, transport, and release of posterior pituitary hormones. Science 207: 373–378, 1980. [DOI] [PubMed] [Google Scholar]
- 25.Burmester V, Higgs S, and Terry P. Rapid-onset anorectic effects of intranasal oxytocin in young men. Appetite 130: 104–109, 2018. [DOI] [PubMed] [Google Scholar]
- 26.Burri A, Heinrichs M, Schedlowski M, and Kruger TH. The acute effects of intranasal oxytocin administration on endocrine and sexual function in males. Psychoneuroendocrinology 33: 591–600, 2008. [DOI] [PubMed] [Google Scholar]
- 27.Camerino C. Low sympathetic tone and obese phenotype in oxytocin-deficient mice. Obesity (Silver Spring) 17: 980–984, 2009. [DOI] [PubMed] [Google Scholar]
- 28.Castel M and Morris JF. The neurophysin-containing innervation of the forebrain of the mouse. Neuroscience 24: 937–966, 1988. [DOI] [PubMed] [Google Scholar]
- 29.Chang SW and Platt ML. Oxytocin and social cognition in rhesus macaques: implications for understanding and treating human psychopathology. Brain research 1580: 57–68, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chelikani PK, Haver AC, Reeve JR Jr., Keire DA, and Reidelberger RD. Daily, intermittent intravenous infusion of peptide YY(3–36) reduces daily food intake and adiposity in rats. American journal of physiology Regulatory, integrative and comparative physiology 290: R298–305, 2006. [DOI] [PubMed] [Google Scholar]
- 31.Chelikani PK, Haver AC, and Reidelberger RD. Effects of intermittent intraperitoneal infusion of salmon calcitonin on food intake and adiposity in obese rats. American journal of physiology Regulatory, integrative and comparative physiology 293: R1798–1808, 2007. [DOI] [PubMed] [Google Scholar]
- 32.Chelikani PK, Haver AC, and Reidelberger RD. Intermittent intraperitoneal infusion of peptide YY(3–36) reduces daily food intake and adiposity in obese rats. American journal of physiology Regulatory, integrative and comparative physiology 293: R39–46, 2007. [DOI] [PubMed] [Google Scholar]
- 33.Dal Monte O, Noble PL, Turchi J, Cummins A, and Averbeck BB. CSF and blood oxytocin concentration changes following intranasal delivery in macaque. PloS one 9: e103677, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Deblon N, Veyrat-Durebex C, Bourgoin L, Caillon A, Bussier AL, Petrosino S, Piscitelli F, Legros JJ, Geenen V, Foti M, Wahli W, Di Marzo V, and Rohner-Jeanrenaud F. Mechanisms of the anti-obesity effects of oxytocin in diet-induced obese rats. PloS one 6: e25565, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Dierickx K and Vandesande F. Immunocytochemical localization of the vasopressinergic and the oxytocinergic neurons in the human hypothalamus. Cell and tissue research 184: 15–27, 1977. [DOI] [PubMed] [Google Scholar]
- 36.Dunning BE, Moltz JH, and Fawcett CP. Actions of neurohypophysial peptides on pancreatic hormone release. The American journal of physiology 246: E108–114, 1984. [DOI] [PubMed] [Google Scholar]
- 37.Dunning BE, Moltz JH, and Fawcett CP. Modulation of insulin and glucagon secretion from the perfused rat pancreas by the neurohypophysial hormones and by desamino-D-arginine vasopressin (DDAVP). Peptides 5: 871–875, 1984. [DOI] [PubMed] [Google Scholar]
- 38.Eckertova M, Ondrejcakova M, Krskova K, Zorad S, and Jezova D. Subchronic treatment of rats with oxytocin results in improved adipocyte differentiation and increased gene expression of factors involved in adipogenesis. British journal of pharmacology 162: 452–463, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fabian M, Forsling ML, Jones JJ, and Pryor JS. The clearance and antidiuretic potency of neurohypophysial hormones in man, and their plasma binding and stability. The Journal of physiology 204: 653–668, 1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fenselau H, Campbell JN, Verstegen AM, Madara JC, Xu J, Shah BP, Resch JM, Yang Z, Mandelblat-Cerf Y, Livneh Y, and Lowell BB. A rapidly acting glutamatergic ARC-->PVH satiety circuit postsynaptically regulated by alpha-MSH. Nature neuroscience 20: 42–51, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Flanagan LM, Blackburn RE, Verbalis JG, and Stricker EM. Hypertonic NaCl inhibits gastric motility and food intake in rats with lesions in the rostral AV3V region. The American journal of physiology 263: R9–14, 1992. [DOI] [PubMed] [Google Scholar]
- 42.Freeman SM, Inoue K, Smith AL, Goodman MM, and Young LJ. The neuroanatomical distribution of oxytocin receptor binding and mRNA in the male rhesus macaque (Macaca mulatta). Psychoneuroendocrinology 45: 128–141, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Freeman SM, Ngo J, Singh B, Masnaghetti M, Bales KL, and Blevins JE. Effects of Chronic Oxytocin Administration and Diet Composition on Oxytocin and Vasopressin 1a Receptor Binding in the Rat Brain. Neuroscience 392: 241–251, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Freeman SM, Samineni S, Allen PC, Stockinger D, Bales KL, Hwa GG, and Roberts JA. Plasma and CSF oxytocin levels after intranasal and intravenous oxytocin in awake macaques. Psychoneuroendocrinology 66: 185–194, 2016. [DOI] [PubMed] [Google Scholar]
- 45.Freund-Mercier MJ, Stoeckel ME, and Klein MJ. Oxytocin receptors on oxytocin neurones: histoautoradiographic detection in the lactating rat. The Journal of physiology 480 (Pt 1): 155–161, 1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fu-Man D, Hong-Yu K, Bin-Hong D, Da-Na L, and Xin-Yang Y. Associations of oxytocin with metabolic parameters in obese women of childbearing age. Endokrynol Pol, 2019. [DOI] [PubMed] [Google Scholar]
- 47.Fuchs AR, Romero R, Keefe D, Parra M, Oyarzun E, and Behnke E. Oxytocin secretion and human parturition: pulse frequency and duration increase during spontaneous labor in women. Am J Obstet Gynecol 165: 1515–1523, 1991. [DOI] [PubMed] [Google Scholar]
- 48.Gajdosechova L, Krskova K, Segarra AB, Spolcova A, Suski M, Olszanecki R, and Zorad S. Hypooxytocinaemia in obese Zucker rats relates to oxytocin degradation in liver and adipose tissue. The Journal of endocrinology 220: 333–343, 2014. [DOI] [PubMed] [Google Scholar]
- 49.Gao ZY, Drews G, and Henquin JC. Mechanisms of the stimulation of insulin release by oxytocin in normal mouse islets. The Biochemical journal 276 (Pt 1): 169–174, 1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.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, and Lowell BB. A neural basis for melanocortin-4 receptor-regulated appetite. Nature neuroscience 18: 863–871, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gartner SN, Aidney F, Klockars A, Prosser C, Carpenter EA, Isgrove K, Levine AS, and Olszewski PK. Intragastric preloads of l-tryptophan reduce ingestive behavior via oxytocinergic neural mechanisms in male mice. Appetite 125: 278–286, 2018. [DOI] [PubMed] [Google Scholar]
- 52.Gelez H, Poirier S, Facchinetti P, Allers KA, Wayman C, Alexandre L, and Giuliano F. Neuroanatomical evidence for a role of central melanocortin-4 receptors and oxytocin in the efferent control of the rodent clitoris and vagina. The journal of sexual medicine 7: 2056–2067, 2010. [DOI] [PubMed] [Google Scholar]
- 53.George JM. Immunoreactive vasopressin and oxytocin: concentration in individual human hypothalamic nuclei. Science 200: 342–343, 1978. [DOI] [PubMed] [Google Scholar]
- 54.Gimpl G and Fahrenholz F. The oxytocin receptor system: structure, function, and regulation. Physiological reviews 81: 629–683, 2001. [DOI] [PubMed] [Google Scholar]
- 55.Ginsberg SD, Hof PR, Young WG, and Morrison JH. Noradrenergic innervation of vasopressin- and oxytocin-containing neurons in the hypothalamic paraventricular nucleus of the macaque monkey: quantitative analysis using double-label immunohistochemistry and confocal laser microscopy. The Journal of comparative neurology 341: 476–491, 1994. [DOI] [PubMed] [Google Scholar]
- 56.Gould BR and Zingg HH. Mapping oxytocin receptor gene expression in the mouse brain and mammary gland using an oxytocin receptor-LacZ reporter mouse. Neuroscience 122: 155–167, 2003. [DOI] [PubMed] [Google Scholar]
- 57.Greenfield JR, Miller JW, Keogh JM, Henning E, Satterwhite JH, Cameron GS, Astruc B, Mayer JP, Brage S, See TC, Lomas DJ, O’Rahilly S, and Farooqi IS. Modulation of blood pressure by central melanocortinergic pathways. The New England journal of medicine 360: 44–52, 2009. [DOI] [PubMed] [Google Scholar]
- 58.Gutkowska J and Jankowski M. Oxytocin revisited: its role in cardiovascular regulation. Journal of neuroendocrinology 24: 599–608, 2012. [DOI] [PubMed] [Google Scholar]
- 59.Hakansson ML, Brown H, Ghilardi N, Skoda RC, and Meister B. Leptin receptor immunoreactivity in chemically defined target neurons of the hypothalamus. J Neurosci 18: 559–572, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hashimoto H, Hyodo S, Kawasaki M, Mera T, Chen L, Soya A, Saito T, Fujihara H, Higuchi T, Takei Y, and Ueta Y. Centrally administered adrenomedullin 2 activates hypothalamic oxytocin-secreting neurons, causing elevated plasma oxytocin level in rats. American journal of physiology Endocrinology and metabolism 289: E753–761, 2005. [DOI] [PubMed] [Google Scholar]
- 61.Head MA, Jewett DC, Gartner SN, Klockars A, Levine AS, and Olszewski PK. Effect of Oxytocin on Hunger Discrimination. Front Endocrinol (Lausanne) 10: 297, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Henry KE, Elfers CT, Burke RM, Chepurny OG, Holz GG, Blevins JE, Roth CL, and Doyle RP. Vitamin B12 conjugation of peptide-YY(3–36) decreases food intake compared to native peptide-YY(3–36) upon subcutaneous administration in male rats. Endocrinology 156: 1739–1749, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Herisson FM, Brooks LL, Waas JR, Levine AS, and Olszewski PK. Functional relationship between oxytocin and appetite for carbohydrates versus saccharin. Neuroreport 25: 909–914, 2014. [DOI] [PubMed] [Google Scholar]
- 64.Herisson FM, Waas JR, Fredriksson R, Schioth HB, Levine AS, and Olszewski PK. Oxytocin Acting in the Nucleus Accumbens Core Decreases Food Intake. Journal of neuroendocrinology 28, 2016. [DOI] [PubMed] [Google Scholar]
- 65.Hicks C, Ramos L, Reekie T, Misagh GH, Narlawar R, Kassiou M, and McGregor IS. Body temperature and cardiac changes induced by peripherally administered oxytocin, vasopressin and the non-peptide oxytocin receptor agonist WAY 267,464: a biotelemetry study in rats. British journal of pharmacology 171: 2868–2887, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hidema S, Fukuda T, Hiraoka Y, Mizukami H, Hayashi R, Otsuka A, Suzuki S, Miyazaki S, and Nishimori K. Generation of Oxtr cDNA(HA)-Ires-Cre Mice for Gene Expression in an Oxytocin Receptor Specific Manner. Journal of cellular biochemistry 117: 1099–1111, 2016. [DOI] [PubMed] [Google Scholar]
- 67.Ho JM, Anekonda VT, Thompson BW, Zhu M, Curry RW, Hwang BH, Morton GJ, Schwartz MW, Baskin DG, Appleyard SM, and Blevins JE. Hindbrain oxytocin receptors contribute to the effects of circulating oxytocin on food intake in male rats. Endocrinology 155: 2845–2857, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hornby PJ. Central neurocircuitry associated with emesis. The American journal of medicine 111 Suppl 8A: 106S–112S, 2001. [DOI] [PubMed] [Google Scholar]
- 69.Hsu EA, Miller JL, Perez FA, and Roth CL. Oxytocin and Naltrexone successfully treat hypothalamic obesity in a boy post-craniopharyngioma resection. The Journal of clinical endocrinology and metabolism, 2017. [DOI] [PubMed] [Google Scholar]
- 70.Hunter DJ, Schulz P, and Wassenaar W. Effect of carbetocin, a long-acting oxytocin analog on the postpartum uterus. Clinical pharmacology and therapeutics 52: 60–67, 1992. [DOI] [PubMed] [Google Scholar]
- 71.Ichinose W, Cherepanov SM, Shabalova AA, Yokoyama S, Yuhi T, Yamaguchi H, Watanabe A, Yamamoto Y, Okamoto H, Horike S, Terakawa J, Daikoku T, Watanabe M, Mano N, Higashida H, and Shuto S. Development of a Highly Potent Analogue and a Long-Acting Analogue of Oxytocin for the Treatment of Social Impairment-Like Behaviors. Journal of medicinal chemistry 62: 3297–3310, 2019. [DOI] [PubMed] [Google Scholar]
- 72.Insel TR, Winslow JT, and Witt DM. Homologous regulation of brain oxytocin receptors. Endocrinology 130: 2602–2608, 1992. [DOI] [PubMed] [Google Scholar]
- 73.Iwasa T, Matsuzaki T, Mayila Y, Kawakita T, Yanagihara R, and Irahara M. The effects of chronic oxytocin administration on body weight and food intake in DHT-induced PCOS model rats. Gynecological endocrinology : the official journal of the International Society of Gynecological Endocrinology: 1–6, 2019. [DOI] [PubMed] [Google Scholar]
- 74.Iwasa T, Matsuzaki T, Mayila Y, Yanagihara R, Yamamoto Y, Kawakita T, Kuwahara A, and Irahara M. Oxytocin treatment reduced food intake and body fat and ameliorated obesity in ovariectomized female rats. Neuropeptides 75: 49–57, 2019. [DOI] [PubMed] [Google Scholar]
- 75.Iwasaki Y, Kumari P, Wang L, Hidema S, Nishimori K, and Yada T. Relay of peripheral oxytocin to central oxytocin neurons via vagal afferents for regulating feeding. Biochemical and biophysical research communications, 2019. [DOI] [PubMed] [Google Scholar]
- 76.Iwasaki Y, Maejima Y, Suyama S, Yoshida M, Arai T, Katsurada K, Kumari P, Nakabayashi H, Kakei M, and Yada T. Peripheral oxytocin activates vagal afferent neurons to suppress feeding in normal and leptinresistant mice: a route for ameliorating hyperphagia and obesity. American journal of physiology Regulatory, integrative and comparative physiology 308: R360–369, 2015. [DOI] [PubMed] [Google Scholar]
- 77.Jansen AS, Wessendorf MW, and Loewy AD. Transneuronal labeling of CNS neuropeptide and monoamine neurons after pseudorabies virus injections into the stellate ganglion. Brain research 683: 1–24, 1995. [DOI] [PubMed] [Google Scholar]
- 78.Jarrett TM, McMurray MS, Walker CH, and Johns JM. Cocaine treatment alters oxytocin receptor binding but not mRNA production in postpartum rat dams. Neuropeptides 40: 161–167, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Johnstone LE, Fong TM, and Leng G. Neuronal activation in the hypothalamus and brainstem during feeding in rats. Cell metabolism 4: 313–321, 2006. [DOI] [PubMed] [Google Scholar]
- 80.Kasahara Y, Sato K, Takayanagi Y, Mizukami H, Ozawa K, Hidema S, So KH, Kawada T, Inoue N, Ikeda I, Roh SG, Itoi K, and Nishimori K. Oxytocin receptor in the hypothalamus is sufficient to rescue normal thermoregulatory function in male oxytocin receptor knockout mice. Endocrinology 154: 4305–4315, 2013. [DOI] [PubMed] [Google Scholar]
- 81.Kasahara Y, Takayanagi Y, Kawada T, Itoi K, and Nishimori K. Impaired thermoregulatory ability of oxytocin-deficient mice during cold-exposure. Bioscience, biotechnology, and biochemistry 71: 3122–3126, 2007. [DOI] [PubMed] [Google Scholar]
- 82.Kasahara Y, Tateishi Y, Hiraoka Y, Otsuka A, Mizukami H, Ozawa K, Sato K, Hidema S, and Nishimori K. Role of the Oxytocin Receptor Expressed in the Rostral Medullary Raphe in Thermoregulation During Cold Conditions. Frontiers in endocrinology 6: 180, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kasiske BL, O’Donnell MP, and Keane WF. The obese Zucker rat model of glomerular injury in type II diabetes. The Journal of diabetic complications 1: 26–29, 1987. [DOI] [PubMed] [Google Scholar]
- 84.Kataoka N, Hioki H, Kaneko T, and Nakamura K. Psychological stress activates a dorsomedial hypothalamus-medullary raphe circuit driving brown adipose tissue thermogenesis and hyperthermia. Cell metabolism 20: 346–358, 2014. [DOI] [PubMed] [Google Scholar]
- 85.Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, and Natori T. Spontaneous long-term hyperglycemic rat with diabetic complications. Otsuka Long-Evans Tokushima Fatty (OLETF) strain. Diabetes 41: 1422–1428, 1992. [DOI] [PubMed] [Google Scholar]
- 86.Kawata M and Sano Y. Immunohistochemical identification of the oxytocin and vasopressin neurons in the hypothalamus of the monkey (Macaca fuscata). Anatomy and embryology 165: 151–167, 1982. [DOI] [PubMed] [Google Scholar]
- 87.Kendrick KM, Keverne EB, Baldwin BA, and Sharman DF. Cerebrospinal fluid levels of acetylcholinesterase, monoamines and oxytocin during labour, parturition, vaginocervical stimulation, lamb separation and suckling in sheep. Neuroendocrinology 44: 149–156, 1986. [DOI] [PubMed] [Google Scholar]
- 88.Kievit P, Halem H, Marks DL, Dong JZ, Glavas MM, Sinnayah P, Pranger L, Cowley MA, Grove KL, and Culler MD. Chronic treatment with a melanocortin-4 receptor agonist causes weight loss, reduces insulin resistance, and improves cardiovascular function in diet-induced obese rhesus macaques. Diabetes 62: 490–497, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kim YJ, Schroeder M, Liang NC, Moran TH, Weller A, and Bi S. Effects of early postnatal environment on hypothalamic gene expression in OLETF rats. PloS one 12: e0178428, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kirchgessner AL, Sclafani A, and Nilaver G. Histochemical identification of a PVN-hindbrain feeding pathway. Physiology & behavior 42: 529–543, 1988. [DOI] [PubMed] [Google Scholar]
- 91.Kirkpatrick MG, Francis SM, Lee R, de Wit H, and Jacob S. Plasma oxytocin concentrations following MDMA or intranasal oxytocin in humans. Psychoneuroendocrinology 46: 23–31, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Klement J, Ott V, Rapp K, Brede S, Piccinini F, Cobelli C, Lehnert H, and Hallschmid M. Oxytocin Improves beta-Cell Responsivity and Glucose Tolerance in Healthy Men. Diabetes 66: 264–271, 2017. [DOI] [PubMed] [Google Scholar]
- 93.Klockars A, Brunton C, Li L, Levine AS, and Olszewski PK. Intravenous administration of oxytocin in rats acutely decreases deprivation-induced chow intake, but it fails to affect consumption of palatable solutions. Peptides 93: 13–19, 2017. [DOI] [PubMed] [Google Scholar]
- 94.Klockars OA, Klockars A, Levine AS, and Olszewski PK. Oxytocin administration in the basolateral and central nuclei of amygdala moderately suppresses food intake. Neuroreport 29: 504–510, 2018. [DOI] [PubMed] [Google Scholar]
- 95.Klockars OA, Waas JR, Klockars A, Levine AS, and Olszewski PK. Neural Basis of Ventromedial Hypothalamic Oxytocin-Driven Decrease in Appetite. Neuroscience 366: 54–61, 2017. [DOI] [PubMed] [Google Scholar]
- 96.Kohno D, Nakata M, Maejima Y, Shimizu H, Sedbazar U, Yoshida N, Dezaki K, Onaka T, Mori M, and Yada T. Nesfatin-1 neurons in paraventricular and supraoptic nuclei of the rat hypothalamus coexpress oxytocin and vasopressin and are activated by refeeding. Endocrinology 149: 1295–1301, 2008. [DOI] [PubMed] [Google Scholar]
- 97.Koutcherov Y, Mai JK, Ashwell KW, and Paxinos G. Organization of the human paraventricular hypothalamic nucleus. The Journal of comparative neurology 423: 299–318, 2000. [PubMed] [Google Scholar]
- 98.Kublaoui BM, Gemelli T, Tolson KP, Wang Y, and Zinn AR. Oxytocin deficiency mediates hyperphagic obesity of Sim1 haploinsufficient mice. Mol Endocrinol 22: 1723–1734, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kujath AS, Quinn L, Elliott ME, Varady KA, LeCaire TJ, Carter CS, and Danielson KK. Oxytocin levels are lower in premenopausal women with type 1 diabetes mellitus compared with matched controls. Diabetes Metab Res Rev 31: 102–112, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kuo JJ, Silva AA, and Hall JE. Hypothalamic melanocortin receptors and chronic regulation of arterial pressure and renal function. Hypertension 41: 768–774, 2003. [DOI] [PubMed] [Google Scholar]
- 101.Labyb M, Chretien C, Caillon A, Rohner-Jeanrenaud F, and Altirriba J. Oxytocin Administration Alleviates Acute but Not Chronic Leptin Resistance of Diet-Induced Obese Mice. International journal of molecular sciences 20, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lawson EA. The effects of oxytocin on eating behaviour and metabolism in humans. Nature reviews Endocrinology 13: 700–709, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lawson EA, Ackerman KE, Estella NM, Guereca G, Pierce L, Sluss PM, Bouxsein ML, Klibanski A, and Misra M. Nocturnal oxytocin secretion is lower in amenorrheic athletes than nonathletes and associated with bone microarchitecture and finite element analysis parameters. European journal of endocrinology / European Federation of Endocrine Societies 168: 457–464, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lawson EA, Donoho DA, Blum JI, Meenaghan EM, Misra M, Herzog DB, Sluss PM, Miller KK, and Klibanski A. Decreased nocturnal oxytocin levels in anorexia nervosa are associated with low bone mineral density and fat mass. The Journal of clinical psychiatry 72: 1546–1551, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lawson EA, Marengi DA, DeSanti RL, Holmes TM, Schoenfeld DA, and Tolley CJ. Oxytocin reduces caloric intake in men. Obesity (Silver Spring), 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Lee MR, Scheidweiler KB, Diao XX, Akhlaghi F, Cummins A, Huestis MA, Leggio L, and Averbeck BB. Oxytocin by intranasal and intravenous routes reaches the cerebrospinal fluid in rhesus macaques: determination using a novel oxytocin assay. Molecular psychiatry 23: 115–122, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Leibowitz SF, Hammer NJ, and Chang K. Hypothalamic paraventricular nucleus lesions produce overeating and obesity in the rat. Physiol Behav 27: 1031–1040, 1981. [DOI] [PubMed] [Google Scholar]
- 108.Leng G and Ludwig M. Intranasal Oxytocin: Myths and Delusions. Biological psychiatry 79: 243–250, 2016. [DOI] [PubMed] [Google Scholar]
- 109.Leng G, Onaka T, Caquineau C, Sabatier N, Tobin VA, and Takayanagi Y. Oxytocin and appetite. Progress in brain research 170: 137–151, 2008. [DOI] [PubMed] [Google Scholar]
- 110.Leng G and Sabatier N. Measuring Oxytocin and Vasopressin: Bioassays, Immunoassays and Random Numbers. Journal of neuroendocrinology 28, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li C, Navarrete J, Liang-Guallpa J, Lu C, Funderburk SC, Chang RB, Liberles SD, Olson DP, and Krashes MJ. Defined Paraventricular Hypothalamic Populations Exhibit Differential Responses to Food Contingent on Caloric State. Cell metabolism 29: 681–694 e685, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Liu CY, Chen LB, Liu PY, Xie DP, and Wang PS. Effects of progesterone on gastric emptying and intestinal transit in male rats. World journal of gastroenterology 8: 338–341, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Liu H, Kishi T, Roseberry AG, Cai X, Lee CE, Montez JM, Friedman JM, and Elmquist JK. Transgenic mice expressing green fluorescent protein under the control of the melanocortin-4 receptor promoter. The Journal of neuroscience : the official journal of the Society for Neuroscience 23: 7143–7154, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Loup F, Tribollet E, Dubois-Dauphin M, and Dreifuss JJ. Localization of high-affinity binding sites for oxytocin and vasopressin in the human brain. An autoradiographic study. Brain research 555: 220–232, 1991. [DOI] [PubMed] [Google Scholar]
- 115.Loup F, Tribollet E, Dubois-Dauphin M, Pizzolato G, and Dreifuss JJ. Localization of oxytocin binding sites in the human brainstem and upper spinal cord: an autoradiographic study. Brain research 500: 223–230, 1989. [DOI] [PubMed] [Google Scholar]
- 116.Lucio-Oliveira F and Franci CR. Effect of the interaction between food state and the action of estrogen on oxytocinergic system activity. The Journal of endocrinology 212: 129–138, 2012. [DOI] [PubMed] [Google Scholar]
- 117.Ludwig M and Leng G. Dendritic peptide release and peptide-dependent behaviours. Nature reviews Neuroscience 7: 126–136, 2006. [DOI] [PubMed] [Google Scholar]
- 118.Ludwig M, Tobin VA, Callahan MF, Papadaki E, Becker A, Engelmann M, and Leng G. Intranasal application of vasopressin fails to elicit changes in brain immediate early gene expression, neural activity and behavioural performance of rats. Journal of neuroendocrinology 25: 655–667, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.MacLean EL, Wilson SR, Martin WL, Davis JM, Nazarloo HP, and Carter CS. Challenges for measuring oxytocin: The blind men and the elephant? Psychoneuroendocrinology 107: 225–231, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Maejima Y, Aoyama M, Sakamoto K, Jojima T, Aso Y, Takasu K, Takenosihita S, and Shimomura K. Impact of sex, fat distribution and initial body weight on oxytocin’s body weight regulation. Scientific reports 7: 8599, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Maejima Y, Iwasaki Y, Yamahara Y, Kodaira M, Sedbazar U, and Yada T. Peripheral oxytocin treatment ameliorates obesity by reducing food intake and visceral fat mass. Aging 3: 1169–1177, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Maejima Y, Rita RS, Santoso P, Aoyama M, Hiraoka Y, Nishimori K, Gantulga D, Shimomura K, and Yada T. Nasal oxytocin administration reduces food intake without affecting locomotor activity and glycemia with c-Fos induction in limited brain areas. Neuroendocrinology 101: 35–44, 2015. [DOI] [PubMed] [Google Scholar]
- 123.Maejima Y, Sakuma K, Santoso P, Gantulga D, Katsurada K, Ueta Y, Hiraoka Y, Nishimori K, Tanaka S, Shimomura K, and Yada T. Oxytocinergic circuit from paraventricular and supraoptic nuclei to arcuate POMC neurons in hypothalamus. FEBS letters 588: 4404–4412, 2014. [DOI] [PubMed] [Google Scholar]
- 124.Maejima Y, Sedbazar U, Suyama S, Kohno D, Onaka T, Takano E, Yoshida N, Koike M, Uchiyama Y, Fujiwara K, Yashiro T, Horvath TL, Dietrich MO, Tanaka S, Dezaki K, Oh IS, Hashimoto K, Shimizu H, Nakata M, Mori M, and Yada T. Nesfatin-1-regulated oxytocinergic signaling in the paraventricular nucleus causes anorexia through a leptin-independent melanocortin pathway. Cell metabolism 10: 355–365, 2009. [DOI] [PubMed] [Google Scholar]
- 125.Maestrini S, Mele C, Mai S, Vietti R, Di Blasio A, Castello L, Surico D, Aimaretti G, Scacchi M, and Marzullo P. Plasma Oxytocin Concentration in Pre- and Postmenopausal Women: Its Relationship with Obesity, Body Composition and Metabolic Variables. Obes Facts 11: 429–439, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Manning M, Misicka A, Olma A, Bankowski K, Stoev S, Chini B, Durroux T, Mouillac B, Corbani M, and Guillon G. Oxytocin and vasopressin agonists and antagonists as research tools and potential therapeutics. Journal of neuroendocrinology 24: 609–628, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Martin R, Geis R, Holl R, Schafer M, and Voigt KH. Co-existence of unrelated peptides in oxytocin and vasopressin terminals of rat neurohypophyses: immunoreactive methionine-enkephalin-, leucine-enkephalin- and cholecystokinin-like substances. Neuroscience 8: 213–227, 1983. [DOI] [PubMed] [Google Scholar]
- 128.Matarazzo V, Schaller F, Nedelec E, Benani A, Penicaud L, Muscatelli F, Moyse E, and Bauer S. Inactivation of Socs3 in the hypothalamus enhances the hindbrain response to endogenous satiety signals via oxytocin signaling. The Journal of neuroscience : the official journal of the Society for Neuroscience 32: 17097–17107, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.McCullough ME, Churchland PS, and Mendez AJ. Problems with measuring peripheral oxytocin: can the data on oxytocin and human behavior be trusted? Neuroscience and biobehavioral reviews 37: 1485–1492, 2013. [DOI] [PubMed] [Google Scholar]
- 130.Mens WB, Witter A, and van Wimersma Greidanus TB. Penetration of neurohypophyseal hormones from plasma into cerebrospinal fluid (CSF): half-times of disappearance of these neuropeptides from CSF. Brain research 262: 143–149, 1983. [DOI] [PubMed] [Google Scholar]
- 131.Miaskiewicz SL, Stricker EM, and Verbalis JG. Neurohypophyseal secretion in response to cholecystokinin but not meal-induced gastric distention in humans. The Journal of clinical endocrinology and metabolism 68: 837–843, 1989. [DOI] [PubMed] [Google Scholar]
- 132.Miedlar JA, Rinaman L, Vollmer RR, and Amico JA. Oxytocin gene deletion mice overconsume palatable sucrose solution but not palatable lipid emulsions. American journal of physiology Regulatory, integrative and comparative physiology 293: R1063–1068, 2007. [DOI] [PubMed] [Google Scholar]
- 133.Mitra A, Gosnell BA, Schioth HB, Grace MK, Klockars A, Olszewski PK, and Levine AS. Chronic sugar intake dampens feeding-related activity of neurons synthesizing a satiety mediator, oxytocin. Peptides 31: 1346–1352, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Modi ME, Connor-Stroud F, Landgraf R, Young LJ, and Parr LA. Aerosolized oxytocin increases cerebrospinal fluid oxytocin in rhesus macaques. Psychoneuroendocrinology 45: 49–57, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Modi ME, Inoue K, Barrett CE, Kittelberger KA, Smith DG, Landgraf R, and Young LJ. Melanocortin Receptor Agonists Facilitate Oxytocin-Dependent Partner Preference Formation in the Prairie Vole. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 40: 1856–1865, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Monteleone AM, Scognamiglio P, Volpe U, Di Maso V, and Monteleone P. Investigation of Oxytocin Secretion in Anorexia Nervosa and Bulimia Nervosa: Relationships to Temperament Personality Dimensions. Eur Eat Disord Rev 24: 52–56, 2016. [DOI] [PubMed] [Google Scholar]
- 137.Morton GJ, Thatcher BS, Reidelberger RD, Ogimoto K, Wolden-Hanson T, Baskin DG, Schwartz MW, and Blevins JE. Peripheral oxytocin suppresses food intake and causes weight loss in diet-induced obese rats. Am J Physiol-Endoc M 302: E134–E144, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Mullis K, Kay K, and Williams DL. Oxytocin action in the ventral tegmental area affects sucrose intake. Brain research 1513: 85–91, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Murabe M, Yamauchi J, Fujiwara Y, Miyamoto Y, Hiroyama M, Sanbe A, and Tanoue A. Estimation of the embryotoxic effect of CBZ using an ES cell differentiation system. Biochemical and biophysical research communications 356: 739–744, 2007. [DOI] [PubMed] [Google Scholar]
- 140.Muttenthaler M, Andersson A, de Araujo AD, Dekan Z, Lewis RJ, and Alewood PF. Modulating oxytocin activity and plasma stability by disulfide bond engineering. Journal of medicinal chemistry 53: 8585–8596, 2010. [DOI] [PubMed] [Google Scholar]
- 141.Nasanbuyan N, Yoshida M, Takayanagi Y, Inutsuka A, Nishimori K, Yamanaka A, and Onaka T. Oxytocin-Oxytocin Receptor Systems Facilitate Social Defeat Posture in Male Mice. Endocrinology 159: 763–775, 2018. [DOI] [PubMed] [Google Scholar]
- 142.Nation DA, Szeto A, Mendez AJ, Brooks LG, Zaias J, Herderick EE, Gonzales J, Noller CM, Schneiderman N, and McCabe PM. Oxytocin attenuates atherosclerosis and adipose tissue inflammation in socially isolated ApoE−/− mice. Psychosomatic medicine 72: 376–382, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Nelson EE, Alberts JR, Tian Y, and Verbalis JG. Oxytocin is elevated in plasma of 10-day-old rats following gastric distension. Brain research Developmental brain research 111: 301–303, 1998. [DOI] [PubMed] [Google Scholar]
- 144.Neumann ID, Maloumby R, Beiderbeck DI, Lukas M, and Landgraf R. Increased brain and plasma oxytocin after nasal and peripheral administration in rats and mice. Psychoneuroendocrinology 38: 1985–1993, 2013. [DOI] [PubMed] [Google Scholar]
- 145.Nguyen NT, Magno CP, Lane KT, Hinojosa MW, and Lane JS. Association of hypertension, diabetes, dyslipidemia, and metabolic syndrome with obesity: findings from the National Health and Nutrition Examination Survey, 1999 to 2004. Journal of the American College of Surgeons 207: 928–934, 2008. [DOI] [PubMed] [Google Scholar]
- 146.Nissen E, Uvnas-Moberg K, Svensson K, Stock S, Widstrom AM, and Winberg J. Different patterns of oxytocin, prolactin but not cortisol release during breastfeeding in women delivered by caesarean section or by the vaginal route. Early Hum Dev 45: 103–118, 1996. [DOI] [PubMed] [Google Scholar]
- 147.Noble EE, Billington CJ, Kotz CM, and Wang C. Oxytocin in the ventromedial hypothalamic nucleus reduces feeding and acutely increases energy expenditure. American journal of physiology Regulatory, integrative and comparative physiology 307: R737–745, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Northway MG, Morris M, Geisinger KR, and MacLean DB. Effects of a gastric implant on body weight and gastrointestinal hormones in cafeteria diet obese rats. Physiology & behavior 45: 331–335, 1989. [DOI] [PubMed] [Google Scholar]
- 149.Nussey SS, Hawthorn J, Page SR, Ang VT, and Jenkins JS. Responses of plasma oxytocin and arginine vasopressin to nausea induced by apomorphine and ipecacuanha. Clinical endocrinology 28: 297–304, 1988. [DOI] [PubMed] [Google Scholar]
- 150.Ohlsson B, Forsling ML, Rehfeld JF, and Sjolund K. Cholecystokinin stimulation leads to increased oxytocin secretion in women. The European journal of surgery = Acta chirurgica 168: 114–118, 2002. [DOI] [PubMed] [Google Scholar]
- 151.Oldfield BJ, Giles ME, Watson A, Anderson C, Colvill LM, and McKinley MJ. The neurochemical characterisation of hypothalamic pathways projecting polysynaptically to brown adipose tissue in the rat. Neuroscience 110: 515–526, 2002. [DOI] [PubMed] [Google Scholar]
- 152.Olson BR, Drutarosky MD, Chow MS, Hruby VJ, Stricker EM, and Verbalis JG. Oxytocin and an oxytocin agonist administered centrally decrease food intake in rats. Peptides 12: 113–118, 1991. [DOI] [PubMed] [Google Scholar]
- 153.Olson BR, Drutarosky MD, Stricker EM, and Verbalis JG. Brain oxytocin receptor antagonism blunts the effects of anorexigenic treatments in rats: evidence for central oxytocin inhibition of food intake. Endocrinology 129: 785–791, 1991. [DOI] [PubMed] [Google Scholar]
- 154.Olson BR, Freilino M, Hoffman GE, Stricker EM, Sved AF, and Verbalis JG. c-Fos Expression in Rat Brain and Brainstem Nuclei in Response to Treatments That Alter Food Intake and Gastric Motility. Molecular and cellular neurosciences 4: 93–106, 1993. [DOI] [PubMed] [Google Scholar]
- 155.Olson BR, Hoffman GE, Sved AF, Stricker EM, and Verbalis JG. Cholecystokinin induces c-fos expression in hypothalamic oxytocinergic neurons projecting to the dorsal vagal complex. Brain Res 569: 238–248, 1992. [DOI] [PubMed] [Google Scholar]
- 156.Olszewski PK, Klockars A, Olszewska AM, Fredriksson R, Schioth HB, and Levine AS. Molecular, immunohistochemical, and pharmacological evidence of oxytocin’s role as inhibitor of carbohydrate but not fat intake. Endocrinology 151: 4736–4744, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Olszewski PK and Levine AS. Central opioids and consumption of sweet tastants: when reward outweighs homeostasis. Physiol Behav 91: 506–512, 2007. [DOI] [PubMed] [Google Scholar]
- 158.Olszewski PK, Shi Q, Billington CJ, and Levine AS. Opioids affect acquisition of LiCl-induced conditioned taste aversion: involvement of OT and VP systems. Am J Physiol Regul Integr Comp Physiol 279: R1504–1511, 2000. [DOI] [PubMed] [Google Scholar]
- 159.Olszewski PK, Waas JR, Brooks LL, Herisson F, and Levine AS. Oxytocin receptor blockade reduces acquisition but not retrieval of taste aversion and blunts responsiveness of amygdala neurons to an aversive stimulus. Peptides 50: 36–41, 2013. [DOI] [PubMed] [Google Scholar]
- 160.Olszewski PK, Wirth MM, Shaw TJ, Grace MK, Billington CJ, Giraudo SQ, and Levine AS. Role of alpha-MSH in the regulation of consummatory behavior: immunohistochemical evidence. American journal of physiology Regulatory, integrative and comparative physiology 281: R673–680, 2001. [DOI] [PubMed] [Google Scholar]
- 161.Ong ZY, Alhadeff AL, and Grill HJ. Medial nucleus tractus solitarius oxytocin receptor signaling and food intake control: the role of gastrointestinal satiation signal processing. American journal of physiology Regulatory, integrative and comparative physiology 308: R800–806, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Ong ZY, Bongiorno DM, Hernando MA, and Grill HJ. Effects of Endogenous Oxytocin Receptor Signaling in Nucleus Tractus Solitarius on Satiation-Mediated Feeding and Thermogenic Control in Male Rats. Endocrinology 158: 2826–2836, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Oppong-Damoah A, Zaman RU, D’Souza MJ, and Murnane KS. Nanoparticle encapsulation increases the brain penetrance and duration of action of intranasal oxytocin. Hormones and behavior 108: 20–29, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Otsubo H, Hyodo S, Hashimoto H, Kawasaki M, Suzuki H, Saito T, Ohbuchi T, Yokoyama T, Fujihara H, Matsumoto T, Takei Y, and Ueta Y. Centrally administered adrenomedullin 5 activates oxytocin-secreting neurons in the hypothalamus and elevates plasma oxytocin level in rats. The Journal of endocrinology 202: 237–247, 2009. [DOI] [PubMed] [Google Scholar]
- 165.Ott V, Finlayson G, Lehnert H, Heitmann B, Heinrichs M, Born J, and Hallschmid M. Oxytocin reduces reward-driven food intake in humans. Diabetes 62: 3418–3425, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Pataky Z, Guessous I, Caillon A, Golay A, Rohner-Jeanrenaud F, and Altirriba J. Variable oxytocin levels in humans with different degrees of obesity and impact of gastric bypass surgery. Int J Obes (Lond) 43: 1120–1124, 2019. [DOI] [PubMed] [Google Scholar]
- 167.Paulin C, Dubois PM, Czernichow P, and Dubois MP. Immunocytological evidence for oxytocin neurons in the human fetal hypothalamus. Cell and tissue research 188: 259–264, 1978. [DOI] [PubMed] [Google Scholar]
- 168.Pei H, Sutton AK, Burnett KH, Fuller PM, and Olson DP. AVP neurons in the paraventricular nucleus of the hypothalamus regulate feeding. Molecular metabolism 3: 209–215, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Perello M and Raingo J. Leptin activates oxytocin neurons of the hypothalamic paraventricular nucleus in both control and diet-induced obese rodents. PloS one 8: e59625, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Peris J, MacFadyen K, Smith JA, de Kloet AD, Wang L, and Krause EG. Oxytocin receptors are expressed on dopamine and glutamate neurons in the mouse ventral tegmental area that project to nucleus accumbens and other mesolimbic targets. The Journal of comparative neurology 525: 1094–1108, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Peters S, Slattery DA, Uschold-Schmidt N, Reber SO, and Neumann ID. Dose-dependent effects of chronic central infusion of oxytocin on anxiety, oxytocin receptor binding and stress-related parameters in mice. Psychoneuroendocrinology 42: 225–236, 2014. [DOI] [PubMed] [Google Scholar]
- 172.Petersson M. Cardiovascular effects of oxytocin. Progress in brain research 139: 281–288, 2002. [DOI] [PubMed] [Google Scholar]
- 173.Petersson M, Alster P, Lundeberg T, and Uvnas-Moberg K. Oxytocin causes a long-term decrease of blood pressure in female and male rats. Physiology & behavior 60: 1311–1315, 1996. [DOI] [PubMed] [Google Scholar]
- 174.Phillips MS, Liu Q, Hammond HA, Dugan V, Hey PJ, Caskey CJ, and Hess JF. Leptin receptor missense mutation in the fatty Zucker rat. Nature genetics 13: 18–19, 1996. [DOI] [PubMed] [Google Scholar]
- 175.Pinol RA, Bateman R, and Mendelowitz D. Optogenetic approaches to characterize the long-range synaptic pathways from the hypothalamus to brain stem autonomic nuclei. Journal of neuroscience methods 210: 238–246, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Pinol RA, Jameson H, Popratiloff A, Lee NH, and Mendelowitz D. Visualization of oxytocin release that mediates paired pulse facilitation in hypothalamic pathways to brainstem autonomic neurons. PloS one 9: e112138, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Plante E, Menaouar A, Danalache BA, Yip D, Broderick TL, Chiasson JL, Jankowski M, and Gutkowska J. Oxytocin treatment prevents the cardiomyopathy observed in obese diabetic male db/db mice. Endocrinology 156: 1416–1428, 2015. [DOI] [PubMed] [Google Scholar]
- 178.Plessow F, Marengi DA, Perry SK, Felicione JM, Franklin R, Holmes TM, Holsen LM, Makris N, Deckersbach T, and Lawson EA. Effects of Intranasal Oxytocin on the Blood Oxygenation Level-Dependent Signal in Food Motivation and Cognitive Control Pathways in Overweight and Obese Men. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 43: 638–645, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Postina R, Kojro E, and Fahrenholz F. Separate agonist and peptide antagonist binding sites of the oxytocin receptor defined by their transfer into the V2 vasopressin receptor. The Journal of biological chemistry 271: 31593–31601, 1996. [DOI] [PubMed] [Google Scholar]
- 180.Pow DV and Morris JF. Dendrites of hypothalamic magnocellular neurons release neurohypophysial peptides by exocytosis. Neuroscience 32: 435–439, 1989. [DOI] [PubMed] [Google Scholar]
- 181.Qi J, Yang JY, Song M, Li Y, Wang F, and Wu CF. Inhibition by oxytocin of methamphetamine-induced hyperactivity related to dopamine turnover in the mesolimbic region in mice. Naunyn Schmiedebergs Arch Pharmacol 376: 441–448, 2008. [DOI] [PubMed] [Google Scholar]
- 182.Qian W, Zhu T, Tang B, Yu S, Hu H, Sun W, Pan R, Wang J, Wang D, Yang L, Mao C, Zhou L, and Yuan G. Decreased circulating levels of oxytocin in obesity and newly diagnosed type 2 diabetic patients. J Clin Endocrinol Metab 99: 4683–4689, 2014. [DOI] [PubMed] [Google Scholar]
- 183.Qin J, Feng M, Wang C, Ye Y, Wang PS, and Liu C. Oxytocin receptor expressed on the smooth muscle mediates the excitatory effect of oxytocin on gastric motility in rats. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society 21: 430–438, 2009. [DOI] [PubMed] [Google Scholar]
- 184.Ragen BJ and Bales KL. Oxytocin and vasopressin in non-human primates In: Oxytocin, vasopressin and related peptides in the regulation of behavior, edited by Choleris E, Pfaff DW and Kavaliers M. Cambridge, United Kingdom: Cambridge University Press, 2013, p. 288–306. [Google Scholar]
- 185.Rault JL. Effects of positive and negative human contacts and intranasal oxytocin on cerebrospinal fluid oxytocin. Psychoneuroendocrinology 69: 60–66, 2016. [DOI] [PubMed] [Google Scholar]
- 186.Reiter MK, Kremarik P, Freund-Mercier MJ, Stoeckel ME, Desaulles E, and Feltz P. Localization of oxytocin binding sites in the thoracic and upper lumbar spinal cord of the adult and postnatal rat: a histoautoradiographic study. The European journal of neuroscience 6: 98–104, 1994. [DOI] [PubMed] [Google Scholar]
- 187.Renaud LP and Bourque CW. Neurophysiology and neuropharmacology of hypothalamic magnocellular neurons secreting vasopressin and oxytocin. Progress in neurobiology 36: 131–169, 1991. [DOI] [PubMed] [Google Scholar]
- 188.Renaud LP, Tang M, McCann MJ, Stricker EM, and Verbalis JG. Cholecystokinin and gastric distension activate oxytocinergic cells in rat hypothalamus. The American journal of physiology 253: R661–665, 1987. [DOI] [PubMed] [Google Scholar]
- 189.Rinaman L. Oxytocinergic inputs to the nucleus of the solitary tract and dorsal motor nucleus of the vagus in neonatal rats. The Journal of comparative neurology 399: 101–109, 1998. [DOI] [PubMed] [Google Scholar]
- 190.Ring RH, Malberg JE, Potestio L, Ping J, Boikess S, Luo B, Schechter LE, Rizzo S, Rahman Z, and Rosenzweig-Lipson S. Anxiolytic-like activity of oxytocin in male mice: behavioral and autonomic evidence, therapeutic implications. Psychopharmacology 185: 218–225, 2006. [DOI] [PubMed] [Google Scholar]
- 191.Roberts ZS, Wolden-Hanson TH, Matsen ME, Ryu V, Vaughan CH, Graham JL, Havel PJ, Chukri DW, Schwartz MW, Morton GJ, and Blevins JE. Chronic Hindbrain Administration of Oxytocin is Sufficient to Elicit Weight Loss in Diet-Induced Obese Rats. American journal of physiology Regulatory, integrative and comparative physiology: ajpregu 00169 02017, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Rosen GJ, de Vries GJ, Goldman SL, Goldman BD, and Forger NG. Distribution of oxytocin in the brain of a eusocial rodent. Neuroscience 155: 809–817, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Ryan PJ, Ross SI, Campos CA, Derkach VA, and Palmiter RD. Oxytocin-receptor-expressing neurons in the parabrachial nucleus regulate fluid intake. Nature neuroscience 20: 1722–1733, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Sabatier N, Caquineau C, Dayanithi G, Bull P, Douglas AJ, Guan XM, Jiang M, Van der Ploeg L, and Leng G. Alpha-melanocyte-stimulating hormone stimulates oxytocin release from the dendrites of hypothalamic neurons while inhibiting oxytocin release from their terminals in the neurohypophysis. The Journal of neuroscience : the official journal of the Society for Neuroscience 23: 10351–10358, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Sabatier N, Leng G, and Menzies J. Oxytocin, feeding, and satiety. Frontiers in endocrinology 4: 35, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Sawchenko PE and Swanson LW. Immunohistochemical identification of neurons in the paraventricular nucleus of the hypothalamus that project to the medulla or to the spinal cord in the rat. The Journal of comparative neurology 205: 260–272, 1982. [DOI] [PubMed] [Google Scholar]
- 197.Sawchenko PE, Swanson LW, and Vale WW. Corticotropin-releasing factor: co-expression within distinct subsets of oxytocin-, vasopressin-, and neurotensin-immunoreactive neurons in the hypothalamus of the male rat. The Journal of neuroscience : the official journal of the Society for Neuroscience 4: 1118–1129, 1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Schaffler A, Binart N, Scholmerich J, and Buchler C. Hypothesis paper Brain talks with fat--evidence for a hypothalamic-pituitary-adipose axis? Neuropeptides 39: 363–367, 2005. [DOI] [PubMed] [Google Scholar]
- 199.Schorr M, Marengi DA, Pulumo RL, Yu E, Eddy KT, Klibanski A, Miller KK, and Lawson EA. Oxytocin and Its Relationship to Body Composition, Bone Mineral Density, and Hip Geometry Across the Weight Spectrum. The Journal of clinical endocrinology and metabolism 102: 2814–2824, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Schorscher-Petcu A, Dupre A, and Tribollet E. Distribution of vasopressin and oxytocin binding sites in the brain and upper spinal cord of the common marmoset. Neuroscience letters 461: 217–222, 2009. [DOI] [PubMed] [Google Scholar]
- 201.Schramme AR, Pinto CR, Davis J, Whisnant CS, and Whitacre MD. Pharmacokinetics of carbetocin, a long-acting oxytocin analogue, following intravenous administration in horses. Equine veterinary journal 40: 658–661, 2008. [DOI] [PubMed] [Google Scholar]
- 202.Schroeder M, Gelber V, Moran TH, and Weller A. Long-term obesity levels in female OLETF rats following time-specific post-weaning food restriction. Hormones and behavior 58: 844–853, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Schroeder M, Moran TH, and Weller A. Attenuation of obesity by early-life food restriction in genetically hyperphagic male OLETF rats: peripheral mechanisms. Hormones and behavior 57: 455–462, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Schroeder M, Zagoory-Sharon O, Shbiro L, Marco A, Hyun J, Moran TH, Bi S, and Weller A. Development of obesity in the Otsuka Long-Evans Tokushima Fatty rat. American journal of physiology Regulatory, integrative and comparative physiology 297: R1749–1760, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Sclafani A, Rinaman L, Vollmer RR, and Amico JA. Oxytocin knockout mice demonstrate enhanced intake of sweet and nonsweet carbohydrate solutions. American journal of physiology Regulatory, integrative and comparative physiology 292: R1828–1833, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Seelke AM, Rhine MA, Khun K, Shweyk AN, Scott AM, Bond JM, Graham JL, Havel PJ, Wolden-Hanson T, Bales KL, and Blevins JE. Intranasal oxytocin reduces weight gain in diet-induced obese prairie voles. Physiology & behavior 196: 67–77, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Shahrokh DK, Zhang TY, Diorio J, Gratton A, and Meaney MJ. Oxytocin-dopamine interactions mediate variations in maternal behavior in the rat. Endocrinology 151: 2276–2286, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Shi H and Bartness TJ. Neurochemical phenotype of sympathetic nervous system outflow from brain to white fat. Brain research bulletin 54: 375–385, 2001. [DOI] [PubMed] [Google Scholar]
- 209.Shor-Posner G, Azar AP, Insinga S, and Leibowitz SF. Deficits in the control of food intake after hypothalamic paraventricular nucleus lesions. Physiol Behav 35: 883–890, 1985. [DOI] [PubMed] [Google Scholar]
- 210.Siljee JE, Unmehopa UA, Kalsbeek A, Swaab DF, Fliers E, and Alkemade A. Melanocortin 4 receptor distribution in the human hypothalamus. European journal of endocrinology 168: 361–369, 2013. [DOI] [PubMed] [Google Scholar]
- 211.Sims JS and Lorden JF. Effect of paraventricular nucleus lesions on body weight, food intake and insulin levels. Behav Brain Res 22: 265–281, 1986. [DOI] [PubMed] [Google Scholar]
- 212.Singru PS, Wittmann G, Farkas E, Zseli G, Fekete C, and Lechan RM. Refeeding-activated glutamatergic neurons in the hypothalamic paraventricular nucleus (PVN) mediate effects of melanocortin signaling in the nucleus tractus solitarius (NTS). Endocrinology 153: 3804–3814, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Skibicka KP and Grill HJ. Hypothalamic and hindbrain melanocortin receptors contribute to the feeding, thermogenic, and cardiovascular action of melanocortins. Endocrinology 150: 5351–5361, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Smith AS, Korgan AC, and Young WS. Oxytocin delivered nasally or intraperitoneally reaches the brain and plasma of normal and oxytocin knockout mice. Pharmacological research: 104324, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Smith JE, Jansen AS, Gilbey MP, and Loewy AD. CNS cell groups projecting to sympathetic outflow of tail artery: neural circuits involved in heat loss in the rat. Brain research 786: 153–164, 1998. [DOI] [PubMed] [Google Scholar]
- 216.Snider B, Geiser A, Yu XP, Beebe EC, Willency JA, Qing K, Guo L, Lu J, Wang X, Yang Q, Efanov A, Adams AC, Coskun T, Emmerson PJ, Alsina-Fernandez J, and Ai M. Long-Acting and Selective Oxytocin Peptide Analogs Show Antidiabetic and Antiobesity Effects in Male Mice. Journal of the Endocrine Society 3: 1423–1444, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Sofroniew MV. Projections from vasopressin, oxytocin, and neurophysin neurons to neural targets in the rat and human. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society 28: 475–478, 1980. [DOI] [PubMed] [Google Scholar]
- 218.Sofroniew MV, Weindl A, Schrell U, and Wetzstein R. Immunohistochemistry of vasopressin, oxytocin and neurophysin in the hypothalamus and extrahypothalamic regions of the human and primate brain. Acta histochemica Supplementband 24: 79–95, 1981. [PubMed] [Google Scholar]
- 219.Spetter MS, Feld GB, Thienel M, Preissl H, Hege MA, and Hallschmid M. Oxytocin curbs calorie intake via food-specific increases in the activity of brain areas that process reward and establish cognitive control. Scientific reports 8: 2736, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Stanley S, Pinto S, Segal J, Perez CA, Viale A, DeFalco J, Cai X, Heisler LK, and Friedman JM. Identification of neuronal subpopulations that project from hypothalamus to both liver and adipose tissue polysynaptically. Proceedings of the National Academy of Sciences of the United States of America 107: 7024–7029, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Steinwall M, Akerlund M, Bossmar T, and Forsling ML. Osmotically-induced release of vasopressin and oxytocin in non-pregnant women--influence of estrogen and progesterone. Acta obstetricia et gynecologica Scandinavica 77: 983–987, 1998. [PubMed] [Google Scholar]
- 222.Stock S, Granstrom L, Backman L, Matthiesen AS, and Uvnas-Moberg K. Elevated plasma levels of oxytocin in obese subjects before and after gastric banding. International journal of obesity 13: 213–222, 1989. [PubMed] [Google Scholar]
- 223.Striepens N, Kendrick KM, Hanking V, Landgraf R, Wullner U, Maier W, and Hurlemann R. Elevated cerebrospinal fluid and blood concentrations of oxytocin following its intranasal administration in humans. Scientific reports 3: 3440, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Striepens N, Schroter F, Stoffel-Wagner B, Maier W, Hurlemann R, and Scheele D. Oxytocin enhances cognitive control of food craving in women. Human brain mapping 37: 4276–4285, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Stymiest JL, Mitchell BF, Wong S, and Vederas JC. Synthesis of oxytocin analogues with replacement of sulfur by carbon gives potent antagonists with increased stability. The Journal of organic chemistry 70: 7799–7809, 2005. [DOI] [PubMed] [Google Scholar]
- 226.Succu S, Sanna F, Cocco C, Melis T, Boi A, Ferri GL, Argiolas A, and Melis MR. Oxytocin induces penile erection when injected into the ventral tegmental area of male rats: role of nitric oxide and cyclic GMP. Eur J Neurosci 28: 813–821, 2008. [DOI] [PubMed] [Google Scholar]
- 227.Sukhov RR, Walker LC, Rance NE, Price DL, and Young WS, 3rd. Vasopressin and oxytocin gene expression in the human hypothalamus. The Journal of comparative neurology 337: 295–306, 1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Sutton AK, Pei H, Burnett KH, Myers MG Jr., Rhodes CJ, and Olson DP. Control of food intake and energy expenditure by Nos1 neurons of the paraventricular hypothalamus. The Journal of neuroscience : the official journal of the Society for Neuroscience 34: 15306–15318, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Suzuki M, Honda Y, Li MZ, Masuko S, and Murata Y. The localization of oxytocin receptors in the islets of Langerhans in the rat pancreas. Regulatory peptides 183: 42–45, 2013. [DOI] [PubMed] [Google Scholar]
- 230.Swaab DF, Nijveldt F, and Pool CW. Distribution of oxytocin and vasopressin in the rat supraoptic and paraventricular nucleus. The Journal of endocrinology 67: 461–462, 1975. [DOI] [PubMed] [Google Scholar]
- 231.Swaab DF, Pool CW, and Nijveldt F. Immunofluorescence of vasopressin and oxytocin in the rat hypothalamo-neurohypophypopseal system. Journal of neural transmission 36: 195–215, 1975. [DOI] [PubMed] [Google Scholar]
- 232.Sweeney G, Holbrook AM, Levine M, Yip M, Alfredsson K, Cappi S, Ohlin M, Schulz P, and Wassenaar W. Curr Ther Res Clin Exper 47: 528–539, 1990. [Google Scholar]
- 233.Szeto A, McCabe PM, Nation DA, Tabak BA, Rossetti MA, McCullough ME, Schneiderman N, and Mendez AJ. Evaluation of enzyme immunoassay and radioimmunoassay methods for the measurement of plasma oxytocin. Psychosomatic medicine 73: 393–400, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Szulc P, Amri EZ, Varennes A, Panaia-Ferrari P, Fontas E, Goudable J, Chapurlat R, and Breuil V. High serum oxytocin is associated with metabolic syndrome in older men - The MINOS study. Diabetes Res Clin Pract 122: 17–27, 2016. [DOI] [PubMed] [Google Scholar]
- 235.Takayanagi Y, Kasahara Y, Onaka T, Takahashi N, Kawada T, and Nishimori K. Oxytocin receptor-deficient mice developed late-onset obesity. Neuroreport 19: 951–955, 2008. [DOI] [PubMed] [Google Scholar]
- 236.Tanaka A, Furubayashi T, Arai M, Inoue D, Kimura S, Kiriyama A, Kusamori K, Katsumi H, Yutani R, Sakane T, and Yamamoto A. Delivery of Oxytocin to the Brain for the Treatment of Autism Spectrum Disorder by Nasal Application. Molecular pharmaceutics 15: 1105–1111, 2018. [DOI] [PubMed] [Google Scholar]
- 237.Tauchi M, Zhang R, D’Alessio DA, Stern JE, and Herman JP. Distribution of glucagon-like peptide-1 immunoreactivity in the hypothalamic paraventricular and supraoptic nuclei. Journal of chemical neuroanatomy 36: 144–149, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Thiele TE, Roitman MF, and Bernstein IL. c-Fos induction in rat brainstem in response to ethanol- and lithium chloride-induced conditioned taste aversions. Alcoholism, clinical and experimental research 20: 1023–1028, 1996. [DOI] [PubMed] [Google Scholar]
- 239.Thienel M, Fritsche A, Heinrichs M, Peter A, Ewers M, Lehnert H, Born J, and Hallschmid M. Oxytocin’s inhibitory effect on food intake is stronger in obese than normal-weight men. International journal of obesity 40: 1707–1714, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Tribollet E, Dubois-Dauphin M, Dreifuss JJ, Barberis C, and Jard S. Oxytocin receptors in the central nervous system. Distribution, development, and species differences. Annals of the New York Academy of Sciences 652: 29–38, 1992. [DOI] [PubMed] [Google Scholar]
- 241.Tung YC, Ma M, Piper S, Coll A, O’Rahilly S, and Yeo GS. Novel leptin-regulated genes revealed by transcriptional profiling of the hypothalamic paraventricular nucleus. J Neurosci 28: 12419–12426, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Ueta Y, Kannan H, Higuchi T, Negoro H, Yamaguchi K, and Yamashita H. Activation of gastric afferents increases noradrenaline release in the paraventricular nucleus and plasma oxytocin level. Journal of the autonomic nervous system 78: 69–76, 2000. [DOI] [PubMed] [Google Scholar]
- 243.Vaccari C, Lolait SJ, and Ostrowski NL. Comparative distribution of vasopressin V1b and oxytocin receptor messenger ribonucleic acids in brain. Endocrinology 139: 5015–5033, 1998. [DOI] [PubMed] [Google Scholar]
- 244.Vallera C, Choi LO, Cha CM, and Hong RW. Uterotonic Medications: Oxytocin, Methylergonovine, Carboprost, Misoprostol. Anesthesiol Clin 35: 207–219, 2017. [DOI] [PubMed] [Google Scholar]
- 245.van der Klaauw AA, Ziauddeen H, Keogh JM, Henning E, Dachi S, Fletcher PC, and Farooqi IS. Oxytocin administration suppresses hypothalamic activation in response to visual food cues. Scientific reports 7: 4266, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.van Leeuwen FW, van Heerikhuize J, van der Meulen G, and Wolters P. Light microscopic autoradiographic localization of [3H]oxytocin binding sites in the rat brain, pituitary and mammary gland. Brain research 359: 320–325, 1985. [DOI] [PubMed] [Google Scholar]
- 247.Vankrieken L, Godart A, and Thomas K. Oxytocin determination by radioimmunoassay. Gynecologic and obstetric investigation 16: 180–185, 1983. [DOI] [PubMed] [Google Scholar]
- 248.Vaughan CH, Shrestha YB, and Bartness TJ. Characterization of a novel melanocortin receptor-containing node in the SNS outflow circuitry to brown adipose tissue involved in thermogenesis. Brain research 1411: 17–27, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Velmurugan S, Russell JA, and Leng G. Systemic leptin increases the electrical activity of supraoptic nucleus oxytocin neurones in virgin and late pregnant rats. Journal of neuroendocrinology 25: 383–390, 2013. [DOI] [PubMed] [Google Scholar]
- 250.Verbalis JG. The brain oxytocin receptor(s)? Frontiers in neuroendocrinology 20: 146–156, 1999. [DOI] [PubMed] [Google Scholar]
- 251.Verbalis JG, Blackburn AN, Hoffman GE, and Stricker EM. Establishing behavioral and physiological functions of central oxytocin: insights from studies of oxytocin and ingestive behvaviors In: Oxytocin, edited by Ivell R and Russell J. New York: Plenum Press, 1995, p. 209–225. [PubMed] [Google Scholar]
- 252.Verbalis JG, McCann MJ, McHale CM, and Stricker EM. Oxytocin secretion in response to cholecystokinin and food: differentiation of nausea from satiety. Science 232: 1417–1419, 1986. [DOI] [PubMed] [Google Scholar]
- 253.Vrang N, Larsen PJ, Kristensen P, and Tang-Christensen M. Central administration of cocaine-amphetamine-regulated transcript activates hypothalamic neuroendocrine neurons in the rat. Endocrinology 141: 794–801, 2000. [DOI] [PubMed] [Google Scholar]
- 254.Weingarten MFJ, Scholz M, Wohland T, Horn K, Stumvoll M, Kovacs P, and Tonjes A. Circulating oxytocin is genetically determined and associated with obesity and impaired glucose tolerance. The Journal of clinical endocrinology and metabolism, 2019. [DOI] [PubMed] [Google Scholar]
- 255.Welch MG, Tamir H, Gross KJ, Chen J, Anwar M, and Gershon MD. Expression and developmental regulation of oxytocin (OT) and oxytocin receptors (OTR) in the enteric nervous system (ENS) and intestinal epithelium. The Journal of comparative neurology 512: 256–270, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Wirth MM, Olszewski PK, Levine AS, and Giraudo SQ. Effect of Agouti-related protein on development of conditioned taste aversion and oxytocin neuronal activation. Neuroreport 13: 1355–1358, 2002. [DOI] [PubMed] [Google Scholar]
- 257.Wrobel L, Schorscher-Petcu A, Dupre A, Yoshida M, Nishimori K, and Tribollet E. Distribution and identity of neurons expressing the oxytocin receptor in the mouse spinal cord. Neuroscience letters 495: 49–54, 2011. [DOI] [PubMed] [Google Scholar]
- 258.Wu CL, Hung CR, Chang FY, Pau KY, Wang JL, and Wang PS. Involvement of cholecystokinin receptor in the inhibition of gastric emptying by oxytocin in male rats. Pflugers Archiv : European journal of physiology 445: 187–193, 2002. [DOI] [PubMed] [Google Scholar]
- 259.Wu CL, Hung CR, Chang FY, Pau KY, and Wang PS. Pharmacological effects of oxytocin on gastric emptying and intestinal transit of a non-nutritive liquid meal in female rats. Naunyn-Schmiedeberg’s archives of pharmacology 367: 406–413, 2003. [DOI] [PubMed] [Google Scholar]
- 260.Wu L, Meng J, Shen Q, Zhang Y, Pan S, Chen Z, Zhu LQ, Lu Y, Huang Y, and Zhang G. Caffeine inhibits hypothalamic A1R to excite oxytocin neuron and ameliorate dietary obesity in mice. Nature communications 8: 15904, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Wu Z, Xu Y, Zhu Y, Sutton AK, Zhao R, Lowell BB, Olson DP, and Tong Q. An obligate role of oxytocin neurons in diet induced energy expenditure. PloS one 7: e45167, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Xi D, Long C, Lai M, Casella A, O’Lear L, Kublaoui B, and Roizen JD. Ablation of Oxytocin Neurons Causes a Deficit in Cold Stress Response. Journal of the Endocrine Society 1: 1041–1055, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Yamamoto T, Shimura T, Sako N, Azuma S, Bai WZ, and Wakisaka S. C-fos expression in the rat brain after intraperitoneal injection of lithium chloride. Neuroreport 3: 1049–1052, 1992. [DOI] [PubMed] [Google Scholar]
- 264.Yamamoto T, Shimura T, Sako N, Yasoshima Y, and Sakai N. Neural substrates for conditioned taste aversion in the rat. Behavioural brain research 65: 123–137, 1994. [DOI] [PubMed] [Google Scholar]
- 265.Yamashita M, Takayanagi Y, Yoshida M, Nishimori K, Kusama M, and Onaka T. Involvement of prolactin-releasing peptide in the activation of oxytocin neurones in response to food intake. Journal of neuroendocrinology 25: 455–465, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Yang Z, Han D, and Coote JH. Cardiac sympatho-excitatory action of PVN-spinal oxytocin neurones. Autonomic neuroscience : basic & clinical 147: 80–85, 2009. [DOI] [PubMed] [Google Scholar]
- 267.Yaseen A, Shrivastava K, Zuri Z, Hatoum OA, and Maroun M. Prefrontal Oxytocin is Involved in Impairments in Prefrontal Plasticity and Social Memory Following Acute Exposure to High Fat Diet in Juvenile Animals. Cerebral cortex, 2018. [DOI] [PubMed] [Google Scholar]
- 268.Yi KJ, So KH, Hata Y, Suzuki Y, Kato D, Watanabe K, Aso H, Kasahara Y, Nishimori K, Chen C, Katoh K, and Roh SG. The regulation of oxytocin receptor gene expression during adipogenesis. Journal of neuroendocrinology 27: 335–342, 2015. [DOI] [PubMed] [Google Scholar]
- 269.Yoshida M, Takayanagi Y, Inoue K, Kimura T, Young LJ, Onaka T, and Nishimori K. Evidence that oxytocin exerts anxiolytic effects via oxytocin receptor expressed in serotonergic neurons in mice. The Journal of neuroscience : the official journal of the Society for Neuroscience 29: 2259–2271, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Yoshimura R, Kiyama H, Kimura T, Araki T, Maeno H, Tanizawa O, and Tohyama M. Localization of oxytocin receptor messenger ribonucleic acid in the rat brain. Endocrinology 133: 1239–1246, 1993. [DOI] [PubMed] [Google Scholar]
- 271.Yosten GL and Samson WK. Neural circuitry underlying the central hypertensive action of nesfatin-1: melanocortins, corticotropin-releasing hormone, and oxytocin. American journal of physiology Regulatory, integrative and comparative physiology 306: R722–727, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Yuan G, Qian W, Pan R, Jia J, Jiang D, Yang Q, Wang S, Liu Y, Yu S, Hu H, Sun W, Ye J, Mao C, Zhuang R, and Zhou L. Reduced circulating oxytocin and High-Molecular-Weight adiponectin are risk factors for metabolic syndrome. Endocr J 63: 655–662, 2016. [DOI] [PubMed] [Google Scholar]
- 273.Zagoory-Sharon O, Schroeder M, Levine A, Moran TH, and Weller A. Adaptation to lactation in OLETF rats lacking CCK-1 receptors: body weight, fat tissues, leptin and oxytocin. International journal of obesity 32: 1211–1221, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Zaman RU, Mulla NS, Braz Gomes K, D’Souza C, Murnane KS, and D’Souza MJ. Nanoparticle formulations that allow for sustained delivery and brain targeting of the neuropeptide oxytocin. International journal of pharmaceutics 548: 698–706, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Zhang G, Bai H, Zhang H, Dean C, Wu Q, Li J, Guariglia S, Meng Q, and Cai D. Neuropeptide exocytosis involving synaptotagmin-4 and oxytocin in hypothalamic programming of body weight and energy balance. Neuron 69: 523–535, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Zhang G and Cai D. Circadian intervention of obesity development via resting-stage feeding manipulation or oxytocin treatment. American journal of physiology Endocrinology and metabolism 301: E1004–1012, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Zhang H, Wu C, Chen Q, Chen X, Xu Z, Wu J, and Cai D. Treatment of obesity and diabetes using oxytocin or analogs in patients and mouse models. PloS one 8: e61477, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Zhang Y, Collazo R, Gao Y, Li G, and Scarpace PJ. Intermittent MTII application evokes repeated anorexia and robust fat and weight loss. Peptides 31: 639–643, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Zueco JA, Esquifino AI, Chowen JA, Alvarez E, Castrillon PO, and Blazquez E. Coexpression of glucagon-like peptide-1 (GLP-1) receptor, vasopressin, and oxytocin mRNAs in neurons of the rat hypothalamic supraoptic and paraventricular nuclei: effect of GLP-1(7–36)amide on vasopressin and oxytocin release. Journal of neurochemistry 72: 10–16, 1999. [DOI] [PubMed] [Google Scholar]