Abstract
We have previously shown that neuropeptide Y (NPY) reduces social fear in an animal model that closely mimics the key behavioral symptoms of social anxiety disorder (SAD). Since NPY cannot yet be routinely administered to patients, we investigated the effects of sitagliptin, a dipeptidyl peptidase-4 (DPP4) inhibitor approved for the treatment of type 2 diabetes mellitus, on social fear and comorbid depression in mice. In addition to its well-known effects on glucose metabolism, sitagliptin also prevents the degradation of NPY, thereby increasing its concentration in the blood and the brain. We show that sitagliptin administration via drinking water (50 and 100 mg/kg/day, for 4 weeks) not only reduced social fear but also prevented the onset of comorbid depressive-like behavior in outbred CD1 mice. A similar phenotype was observed in homozygous DPP4-deficient mice, emphasizing the role of DPP4 in regulating these behaviors. However, in NPY-deficient mice, sitagliptin showed reduced efficacy, suggesting that NPY plays an important role in mediating the effects of sitagliptin on social fear and comorbid depression. These findings have important clinical implications, indicating that early intervention with sitagliptin could be an effective strategy for treating SAD, alleviating both core symptoms and reducing the risk of developing comorbid mood disorders that often complicate treatment outcomes.
Similar content being viewed by others
Introduction
Social anxiety disorder (SAD) is the second most common anxiety disorder, with a lifetime prevalence of around 12%, and is characterized by persistent fear and avoidance of social situations [1]. SAD is highly comorbid with major depressive disorder (affecting 35–70% of SAD patients), specific phobia (14–61%), panic disorder (5–27%), agoraphobia (8–45%), generalized anxiety disorder (0.6–27%), alcohol use disorders (up to 50%), obsessive compulsive disorder (2–19%) and posttraumatic stress disorder (3–16%) [1]. Except for specific phobia, SAD has an earlier onset, suggesting that SAD is a risk factor for developing additional psychiatric disorders [1]. Early diagnosis and intervention could help reduce this risk.
Animal models are essential for understanding the neurobiology of SAD and its comorbidities. They should ideally replicate both specific social anxiety (i.e., without any comorbidities) and social anxiety with comorbid conditions to provide valuable insights for developing targeted treatments for SAD patients with and without comorbidities. One such model is social fear conditioning (SFC), which mimics the key behavioral symptoms of SAD, like social avoidance and reduced social investigation [2, 3]. The SFC model has face, predictive and construct validity for human SAD [2, 4] and induces specific social fear without symptoms of generalized anxiety or depressive-like behavior for up to 2–3 weeks post-conditioning [2, 5]. A depressive-like phenotype can emerge later, around 5–6 weeks post-conditioning [5].
In previous studies, we demonstrated that central administration of neuropeptide Y (NPY), a neuropeptide well known for its fear-reducing properties in animal models of conditioned fear [6], effectively reduced SFC-induced social fear in male mice [7,8,9]. Central NPY also alleviated an antidepressant-resistant type of social fear in both male and female mice [10], supporting its potential in treating SAD. Elevated NPY levels have also been associated with reduced anxiety- and depressive-like behaviors, while lower NPY levels often correlate with heightened stress sensitivity and more pronounced depressive-like symptoms [11, 12]. This ability of NPY to modulate the stress response and to reduce both fear-related behaviors and anxiety-/depressive-like symptoms suggests that NPY may be a promising target for developing new therapies for SAD with comorbid depression. However, NPY has not yet been developed or approved for routine clinical use, and its administration to patients is currently restricted to controlled research settings.
One effective way to increase NPY concentration is by inhibiting its breakdown. Dipeptidyl peptidase-4 (DPP4) is an enzyme that cleaves the amino-terminal dipeptide from several peptides, including the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP), as well as other peptides such as NPY [13,14,15,16]. DPP4 inhibitors, also known as gliptins, are used in the treatment of type 2 diabetes mellitus to improve glucose homeostasis. They inhibit the degradation of GLP-1 and GIP, leading to an increased insulin secretion and reduced glucagon secretion [14, 15]. In addition to their effects on glucose metabolism, DPP4 inhibitors such as sitagliptin, vildagliptin, linagliptin and saxagliptin also inhibit the degradation of NPY, thereby increasing its blood concentration in healthy individuals and patients with type 2 diabetes mellitus [17,18,19,20,21]. In rodents, sitagliptin has been shown to increase NPY levels in brain regions such as the hippocampus, amygdala and hypothalamus [22]. Preclinical studies suggest that DPP4 inhibition may offer potential benefits for treating conditioned fear, anxiety and mood disorders. For example, sitagliptin exerts antidepressant-like effects in naïve mice [23]. DPP4 inhibitors also improve social interaction deficits [24] and reduce depressive-like behaviors in various animal models of depression, including chronic restraint stress [24], chronic unpredictable mild stress [25], high fat diet [24], diabetes [26] and morphine withdrawal [27]. Additional insights come from studies on DPP4-deficient rodents, which show enhanced extinction of cued fear [28], reduced anxiety-like behavior [29, but see 30] and reduced depressive-like behavior [31]. These DPP4-deficient animals also exhibit increased NPY levels in the cerebrospinal fluid [28], supporting the idea that the effects of DPP4 inhibition may be mediated, at least in part, by increased NPY levels.
In this study, we investigated whether early intervention with sitagliptin reduces SFC-induced social fear and prevents the development of comorbid depressive-like behavior. Additionally, we examined whether DPP4-deficient mice show reduced social fear compared with wild-type littermates, and whether NPY-deficient mice exhibit a diminished response to sitagliptin.
Materials and methods
Animals
Experiments were performed in male CD1 mice (Charles River, Sulzfeld, Germany), male and female homozygous DPP4-deficient (DPP4 −/−) [32] and NPY-deficient (NPY −/−) mice [33,34,35] (Supplementary Materials and Methods), as well as in wild-type controls (DPP4 + /+ and NPY + /+ mice), at an age of 8 − 9 weeks (see Table S1 for a detailed depiction of the experimental groups). Mice were individually housed for one week before experiments started and remained single-housed throughout the experiments. Age-, sex- and strain-matched wild-type mice were used as social stimuli for the SFC experiments. Mice were kept under standard laboratory conditions (12:12 light/dark cycle, lights on at 07:00 h, 22 °C, 60% humidity, food and water ad libitum). Experiments were performed during the light phase between 09:00 and 14:00 in accordance with the EU Directive 2010/63 for the protection of animals used for scientific purposes and were approved by the local government commission for animal protection (Regierung von Unterfranken).
Gliptin treatment
Sitagliptin (sitagliptin phosphate; PHR1857, Sigma-Aldrich, Darmstadt, Germany) was administered for 4 weeks via drinking water at a daily dose of 50 mg/kg or 100 mg/kg [36, 37]. The treatment was started one day after SFC to avoid potential treatment-induced alterations in social fear memory consolidation and was maintained throughout the experiment. The mice’s body weight and drinking volume were assessed every two to three days, and the sitagliptin solution was freshly prepared accordingly.
Social fear conditioning (SFC) paradigm
To induce social fear, mice were conditioned during SFC on day 1, and social investigation was assessed during social fear extinction on day 30 as a read-out of social fear.
SFC (day 1)
SFC was performed with a computerized fear conditioning system (TSE System GmbH, Bad Homburg, Germany), as previously described [3, 4, 7,8,9,10, 38,39,40]. Mice were placed in the conditioning chamber (45 × 22 × 40 cm) and, after a 30-sec habituation, an empty wire mesh cage (7 × 7 × 6 cm) was placed as a non-social stimulus near one of the short walls. After 3 min, the non-social stimulus was replaced by an identical cage containing an unfamiliar age-, sex- and strain-matched mouse. Unconditioned control mice (SFC –) were allowed to investigate this social stimulus for 3 min, whereas socially fear conditioned mice (SFC + ) received a 1-sec mild electric foot-shock (0.7 mA) each time they investigated, i.e., made direct contact with the social stimulus. Mice received between one and four foot-shocks with a variable inter-shock interval, depending on when direct social contact was made. The number of foot-shocks was assessed as a measure of distress and of social fear learning. Mice were returned to their home cage when no further social contact was made for 2 min (average duration of SFC approximately 10 min). The time spent investigating the non-social stimulus was analyzed as a preconditioning measure of non-social anxiety.
Social fear extinction (day 30)
After 4 weeks of sitagliptin treatment, mice were exposed in their home cage to three non-social stimuli, to assess non-social investigation as a parameter of non-social fear. Mice were then exposed to six unfamiliar social stimuli to assess social investigation as a parameter of social fear. Each stimulus was presented for 3 min, with a 3-min inter-exposure interval. The test was recorded and analyzed using JWatcher (V 1.0, Macquarie University, Sydney, Australia, and UCLA, Los Angeles, CA, USA). Non-social investigation was defined as direct sniffing of the empty cage, whereas social investigation was defined as direct sniffing of the cage and/or the social stimulus inside the cage.
Elevated plus-maze (EPM) test
To investigate whether SFC leads to the development of a comorbid anxious phenotype, mice were tested in the EPM on day 34 as previously described [5, 41, 42]. The test was recorded and analyzed using JWatcher. The time spent on the open arms (150 lx) indicated non-social anxiety-like behavior. The number of entries into the closed arms (30 lx) during the 5-min testing period indicated locomotor activity.
Forced swim test (FST)
To investigate whether SFC leads to the development of a comorbid depressive-like phenotype, mice were tested in the FST on day 39 as previously described [5, 41, 42]. Mice were individually placed into a Plexiglas cylinder filled with 25 °C water for 6 min. The test was recorded and analyzed using JWatcher. An increased immobility time during the last 4 min of the test indicated a depressive-like phenotype.
Quantification of DPP4 activity, DPP4 protein levels and baseline corticosterone (CORT) levels in the serum
To verify whether the selected doses of sitagliptin effectively inhibited DPP4 activity, CD1 mice were rapidly killed one day after behavioral testing, on day 40. Trunk blood was collected via cardiac puncture and left to coagulate. After centrifugation (4 °C, 4000 rpm, 10 min), the serum was extracted and stored at –80 °C.
DPP4 activity was quantified in 4 µl serum duplicates using the DPP4 Activity Assay Kit (MAK088, Sigma-Aldrich, Saint Louis, MO, USA), based on the cleavage of the non-fluorescent substrate H-Gly-Pro-AMC to release the fluorescent product7-Amino-4-Methyl Coumarin (AMC). The kinetics of fluorescence increase at 37 °C were assessed on a CLARIOstar Plus multi-mode plate reader (BMG LABTECh GmbH, Ortenberg, Germany) and corrected for the AMC release in a reaction with sitagliptin as DPP4 inhibitor. All samples were assessed within one assay with an intra-assay coefficient of variation (CV) of 2%.
DPP4 protein levels were quantified in 1 µL serum duplicates using the Mouse DPPIV ELISA Kit (RAB0148, Sigma-Aldrich, Saint Louis, MO, USA), based on a standard curve ranging from 0.3 to 100 ng/mL, within one assay with an intra-assay CV of 3%.
To assess whether SFC and/or sitagliptin treatment affected baseline CORT levels, CORT was quantified in 4 µL serum duplicates using a competitive CORT ELISA Kit (EIA-4164, DRG Instruments GmbH, Marburg, Germany), based on a standard curve ranging from 5 to 240 nmol/L. The intra-assay CV was 5% and the inter-assay CV was 12%.
Statistical analysis
For statistical analysis, IBM SPSS (Version 28.0, SPSS Inc., Chicago, IL, USA) was used. Data were analyzed by Student’s t-test, one-way, two-way or three-way ANOVA for repeated measures, followed by Bonferroni’s post hoc analysis whenever appropriate. Statistical significance was set at p < 0.05.
Results
Sitagliptin did not influence body weight in either CD1 mice (Fig. S1; Table S2) or NPY-deficient mice (Fig. S2; Table S2). Sitagliptin consumption was consistent within each group, and neither sitagliptin nor its taste influenced fluid intake (Fig. S1; Fig. S2; Table S2). Male and female mice were pooled together, as no sex differences in behavior were observed (Table S3). Sex differences in body weight and fluid intake in NPY-deficient mice are provided in the supplementary information (Fig. S2; Table S2).
Sitagliptin reduces social fear and prevents the development of comorbid depressive-like behavior in CD1 mice
During SFC on day 1, all groups of mice showed similar investigation of the non-social stimulus (empty cage), reflecting similar preconditioning non-social anxiety (Fig. 1a; F(3,56) = 0.058; p = 0.981). All SFC + mice received a similar number of foot-shocks, reflecting similar levels of distress during conditioning and similar social fear learning between the groups (Fig. 1b; F(2,43) = 0.092; p = 0.912). During social fear extinction on day 30, all mice showed similar non-social investigation (i.e., investigation of the three empty cages), indicating that SFC did not induce an unspecific non-social fear and that sitagliptin did not alter non-social investigation (Fig. 1c). Water-drinking SFC + mice showed reduced social investigation (i.e., investigation of the six social stimuli) compared with all other groups, reflecting social fear (Fig. 1c; group effect F(3,56) = 17.494; p < 0.001; stimulus x group effect F(24,448) = 6.603; p < 0.001). Both doses of sitagliptin, however, increased social investigation to levels observed in SFC– mice starting from the first (100 mg/kg/day) and second (50 mg/kg/day) social stimulus, reflecting significantly reduced social fear in SFC + mice after sitagliptin treatment.
a Preconditioning investigation of the non-social stimulus (empty cage) shown by unconditioned (SFC − ) and conditioned (SFC + ) mice during social fear conditioning (SFC) on day 1. b Number of foot-shocks received during SFC. c Investigation of the non-social (ns1 − ns3) and social (cages with mice; s1 − s6) stimuli during social fear extinction on day 30; # p < 0.05 versus SFC − /W and SFC + /100 Sita; * p < 0.05 versus all groups. d Time spent on the open arms of the elevated plus-maze (EPM) and e number of closed-arm entries on day 34. f Immobility time in the forced swim test (FST) on day 39; * p < 0.05. Mice were administered sitagliptin (Sita; 50 or 100 mg/kg/day) via drinking water (W). Data represent means ± SEM and numbers on the bars and in parentheses indicate group sizes.
On day 34, all mice showed similar non-social anxiety-like behavior (Fig. 1d; F(3,56) = 0.395; p = 0.757) and similar locomotor activity (Fig. 1e; F(3,56) = 0.672; p = 0.573) on the EPM, as reflected by similar time spent on the open arms and similar number of entries into the closed arms between the groups, respectively. However, on day 39, water-drinking SFC + mice showed increased immobility in the FST compared with water-drinking SFC − mice (Fig. 1f; F(3,56) = 5.550; p = 0.002), indicating that SFC induced a depressive-like phenotype. This depressive-like phenotype was not observed in sitagliptin-treated mice, indicating that sitagliptin prevented the development of comorbid depressive-like behavior in SFC + mice.
Sitagliptin treatment effectively reduced DPP4 activity (Fig. 2a; F(3,26) = 65.249; p < 0.001) and increased DPP4 protein levels (Fig. 2b; F(3,26) = 44.675; p < 0.001) in the serum, regardless of the dose applied, supporting previous findings in both blood and brain [43,44,45]. This indicates that the selected sitagliptin doses were appropriate for the intended therapeutic effect. Neither SFC nor sitagliptin treatment altered the baseline serum CORT levels (Fig. 2c; F(3,54) = 0.299; p = 0.826), suggesting that the observed effects on behavior were not mediated by changes in baseline stress levels. In a pilot analysis, sitagliptin treatment (50 mg/kg/day) increased serum NPY protein levels in socially fear conditioned male CD1 mice (T(11) = −2.349; p = 0.039; Fig. S3), confirming previous findings [17, 18].
Mice were administered sitagliptin (Sita; 50 or 100 mg/kg/day) via drinking water (W). a Dipeptidyl dipeptidase 4 (DPP4) activity, b DPP4 protein levels and c baseline CORT levels in serum after 39 days of sitagliptin treatment. Data represent means + SEM and numbers on the bars indicate group sizes. * p < 0.05 versus SFC − /W and SFC + /W.
DPP4 deficiency reduces social fear and prevents the development of comorbid depressive-like behavior
During SFC on day 1, all mice showed similar preconditioning non-social anxiety (Fig. 3a; F(1,28) = 0.017; p = 0.897). All SFC + mice received a similar number of foot-shocks (Fig. 3b; T(15) = − 1.265; p = 0.225). During social fear extinction on day 30, all mice showed similar non-social investigation (Fig. 3c). Both SFC + /DPP4 −/− and SFC + /DPP4 + /+ mice showed reduced social investigation compared with respective SFC − mice, reflecting social fear (Fig. 3c; conditioning x genotype effect F(1,28) = 6.728; p = 0.015; stimulus x genotype effect F(8,224) = 4.090; p < 0.001). SFC + /DPP4 −/− mice, however, showed higher social investigation compared with SFC + /DPP4 + /+ mice starting from the second social stimulus, reflecting reduced social fear.
a Preconditioning investigation of the non-social stimulus (empty cage) shown by unconditioned (SFC − ) and conditioned (SFC + ) homozygous dipeptidyl dipeptidase 4-deficient mice (DPP4 − /−) and wild-type controls (DPP4 + /+) during social fear conditioning (SFC) on day 1. b Number of foot-shocks received during SFC. c Investigation of the non-social (ns1 − ns3) and social (cages with mice; s1 − s6) stimuli during social fear extinction on day 30; # p < 0.05 SFC + versus SFC − , independent of genotype; * p < 0.05 versus SFC − /DPP4 − /− and SFC + /DPP4 + /+. d Time spent on the open arms of the elevated plus-maze (EPM) and e number of closed-arm entries on day 34. f Immobility time in the forced swim test (FST) on day 39; * p < 0.05. Data represent means ± SEM and numbers on the bars and in parentheses indicate group sizes. Blue and red dots represent individual male and female mice, respectively.
On day 34, all mice showed similar non-social anxiety-like behavior (Fig. 3d; F(1,28) = 0.008; p = 0.931) and similar locomotor activity (Fig. 3e; F(1,28) = 0.078; p = 0.783) on the EPM. However, on day 39, SFC + /DPP4 + /+ but not SFC + /DPP4 −/− mice showed increased immobility in the FST compared with respective SFC − mice (Fig. 3f; conditioning effect F(1,28) = 4.450; p = 0.044; conditioning x genotype effect F(1,28) = 2.177; p = 0.151), indicating that DPP4 deficiency prevented the development of comorbid depressive-like behavior in SFC + mice.
Sitagliptin reduces social fear and prevents the development of comorbid depressive-like behavior in NPY + /+ mice; reduced efficacy in NPY −/− mice
During SFC on day 1, all mice showed similar preconditioning non-social anxiety (Fig. 4a; F(5,62) = 0.326; p = 0.895). All SFC + mice received a similar number of foot-shocks (Fig. 4b; F(3,40) = 0.303; p = 0.823). During social fear extinction on day 30, all mice showed similar non-social investigation (Fig. 4c). Both water-drinking SFC + /NPY −/− and SFC + /NPY + /+ mice showed reduced social investigation compared with respective water-drinking SFC − mice, reflecting social fear (Fig. 4c; group effect F(5,62) = 25.368; p < 0.001; stimulus x group effect F(40,496) = 8.874; p < 0.001). In SFC + /NPY + /+ mice, sitagliptin (100 mg/kg/day) increased social investigation to levels observed in SFC – /NPY + /+ mice starting from the second social stimulus, reflecting significantly reduced social fear. In SFC + /NPY −/− mice, however, sitagliptin showed a reduced efficacy as it increased social investigation to levels observed in SFC – /NPY −/− mice starting from the fourth social stimulus.
a Preconditioning investigation of the non-social stimulus (empty cage) shown by unconditioned (SFC − ) and conditioned (SFC + ) homozygous neuropeptide Y-deficient mice (NPY − /−) and wild-type controls (NPY + /+) during social fear conditioning (SFC) on day 1. b Number of foot-shocks received during SFC. c Investigation of the non-social (ns1 − ns3) and social (cages with mice; s1 − s6) stimuli during social fear extinction on day 30; # p < 0.05 versus SFC − /W/NPY + /+ and SFC + /W/NPY + /+; * p < 0.05 versus SFC − /W/NPY − /−. d Time spent on the open arms of the elevated plus-maze (EPM) and e number of closed-arm entries on day 34. f Immobility time in the forced swim test (FST) on day 39; * p < 0.05. Mice were administered sitagliptin (Sita; 100 mg/kg/day) via drinking water (W). Data represent means ± SEM and numbers on the bars and in parentheses indicate group sizes. Blue and red dots represent individual male and female mice, respectively.
On day 34, NPY + /+ mice showed similar non-social anxiety-like behavior in the EPM. SFC – /NPY −/− mice, however, were more anxious than SFC – /NPY + /+ mice, reflecting elevated baseline anxiety levels in NPY −/− mice (Fig. 4d; F(5,62) = 12.731; p < 0.001). Interestingly, SFC further increased anxiety levels in NPY −/− mice, although such an effect was not observed in NPY + /+ mice nor in CD1 or DPP4 + /+ mice. This indicates that SFC induces a comorbid anxious phenotype specifically in individuals with increased baseline anxiety levels. Sitagliptin treatment attenuated, but did not reverse the SFC-induced anxious phenotype in SFC + /NPY −/− mice (Fig. 4d; p = 0.128). The locomotor activity on the EPM was similar between the groups (Fig. 4e; F(5,62) = 0.285; p = 0.919).
On day 39, water-drinking SFC + /NPY + /+ mice showed increased immobility in the FST compared with water-drinking SFC −/NPY + /+ mice (Fig. 4f; F(5,62) = 9.375; p < 0.001). This depressive-like phenotype was not observed in sitagliptin-treated NPY + /+ mice, indicating that sitagliptin prevented the development of comorbid depressive-like behavior in wild-type SFC + mice. SFC – /NPY −/− mice showed increased immobility compared with SFC – /NPY + /+ mice, reflecting elevated baseline levels of depressive-like behavior in NPY −/− mice (Fig. 4f; p = 0.006), and SFC did not further accentuate this depressive-like phenotype in NPY −/− mice. Sitagliptin treatment, however, tended to attenuate the depressive-like behavior in SFC + /NPY −/− mice (Fig. 4f; p = 0.058).
Discussion
We demonstrate that the DPP4 inhibitor sitagliptin significantly reduces social fear in an animal model that closely mimics the key behavioral symptoms of SAD, while also preventing the onset of comorbid depressive-like behavior in CD1 mice. Similarly, homozygous DPP4-deficient mice displayed reduced social fear and did not develop comorbid depressive-like behavior, mirroring the effects of sitagliptin treatment. A reduced efficacy of sitagliptin was observed in homozygous NPY-deficient mice, suggesting that NPY plays an important role in mediating sitagliptin’s effects on social fear and depressive-like behavior. These findings provide new insights into the therapeutic potential of early intervention with sitagliptin for SAD, not only in alleviating symptoms but also in reducing the risk of comorbid mood disorders.
Our findings contribute to the growing body of evidence supporting the therapeutic potential of DPP4 inhibition in alleviating symptoms of anxiety and mood disorders. Previous studies have shown that DPP4 inhibitors improve social interaction deficits [24] and reduce depressive-like behaviors in various animal models of depression, including chronic restraint stress [24], chronic unpredictable mild stress [25], high fat diet [24], diabetes [26] and morphine withdrawal [27]. Our study extends these observations by showing that early treatment with sitagliptin has the potential not only to reduce social fear but also to prevent the onset of comorbid anxiety- and depressive-like behavior. This is particularly relevant for SAD, as comorbid anxiety and mood disorders are common and often complicate treatment outcomes [1]. While a larger sample size would be needed for more conclusive results, our findings suggest that both males and females could benefit from sitagliptin treatment.
The observation that DPP4-deficient mice display reduced social fear and do not develop comorbid depressive-like behavior further emphasizes the importance of DPP4 in regulating these behaviors. Notably, the fear learning processes and the initial expression of social fear in response to the first social stimulus were not altered in DPP4-deficient mice, ruling out potential impairments in fear memory. Instead, DPP4-deficient mice displayed a facilitated extinction of social fear, supporting previous reports of enhanced cued fear extinction in DPP4-deficient rats [28]. As fear extinction is a form of associative learning, this finding might suggest that DPP4 deficiency positively influences cognitive function. Supporting this hypothesis, DPP4 inhibition reduced cognitive impairments associated with neurodegenerative [46,47,48] and metabolic disorders [36, 49]. Additionally, DPP4-deficient rats show unaltered pain perception [28, 50], suggesting that the facilitated extinction of cued and social fear is not due to changes in pain sensitivity.
The reduced efficacy of sitagliptin observed in homozygous NPY-deficient mice indicates that NPY plays an important role in mediating sitagliptin’s effects on social fear and comorbid depressive-like behavior. Sitagliptin inhibits the degradation of NPY, resulting in elevated NPY levels in the blood [17, 18] (Fig. S3) and in brain regions such as the hippocampus, amygdala and hypothalamus [22]. Elevated NPY levels have been shown to reduce social fear in our experimental paradigm [7,8,9,10], impair the retention and retrieval of cued fear [51, 52] and facilitate the extinction of both cued and contextual fear [52,53,54]. Additionally, both sitagliptin and NPY have been linked to increased levels of brain-derived neurotrophic factor (BDNF) [46, 48, 55], a key molecule for neuronal survival, growth, differentiation and synaptic plasticity. BDNF has been shown to reduce conditioned fear and prevent the onset of stress-induced depressive-like behaviors [56,57,58]. Moreover, sitagliptin, NPY and BDNF are all implicated in promoting hippocampal neurogenesis, which is essential for memory formation, fear extinction and the mitigation of depressive-like behavior [59,60,61]. Although sitagliptin elevated blood NPY levels in socially fear-conditioned male CD1 mice (Fig. S3) and NPY is known to efficiently cross the blood-brain barrier [62], it remains unclear whether sitagliptin also increases brain NPY levels in these mice. The absence of this data represents a limitation of the study and warrants further investigation. The reduced efficacy, rather than a complete lack of effect, of sitagliptin in homozygous NPY-deficient mice also suggests that while NPY plays an important role, it is unlikely the sole mediator of sitagliptin’s effects. GLP-1 and GIP, other molecules whose degradation is inhibited by sitagliptin, may also contribute to the observed effects. GLP-1 analogs have been shown to alleviate social interaction deficits and reduce anxiety- and depressive-like behaviors in animal models of depression [63, 64] and diabetes [65, 66]. Notably, GLP-1 and GIP also promote synaptic plasticity, stimulate neurogenesis and promote neuroprotection [63, 64, 67, 68]. Importantly, DPP4-deficient mice and rats exhibit elevated levels of GLP-1, NPY and BDNF [28, 69], which are likely key factors contributing to their phenotype observed in this study. Thus, sitagliptin might reduce social fear and prevent the onset of comorbid depressive-like behavior through multiple interconnected pathways, which will be investigated in further studies.
Interestingly, while no effects of SFC on non-social anxiety-like behavior were observed in CD1 [5], C57Bl/6 [5] or DPP4-deficient mice, SFC did induce an anxious phenotype in NPY-deficient mice. Since NPY-deficient mice exhibited heightened anxiety levels compared with wild-type controls, as reported in previous studies [34, 70], it appears that SFC exacerbates anxiety in rodents with pre-existing heightened anxiety traits. Interestingly, sitagliptin has been shown to exert anxiolytic-like effects in rodents with elevated anxiety levels [48], but not in those without such heightened anxiety [23]. Overall, these findings suggest that both the comorbid anxiety induced by SFC and the efficacy of sitagliptin in modulating anxiety-like behavior may be influenced by baseline anxiety levels.
In conclusion, our findings demonstrate that both sitagliptin treatment and DPP4 deficiency reduce social fear and prevent the onset of comorbid depressive-like behavior. The reduced efficacy of sitagliptin in NPY-deficient mice highlights the role of NPY in mediating these effects. Our findings emphasize the complex interplay between DPP4 and NPY in modulating social fear and suggest that early intervention with sitagliptin could offer significant clinical promise for treating SAD. By alleviating social fear and preventing the onset of comorbid depressive-like behavior, sitagliptin may provide a dual therapeutic benefit for SAD patients. Given its approval for type 2 diabetes mellitus, sitagliptin’s potential repurposing for SAD offers an opportunity for faster clinical application, providing a promising strategy to address both the core symptoms of SAD and the associated mood disorders that often complicate treatment outcomes.
Data availability
The data generated during the current study are available from the corresponding author on request.
References
Koyuncu A, İnce E, Ertekin E, Tükel R. Comorbidity in social anxiety disorder: diagnostic and therapeutic challenges. Drugs Context 2019;8:212573.
Toth I, Neumann ID, Slattery DA Social fear conditioning as an animal model of social anxiety disorder. Curr Protoc Neurosci. 2013;Chapter 9:Unit9.42.
Toth I, Neumann ID, Slattery DA. Social fear conditioning: a novel and specific animal model to study social anxiety disorder. Neuropsychopharmacology 2012;37:1433–43.
Kornhuber J, Huber SE, Zoicas I. Effects of conditioned social fear on ethanol drinking and vice-versa in male mice. Psychopharmacol (Berl) 2019;236:2059–67.
Zoicas I, Mühle C, Schumacher F, Kleuser B, Kornhuber J. Development of comorbid depression after social fear conditioning in mice and its effects on brain sphingolipid metabolism. Cells 2023;12:1355.
Tasan RO, Verma D, Wood J, Lach G, Hörmer B, de Lima TC, et al. The role of neuropeptide Y in fear conditioning and extinction. Neuropeptides 2016;55:111–26.
Kornhuber J, Zoicas I. Neuropeptide Y reduces expression of social fear via simultaneous activation of Y1 and Y2 receptors. J Psychopharmacol 2019;33:1533–39.
Kornhuber J, Zoicas I. Brain region-dependent effects of neuropeptide Y on conditioned social fear and anxiety-like behavior in male mice. Int J Mol Sci. 2021;22:3695.
Kornhuber J, Zoicas I. Neuropeptide Y reduces social fear in male mice: involvement of Y1 and Y2 receptors in the dorsolateral septum and central amygdala. Int J Mol Sci. 2021;22:10142.
Kornhuber J, Zoicas I. Neuropeptide Y as alternative pharmacotherapy for antidepressant-resistant social fear. Int J Mol Sci. 2020;21:8220.
Kormos V, Gaszner B. Role of neuropeptides in anxiety, stress, and depression: from animals to humans. Neuropeptides 2013;47:401–19.
Rana T, Behl T, Sehgal A, Singh S, Sharma N, Abdeen A, et al. Exploring the role of neuropeptides in depression and anxiety. Prog Neuropsychopharmacol Biol Psychiatry 2022;114:110478.
Frerker N, Wagner L, Wolf R, Heiser U, Hoffmann T, Rahfeld JU, et al. Neuropeptide Y (NPY) cleaving enzymes: structural and functional homologues of dipeptidyl peptidase 4. Peptides 2007;28:257–68.
Golightly LK, Drayna CC, McDermott MT. Comparative clinical pharmacokinetics of dipeptidyl peptidase-4 inhibitors. Clin Pharmacokinet 2012;51(8):501-14 Erratum : Clin Pharmacokinet 2012;51:831.
Filippatos TD, Athyros VG, Elisaf MS. The pharmacokinetic considerations and adverse effects of DPP-4 inhibitors [corrected]. Expert Opin Drug Metab Toxicol 2014;10:787–812.
Wagner L, Wolf R, Zeitschel U, Rossner S, Petersén Å, Leavitt BR, et al. Proteolytic degradation of neuropeptide Y (NPY) from head to toe: Identification of novel NPY-cleaving peptidases and potential drug interactions in CNS and Periphery. J Neurochem 2015;135:1019–37.
Hubers SA, Wilson JR, Yu C, Nian H, Grouzmann E, Eugster P, et al. DPP (dipeptidyl peptidase)-4 inhibition potentiates the vasoconstrictor response to NPY (neuropeptide Y) in humans during renin-angiotensin-aldosterone system inhibition. Hypertension 2018;72:712–719.
Wilson JR, Kerman SJ, Hubers SA, Yu C, Nian H, Grouzmann E, et al. Dipeptidyl peptidase 4 inhibition increases postprandial norepinephrine via substance P (NK1 receptor) during RAAS inhibition. J Endocr Soc. 2019;3:1784–98.
Hansen HH, Grønlund RV, Baader-Pagler T, Haebel P, Tammen H, Larsen LK, et al. Characterization of combined linagliptin and Y2R agonist treatment in diet-induced obese mice. Sci Rep. 2021;11:8060.
Bourdillon N, Eugster PJ, Vocat C, Nguyen T, Wuerzner G, Grouzmann E, et al. Saxagliptin: A potential doping agent? A randomized, double-blinded, placebo-controlled, and crossover pilot study in young active men. Physiol Rep. 2022;10:e15515.
Kalkan EA, Aydogan BI, Emra Kalkan EA. Aydoğan Bİ, Dinçer İ, Güllü S. Effects of DPP-4 inhibitors on brain natriuretic peptide, neuropeptide Y, glucagon like peptide-1, substance P levels and global longitudinal strain measurements in type 2 diabetes mellitus patients. J Health Sci Med. 2022;5:1424–30.
Dahan M, Zohar J, Todder D, Mathé AA, Cohen H. Exploring the anxiolytic potential of NPY by a dipeptidyl peptidase-IV inhibitor in an animal model of PTSD. Int J Neuropsychopharmacol 2024;27:pyae062.
Kamble M, Gupta R, Rehan HS, Gupta LK. Neurobehavioral effects of liraglutide and sitagliptin in experimental models. Eur J Pharmacol 2016;774:64–70.
Magdy YM, El-Kharashi OA, Nabih ES, Shaker SM, Abd-Elaziz LF, Aboul-Fotouh S. Potential involvement of JNK1 repression in the hepatic effect of sitagliptin and metformin in rats subjected to high fat diet and chronic mild distress. Biomed Pharmacother 2017;85:225–38.
Nazeem M, Wahdan SA, El-Naga RN, Gad AM. Saxagliptin ameliorated the depressive-like behavior induced by chronic unpredictable mild stress in rats: Impact on incretins and AKT/PI3K pathway. Eur J Pharmacol 2021;912:174602.
Soliman E, Essmat N, Mahmoud MF, Mahmoud AAA. Impact of some oral hypoglycemic agents on type 2 diabetes-associated depression and reserpine-induced depression in rats: the role of brain oxidative stress and inflammation. Naunyn Schmiedebergs Arch Pharmacol 2020;393:1391–1404.
Listos J, Listos P, Baranowska-Bosiacka I, Karpiuk A, Filarowska J, Łupina M, et al. Linagliptin, a selective dipeptidyl peptidase-4 inhibitor, reduces physical and behavioral effects of morphine withdrawal. Molecules 2022;27:2478.
Canneva F, Golub Y, Distler J, Dobner J, Meyer S, von Hörsten S. DPP4-deficient congenic rats display blunted stress, improved fear extinction and increased central NPY. Psychoneuroendocrinology 2015;53:195–206.
Frerker N, Raber K, Bode F, Skripuletz T, Nave H, Klemann C, et al. Phenotyping of congenic dipeptidyl peptidase 4 (DP4) deficient Dark Agouti (DA) rats suggests involvement of DP4 in neuro-, endocrine, and immune functions. Clin Chem Lab Med. 2009;47:275–87.
Karl T, Hoffmann T, Pabst R, von Hörsten S. Behavioral effects of neuropeptide Y in F344 rat substrains with a reduced dipeptidyl-peptidase IV activity. Pharm Biochem Behav 2003;75:869–79.
El Yacoubi M, Vaugeois JM, Marguet D, Sauze N, Guieu R, Costentin J, et al. Behavioral characterization of CD26 deficient mice in animal tests of anxiety and antidepressant-like activity. Behav Brain Res. 2006;171:279–85.
Marguet D, Baggio L, Kobayashi T, Bernard AM, Pierres M, Nielsen PF, et al. Enhanced insulin secretion and improved glucose tolerance in mice lacking CD26. Proc Natl Acad Sci Usa 2000;97:6874–79.
Bannon AW, Seda J, Carmouche M, Francis JM, Norman MH, Karbon B, et al. Behavioral characterization of neuropeptide Y knockout mice. Brain Res. 2000;868:79–87.
Karl T, Duffy L, Herzog H. Behavioural profile of a new mouse model for NPY deficiency. Eur J Neurosci 2008;28:173–80.
Zoicas I, von Hörsten S, Plank AC, Kornhuber J. Dipeptidyl peptidase-4 inhibitors enhance memory retention via neuropeptide Y. Eur J Pharmacol 2025;996:177556.
Gault VA, Lennox R, Flatt PR. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, improves recognition memory, oxidative stress and hippocampal neurogenesis and upregulates key genes involved in cognitive decline. Diabetes Obes Metab 2015;17:403–13.
Sim AY, Choi DH, Kim JY, Kim ER, Goh AR, Lee YH, et al. SGLT2 and DPP4 inhibitors improve Alzheimer’s disease-like pathology and cognitive function through distinct mechanisms in a T2D-AD mouse model. Biomed Pharmacother 2023;168:115755.
Zoicas I, Slattery DA, Neumann ID. Brain oxytocin in social fear conditioning and its extinction: involvement of the lateral septum. Neuropsychopharmacology 2014;39:3027–35.
Kornhuber J, Zoicas I. Social fear memory requires two stages of protein synthesis in mice. Int J Mol Sci. 2020b;21:5537.
Hamann CS, Bankmann J, Mora Maza H, Kornhuber J, Zoicas I, Schmitt-Böhrer A. Social fear affects limbic system neuronal activity and gene expression. Int J Mol Sci. 2022;23:8228.
Zoicas I, Huber SE, Kalinichenko LS, Gulbins E, Müller CP, Kornhuber J. Ceramides affect alcohol consumption and depressive-like and anxiety-like behavior in a brain region- and ceramide species-specific way in male mice. Addict Biol 2020;25:e12847.
Brazdis RM, Zoicas I, Kornhuber J, Mühle C. Brain region-specific expression levels of synuclein genes in an acid sphingomyelinase knockout mouse model: correlation with depression-/anxiety-like behavior and locomotor activity in the absence of genotypic variation. Int J Mol Sci. 2024;25:8685.
Varin EM, Mulvihill EE, Beaudry JL, Pujadas G, Fuchs S, Tanti JF, et al. Circulating levels of soluble dipeptidyl peptidase-4 are dissociated from inflammation and induced by enzymatic DPP4 inhibition. Cell Metab 2019;29:320–34.e5.
Baggio LL, Varin EM, Koehler JA, Cao X, Lokhnygina Y, Stevens SR, et al. Plasma levels of DPP4 activity and sDPP4 are dissociated from inflammation in mice and humans. Nat Commun. 2020;11:3766.
Yu SJ, Wang Y, Shen H, Bae EK, Li Y, Sambamurti K, et al. DPP-4 inhibitors sitagliptin and PF-00734,200 mitigate dopaminergic neurodegeneration, neuroinflammation and behavioral impairment in the rat 6-OHDA model of Parkinson’s disease. Geroscience 2024;46:4349–71.
Li J, Zhang S, Li C, Li M, Ma L. Sitagliptin rescues memory deficits in Parkinsonian rats via upregulating BDNF to prevent neuron and dendritic spine loss. Neurol Res. 2018;40:736–43.
Osman AS, Gad MH, Hareedy AA, Mishriki AA, Rasheed EAMA. Sitagliptin attenuates cognitive impairment in the rat model of aluminum-induced Alzheimer’s disease. J Adv Pharm Educ Res. 2019;9:53–61.
Khodir SA, Faried MA, Abd-Elhafiz HI, Sweed EM. Sitagliptin attenuates the cognitive deficits in L-methionine-induced vascular dementia in rats. Biomed Res Int. 2022;2022:7222590.
Zhang DD, Shi N, Fang H, Ma L, Wu WP, Zhang YZ, et al. Vildagliptin, a DPP4 inhibitor, alleviates diabetes-associated cognitive deficits by decreasing the levels of apoptosis-related proteins in the rat hippocampus. Exp Ther Med. 2018;15:5100–106.
Karl T, Hoffmann T, Pabst R, von Hörsten S. Extreme reduction of dipeptidyl peptidase IV activity in F344 rat substrains is associated with various behavioral differences. Physiol Behav 2003;80:123–34.
Fendt M, Bürki H, Imobersteg S, Lingenhöhl K, McAllister KH, Orain D, et al. Fear-reducing effects of intra-amygdala neuropeptide Y infusion in animal models of conditioned fear: an NPY Y1 receptor independent effect. Psychopharmacol (Berl) 2009;206:291–301.
Verma D, Wood J, Lach G, Mietzsch M, Weger S, Heilbronn R, et al. NPY Y2 receptors in the central amygdala reduce cued but not contextual fear. Neuropharmacology 2015;99:665–74.
Lach G, de Lima TC. Role of NPY Y1 receptor on acquisition, consolidation and extinction on contextual fear conditioning: dissociation between anxiety, locomotion and non-emotional memory behavior. Neurobiol Learn Mem. 2013;103:26–33.
Verma D, Tasan RO, Herzog H, Sperk G. NPY controls fear conditioning and fear extinction by combined action on Y1 and Y2 receptors. Br J Pharmacol 2012;166:1461–73.
Croce N, Ciotti MT, Gelfo F, Cortelli S, Federici G, Caltagirone C, et al. Neuropeptide Y protects rat cortical neurons against β-amyloid toxicity and re-establishes synthesis and release of nerve growth factor. ACS Chem Neurosci. 2012;3:312–18.
Peters J, Dieppa-Perea LM, Melendez LM, Quirk GJ. Induction of fear extinction with hippocampal-infralimbic BDNF. Science 2010;328:1288–90.
Yang SJ, Song ZJ, Wang XC, Zhang ZR, Wu SB, Zhu GQ. Curculigoside facilitates fear extinction and prevents depression-like behaviors in a mouse learned helplessness model through increasing hippocampal BDNF. Acta Pharm Sin. 2019;40:1269–78.
Colucci-D’Amato L, Speranza L, Volpicelli F. Neurotrophic factor BDNF, physiological functions and therapeutic potential in depression, neurodegeneration and brain cancer. Int J Mol Sci 2020;21:7777.
Decressac M, Wright B, David B, Tyers P, Jaber M, Barker RA, et al. Exogenous neuropeptide Y promotes in vivo hippocampal neurogenesis. Hippocampus 2011;21:233–38.
Bachor TP, Marquioni-Ramella MD, Suburo AM. Sitagliptin protects proliferation of neural progenitor cells in diabetic mice. Metab Brain Dis. 2015;30:885–93.
Anacker C, Hen R. Adult hippocampal neurogenesis and cognitive flexibility - linking memory and mood. Nat Rev Neurosci. 2017;18:335–46.
Kastin AJ, Akerstrom V. Nonsaturable entry of neuropeptide Y into brain. Am J Physiol 1999;276:E479–82.
Weina H, Yuhu N, Christian H, Birong L, Feiyu S, Le W. Liraglutide attenuates the depressive- and anxiety-like behaviour in the corticosterone induced depression model via improving hippocampal neural plasticity. Brain Res 2018;1694:55–62.
Abdelkawy YS, Elharoun M, Sheta E, Abdel-Raheem IT, Nematalla HA. Liraglutide and Naringenin relieve depressive symptoms in mice by enhancing neurogenesis and reducing inflammation. Eur J Pharmacol 2024;971:176525.
Yang F, Wang X, Qi J, Zhang K, Jiang Y, Feng B, et al. Glucagon-like peptide 1 receptor activation inhibits microglial pyroptosis via promoting mitophagy to alleviate depression-like behaviors in diabetic mice. Nutrients 2022;15:38.
de Paiva IHR, da Silva RS, Mendonça IP, de Souza JRB, Peixoto CA. Semaglutide attenuates anxious and depressive-like behaviors and reverses the cognitive impairment in a type 2 diabetes mellitus mouse model via the microbiota-gut-brain axis. J Neuroimmune Pharmacol 2024;19:36.
During MJ, Cao L, Zuzga DS, Francis JS, Fitzsimons HL, Jiao X, et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003;9:1173–79.
Yang S, Zhao X, Zhang Y, Tang Q, Li Y, Du Y, et al. Tirzepatide shows neuroprotective effects via regulating brain glucose metabolism in APP/PS1 mice. Peptides 2024;179:171271.
Zhang M, Yue X, Xu S, Piao J, Zhao L, Shu S, et al. Dipeptidyl peptidase-4 disturbs adipocyte differentiation via the negative regulation of the glucagon-like peptide-1/adiponectin-cathepsin K axis in mice under chronic stress conditions. FASEB J. 2024;38:e23684.
Reichmann F, Wegerer V, Jain P, Mayerhofer R, Hassan AM, Fröhlich EE, et al. Environmental enrichment induces behavioural disturbances in neuropeptide Y knockout mice. Sci Rep. 2016;6:28182.
Acknowledgements
We thank Laura Wormser und Peter Dietrich for providing the breeding pairs for the NPY −/− mice. We also thank Juliana Monti, Sabine Müller, Sarah Sheldrick and Jörg Distler for excellent technical assistance.
Funding
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. CM is a member of the research training group DFG-270949263/GRK2162. Open Access funding enabled and organized by Projekt DEAL.
Author information
Authors and Affiliations
Contributions
Iulia Zoicas: Conceptualization, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing—Original Draft, Writing—Reviewing and Editing. Christiane Mühle: Formal analysis, Methodology, Validation, Writing—Reviewing and Editing. Stephan von Hörsten: Resources, Writing—Reviewing and Editing. Anne-Christine Plank: Resources, Writing—Reviewing and Editing. Johannes Kornhuber: Resources, Writing—Reviewing and Editing.
Corresponding author
Ethics declarations
Competing interest
The authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Zoicas, I., Mühle, C., von Hörsten, S. et al. Sitagliptin as a therapeutic approach for social anxiety disorder: the role of DPP4 and NPY in modulating social fear and comorbid depressive-like behavior in mice. Neuropsychopharmacol. 50, 1724–1731 (2025). https://doi.org/10.1038/s41386-025-02146-8
Received:
Revised:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41386-025-02146-8