Skip to main content

Maternal separation and TNBS-induced gut inflammation synergistically alter the sexually differentiated stress response in rats

Abstract

Background

Neonatal maternal separation (MS) has been used to model long-lasting changes in behavior caused by neuroplastic changes associated with exposure to early-life stress. Earlier studies showed that transient gut inflammation can influence the development of irritable bowel syndrome (IBS). A prevailing paradigm of the etiology of IBS is that transient noxious events lead to long-lasting sensitization of the neural pain circuit, despite complete resolution of the initiating event. This study characterizes the changes in behaviors and neuroendocrine parameters after MS and early-phase trinitrobenzene sulfonic acid (TNBS)-induced colitis. We tested the hypothesis that MS and gut inflammation synergistically induce (1) hyperactivity in male rats and anxiety-like behaviors in female rats and (2) activation of the HPA axis in female rats and deactivation of the HPA axis in male rats after colorectal distention (CRD).

Methods

Male and female rat pups were separated from their dams for 180 min daily from postnatal day (PND) 2 to PND 14 (MS). Early-phase colitis was induced by colorectal administration with TNBS on PND 8. The elevated plus-maze test was performed at 7 weeks. Tonic CRD was performed at 60 mmHg for 15 min at 8 weeks. Plasma ACTH and serum corticosterone were measured at baseline or after the CRD. Analysis of variance was performed for comparison among controls, TNBS, MS, and MS + TNBS.

Results

In male rats, the time spent in open arms significantly differed among the groups (p < 0.005). The time spent in open arms in male MS + TNBS rats was significantly higher than that of controls (p < 0.009) or TNBS rats (p < 0.031, post hoc test). Female rats showed no difference in the time spent in open arms among the groups. MS and gut inflammation induced an increase in plasma ACTH in female rats but not in male rats at baseline.

Conclusions

These findings suggest that MS and gut inflammation synergistically induce hyperactive behavior or exaggerated hypothalamic–pituitary–adrenal axis function depending on sex.

Background

Early life experiences are one of the factors affecting psychological and physiological development and may lead to significant alterations in emotion and stress responses in later life [1,2,3,4,5]. Maternally separated animals have been intensively studied for decades and have also served as models of psychopathology [6, 7]. Especially, maternal separation (MS) is a well-characterized model of early-life stress used to study anxiety and depression [3, 8]. Various results have also been reported regarding fearful/anxiety-related behavior after MS protocols, with most separation schedules increasing fearful/anxiety-like behavior [9,10,11,12,13]. However, different behavioral phenotypes have also been reported depending on MS protocols, with some studies reporting hyperactive behavior [14,15,16,17]. Moreover, MS in rodents resulted in less anxious and more hyperactive behavior, which was considered to resemble attention-deficit hyperactive disorder (ADHD) [18, 19]. The factors determining animal behavior after MS are largely unknown.

Irritable bowel syndrome (IBS) is a common disorder characterized by chronic abdominal pain associated with alterations in bowel habits in the absence of a major organic pathology [20]. The original view of the disease as a primary disturbance in the gut is being conceptually refined to include a complex and disordered interaction between the brain and the gut [21, 22]. IBS is triggered or exacerbated by psychosocial stress [22,23,24]. In addition, early-life stressors increase the risk of IBS [25]. Psychological and physiological stress experienced in early life activates and triggers functional changes in visceral functions and visceral sensitivity to noxious stimuli [26,27,28]. In particular, MS induces gut dysfunction [29,30,31,32,33]. Patients with IBS have a higher rate of anxiety, depression, and somatization [34]. Individuals with ADHD have more diagnosis of IBS with odds ratio 1.67 with 95%CI from 1.56 to 1.80 than those without ADHD [35]. Not only stress but also gut inflammation is likely a causative factor for IBS [36]. Prospective studies indicated that a substantial proportion of patients (3–36%) with acute bacterial gastroenteritis develop IBS symptoms [37, 38]. Earlier studies also clarified that recovered animals after the experimental colitis induced by trinitrobenzene sulfonic acid (TNBS) show exaggerated response to colorectal distention (CRD) [39, 40]. These studies suggest that there may be a behavioral link between stress-related disorders (e.g., IBS, ADHD, and anxiety/depression/somatization) and early experience of noxious stimuli (e.g., MS and gut inflammation).

A previous study by our group clearly demonstrated that IBS patients show an exaggerated response to administration of corticotropin-releasing hormone (CRH) [41]. Sensory and motor dysfunctions of the colon in IBS patients are improved by administration of a CRH antagonist [42]. Moreover, the electrophysiological properties of the brain in IBS patients are also dramatically normalized by administration of a CRH antagonist [43]. In an animal model of IBS, CRH receptor-1 (CRH-R1) antagonist reverses anxiety-like behaviors induced by CRD [44]. Combination of previous TNBS colitis and repetitive CRD makes the colon hypersensitive [45]. Another study demonstrated that CRH-R1 is primarily involved in the water avoidance stress-induced colonic motor response [46]. Neonatal rats exposed to MS demonstrate altered function in the hypothalamic–pituitary–adrenal (HPA) axis [47, 48]. An earlier study indicated that neonatal trauma induced phenotypic changes in adulthood, including increased permeability of gut mucosa to stress via mechanisms involving CRH [31]. Therefore, it is natural to assume that altered HPA axis tone is involved in MS and recovered inflammation.

The unsolved pathogenesis and pathophysiology of brain-gut disorders is related to the sex difference [49, 50]. IBS [49, 50] and anxiety/depression [51] are more predominant in female individuals. Most studies on depression have reported with female:male ratios of 2:1 [51]. Increasing evidence from limited studies supports similar prevalence rates for pain-related symptoms in IBS, but a greater female predominance in non-pain-associated symptoms of constipation, bloating, and extraintestinal manifestations [49, 50]. Evidence suggests that IBS symptoms are influenced by the menstrual cycle, with an amplification of symptoms during the late luteal and early menses phases [49, 50]. By contrast, developmental disorder is predominant in male individuals [52]. Although most studies investigating the long-term effects of MS have used male rodents, those that studied both males and females have found significant sex differences in the effects of MS [9, 53]. Male and female rats may differ in numerous neuroendocrine and behavioral parameters, and vulnerability to stress may be sex dependent [54,55,56]. Therefore, precise behavioral comparisons between male and female subjects are necessary for the accurate understanding of stress-related disorders as a typical manifestation of the biopsychosocial model.

In the present study, we focused on the behavioral and neuroendocrine changes induced by MS and neonatal colorectal inflammation. Based on previous findings, we tested our hypotheses that MS and gut inflammation synergistically induce (1) hyperactivity in male rats and anxiety-like behaviors in female rats and (2) activation of the HPA axis in female rats and deactivation of the HPA axis in male rats after CRD.

Materials and methods

Animals

Timed-pregnant female Wister rats (n = 21) on gestation day 16 were obtained from Charles River Breeding Laboratories, Japan. Upon arrival, the rats were singly housed in the same room, which was maintained on a 12-h light/dark cycle (lights on at 8:00 am). Parturition was checked once a day (9:00 am), and the day of birth was considered PND 0. On PND 1, litters were cross-fostered (to minimize effects of genetic or prenatal variability) and adjusted to 12 pups each with about the same numbers of male and female pups. All dams were given free access to food and water on a 12-h light/dark cycle (lights on at 8:00 am) at a temperature of 23 ± 1 °C. Experimental conditions were assigned per litter. The irritation procedure and the experimental testing were conducted during the light component of the cycle. This study was based on the Guide for the Care and Use of Laboratory Animals and approved by the Animal Ethics Committee of Tohoku University School of Medicine.

Protocol

The experimental design was shown in Fig. 1. Rats were randomly assigned to four groups: (1) no separation and no inflammation (Control), (2) no separation and inflammation (TNBS), (3) MS and no inflammation (MS), and (4) MS and inflammation (MS + TNBS). The pups with MS were separated from their dams during PND 2 to 14, whereas the pups without MS remained with their mothers. To induce inflammation, rats were treated with TNBS (WAKO Pure Chemicals, Ltd., Tokyo, Japan) on PND 8. For no inflammation, rats were treated with vehicle (50% ethanol). In the 7th week, rats were exposed to an elevated plus-maze (EPM) and their behaviors were monitored and quantified. In the 8th week, rats were exposed to CRD as a visceral stimulus. After CRD for 15 min, the rats were killed by decapitation. The basal blood of each group was obtained without CRD. Blood was collected in a tube and the plasma was separated by centrifugation at 3000 rpm and stored at –30 °C.

Fig. 1
figure 1

Experimental design. Maternal separation (MS) procedures were performed from postnatal days (PNDs) 2 to 14. On PND 8, rats were exposed to TNBS-induced colitis and allowed to recover. At 7 weeks of age, rats were exposed to the elevated plus-maze test. At 8 weeks, colorectal distention (CRD) was performed

Maternal separation

Litters of 12 pups were randomly assembled for the fostering. The MS protocol was essentially the same as in earlier reports [31, 57,58,59]. Briefly, rats were exposed to a 180-min period of daily separation from dams (MS180) on PND 2 to 14. Separation was loaded at 9:00 am ± 30 min each day. The dams were removed from the maternity cages and placed into separate identical cages until the end of the manipulation. At dam removal, MS180 litters were removed as a group from the nest, weighed, and placed as a group in an isolation cage in an adjacent room. The cage was lined with chip bedding and placed in a neonatal cage and kept at 37 ± 0.5 °C by using a heating mat (KN–474; Natume, Tokyo, Japan) set underneath the cage. At the end of the daily separation, the pups were returned to their maternity cage and rolled in the soiled home cage bedding material before being reunited with the dam. On PND 22, all rats were weaned and the litters were housed in individual cages by sex and in the same treatment pairs.

Menstrual cycle

At the 7th week, the menstrual cycles of female rats were evaluated by a vaginal smear after Giemsa staining. On the basis of the microscopic reading of the smears, rats were subdivided according to the four phases of the cycle (proestrus, estrus, metestrus, and diestrus). Animal studies have shown increased responsiveness to greater autonomic manifestations during the metestrus/diestrus cycle phase, which corresponds to the perimenstrual phase in women [60, 61]. Therefore, all experiments in female rats were performed during the metestrus/diestrus phase.

Previous inflammation

The inflammation model of experimental colitis used here is well documented elsewhere [39, 40, 45]. Briefly, TNBS was dissolved in 50% ethanol to a concentration of 120 mg/ml. At PND8 during maternal separation rats were transferred in the paper container for treatment. A polyethylene-60 catheter was inserted 3 mm past the anus to lie approximately at the level of the splenic flexure. Rats were infused with 1.5 mg/50 µl of the TNBS plus ethanol solution. Control rats were similarly incubated but infused with 50 µL of the 50% ethanol vehicle only. This procedure involves a 6-week recovery period.

Elevated plus-maze test

Anxiety-like behavior was evaluated using a plexiglass EPM [44, 45]. The four arms (50 × 10 cm) were 1 m above the floor with a 10-cm center. The closed arms had 40-cm-high walls. Rats were placed in the center of the maze and were allowed to explore freely for 5 min; their behavior was recorded by video and using EthoVision software (TARGET system). The test belongs to the group of unconditioned anxiety models used for the development of putative anxiolytic compounds. The paradigm is based on rats’ innate aversion to open and high places.

Colorectal distention stimuli

The experimental CRD procedure and visceral sensitivity testing have been extensively reported elsewhere [62] and we essentially used the same method. The rat was lightly restrained in a plastic tube. A polyethylene balloon of 2.5 cm in diameter was inserted into the colorectum via the anus. The distal end of the balloon was positioned 1 cm proximal to the anus and was secured in place by taping the balloon catheter to the base of the tail. The balloon pressure, which represents intracolonic pressure, was continuously monitored online with the aid of a computerized barostat system (G and J Electronics Inc., Toronto, Canada). The colorectum was distended at a pressure of 60 mmHg for 15 min. CRD in rats causes an easily monitored pseudaffective response: a contraction of the abdominal and hind limb musculature (i.e., a visceromotor reflex) [62, 63].

Neuroendocrine function

Immediately after the rats were decapitated, blood was collected in chilled polyethylene tubes containing 200 µl (74.5 mg) of EDTA and separated with a centrifuge. The plasma was stored at –30 °C until analysis. Plasma adrenocorticotropic hormone (ACTH) and serum corticosterone were measured by radioimmunoassay. Some rats could provide less volume of blood for assay of ACTH and corticosterone. Thus, number of rats for HPA axis data was smaller than number of rats for behavioral experiments.

Statistical analysis

All data are expressed as mean ± SE. A two-tailed Student’s t-test was used to analyze differences between two groups. When more than two groups were compared, the significance among groups was evaluated by three-, two-, and one-way analysis of variance (ANOVA), and further statistical post hoc comparisons were performed using a post hoc Tukey’s test. A probability level of < 0.05 was considered to be statistically significant. All statistical calculations were performed using SPSS for Windows (ver. 12.0 J).

Results

Elevated plus-maze test

Table 1 shows the results of a three-way ANOVA performed with each category as a dependent variable and with sex, MS, and TNBS as independent variables. The results indicate that, concerning the time spent in open arms, the factors sex, MS, and the interaction between sex and MS were significant. This pattern was also seen in the percentage of time spent in open arms and the time spent in closed arms, as confirmed by significant effects of the same factors and same interactions. Regarding the time spent in open arm entries, ANOVA revealed significant main effects of sex (F1.102 = 12.073, p < 0.001), main effects of MS (F1.102 = 11.228, p < 0.001), and a sex × MS interaction (F1.102 = 4.772, p < 0.031). Total travel distance of three-way of ANOVA revealed significant main effects of sex (F1, 102 = 8.243, p < 0.005) and MS (F1, 102 = 4.379, p < 0.038). The results of the anxiety-related behavior in the EPM performance are presented in Figs. 2, 3, 4, 5, 6. All analyzed variables of the behavioral response to the EPM, except entries into closed arms, differed significantly between male rats (control, n = 12; TNBS, n = 16; MS, n = 15; and MS + TNBS, n = 15) and female rats (control, n = 12; TNBS, n = 16; MS, n = 11; and MS + TNBS, n = 13). In male rats, the time spent in open arms significantly differed among the groups (ANOVA, F3,54 = 4.795, p < 0.005). The time spent in open arms by male MS + TNBS rats (112.1 ± 10.3 s) was significantly higher than that of controls (50.4 ± 10.3 s, p < 0.009) or TNBS rats (63.1 ± 11 s, p < 0.031, post hoc test) (Fig. 2). Female rats showed no difference in the time spent in open arms among the groups (Fig. 2). The percentage of time spent in open arms by male rats significantly differed among the groups (ANOVA, F3,54 = 4.793, p < 0.005). The percentage of time spent in open arms by male MS + TNBS rats (37.4 ± 3.4%) was also significantly higher than that of controls (16.8 ± 3.4%, p < 0.009) or TNBS rats (21 ± 3.7%, p < 0.032, post hoc test) (Fig. 3). Female rats showed no difference in the percentage of time spent in open arms among the groups (Fig. 3). Entries into open arms were shown in Fig. 4. The percentage of open arm entries of male rats significantly differed among the groups (ANOVA, F3,54 = 4.023, p < 0.012). Male MS + TNBS rats showed a significantly higher percentage of open arm entries (44.7 ± 3.3%) than controls (23.4 ± 4.7%, p < 0.011, post hoc test) (Fig. 5). Female rats showed no difference in the percentage of open arm entries among the groups (Fig. 5). ANOVA of the time spent in closed arms by male rats revealed a significant difference among the groups (F3,54 = 4.795, p < 0.007). Male MS + TNBS rats spent significantly less time in closed arms (119.5 ± 10.3 s) than controls (179.6 ± 15, p < 0.014) or male TNBS rats (170 ± 10.5 s, p < 0.03, post hoc test) (Fig. 6). Female rats showed no difference in the time spent in closed arms among the groups (Fig. 6).

Table 1 Three-way ANOVA results of the elevated plus-maze
Fig. 2
figure 2

Time spent in open arms in the elevated plus-maze. Effects of early-life stress (MS) and/or previous inflammation (TNBS) on behavior are expressed as the mean ± SE. Control (male, n = 12; female, n = 12), TNBS (male, n = 16; female, n = 16), MS (male, n = 15; female, n = 11), and MS + TNBS (male, n = 15; female, n = 13). *P < 0.05, **P < 0.001 MS + TNBS vs. control. #P < .05, ##P < 0.001 MS + TNBS vs. TNBS. left, males; right, females

Fig. 3
figure 3

Percentage of time spent in open arms in the elevated plus-maze. See the legend of Fig. 3 for details. left, males; right, females

Fig. 4
figure 4

Entries into open arms in the elevated plus-maze. See the legend of Fig. 3 for details. left, males; right, females

Fig. 5
figure 5

Percentage of entries into open arms in the elevated plus-maze. See the legend of Fig. 2 for details. left, males; right, females

Fig. 6
figure 6

Time spent in closed arms in the elevated plus-maze. See the legend of Fig. 2 for details. left, males; right, females

Neuroendocrine function

Plasma ACTH

The results of the plasma ACTH concentration are presented in Table 2. Table 2 shows the results of a three-way ANOVA of plasma ACTH, indicating a significant sex difference in plasma ACTH. In the basal condition, plasma ACTH was shown to have a significant sex effect (F1.51 = 14.488, p < 0.0001), sex × MS interaction (F1.51 = 4.721, p = 0.034), and MS × TNBS interaction (F1.51 = 8.541, p = 0.005) with a trend interaction of sex × MS × TNBS (F1.51 = 3.333, p = 0.074). After the CRD, plasma ACTH showed a significant sex × MS × TNBS interaction (F1.51 = 8.418, p = 0.006).

Table 2 Three-way ANOVA results of plasma ACTH and serum corticosterone levels

In male rats, there was no significant difference in plasma ACTH among controls (n = 6), TNBS (n = 6), MS (n = 9), and MS + TNBS (n = 7) at baseline. There was no significant difference in plasma ACTH in rats after the CRD (control + CRD, n = 6; TNBS + CRD, n = 6; MS + CRD, n = 10; and MS + TNBS + CRD, n = 8). ANOVA revealed a significant main effect of CRD (F1.49 = 258.421, p < 0.0001) and a MS × TNBS interaction (F1.49 = 5.277, p < 0.026) (Fig. 7). In male rats, CRD induced a significantly different increase in plasma ACTH in controls (94.3 ± 30.7 pg/mL to 559.7 ± 53.2 pg/mL, p < 0.0001), TNBS (76.1 ± 13.4 pg/mL to 428.8 ± 47.9 pg/mL, p < 0.0001), MS (50.7 ± 6.3 pg/mL to 429.1 ± 32.9 pg/mL, p < 0.0001), and MS + TNBS (78.9 ± 24.1 pg/mL to 480.1 ± 49.8 pg/mL, p < 0.0001, post hoc test) (Fig. 7).

Fig. 7
figure 7

Plasma ACTH and serum corticosterone levels. All data are expressed as the mean ± SE (n = 6–10). Baseline (without colorectal distention [CRD]) and post-CRD values are shown. *P < 0.01, **P < 0.007, ***P < 0.0001 vs. same treatment (post hoc test following ANOVA). #P < 0.001 MS + TNBS vs. TNBS, +P < 0.05 MS + TNBS vs. MS (female rats)

On the other hand, female rats had a different plasma ACTH pattern among the groups (n = 6–10; Fig. 7). ANOVA revealed significant main effects of CRD (F1.49 = 86.295, p < 0.0001) and a MS × TNBS interaction (F1.49 = 10.590, p < 0.002). In female rats, CRD induced significant increases in plasma ACTH in controls (133.3 ± 13.7 pg/mL to 410.2 ± 65.2 pg/mL, p < 0.007), TNBS (106.2 ± 12.1 pg/mL to 573.5 ± 97.7 pg/mL, p < 0.005), and MS (104.4 ± 12.4 pg/mL to 651.1 ± 72.3 pg/mL, p < 0.0001). ANOVA of the plasma ACTH of female rats at baseline showed a significant difference among the groups (F3,27 = 5.186, p < 0.006). Female rats with MS + TNBS at baseline had a significantly higher plasma ACTH (278.3 ± 75.2 pg/mL) than those with TNBS (106.2 ± 12.1 pg/mL, p = 0.008) or MS (104.4 ± 12.4, p < 0.011, post hoc test). Therefore, CRD did not cause a significant increase in plasma ACTH in female rats with MS + TNBS.

Serum corticosterone

The results of the serum corticosterone concentration are presented in Fig. 7. Table 2 shows the results of three-way ANOVA of serum corticosterone, indicating a significant sex difference in serum corticosterone levels. In the basal condition, serum corticosterone was revealed to have a significant sex effect (F1.52 = 130.7, p < 0.0001) and MS × TNBS interaction (F1.52 = 6.843, p = 0.012). After the CRD, serum corticosterone showed a significant sex effect (F1.48 = 35.862, p < 0.0001).

The changes in the serum corticosterone of male rats were essentially similar to those seen for plasma ACTH (n = 6–10; Fig. 7). ANOVA revealed a significant main effect of CRD (F1.49 = 777.245, p < 0.0001). There was a trend interaction between MS × TNBS in serum corticosterone (F1.49 = 3.715, p < 0.060). CRD induced a significant increase in serum corticosterone in controls (75.2 ± 40.4 ng/mL to 531.9 ± 32.8 ng/mL, p < 0.0001), TNBS rats (55.1 ± 19.4 ng/mL to 521.3 ± 31.5 ng/mL, p < 0.0001), MS rats (30.4 ± 2.6 ng/mL to 462.5 ± 14.6 ng/mL, p < 0.0001), and MS + TNBS rats (54.5 ± 15.3 ng/mL to 534.5 ± 27 ng/mL, p < 0.0001, post hoc test).

The changes in the serum corticosterone of female rats were also similar to those in plasma ACTH (n = 6–10; Fig. 7). ANOVA revealed significant main effects of CRD on serum corticosterone (F1.50 = 176.051, p < 0.0001). In female rats, CRD induced a significant increase in serum corticosterone in controls (155.9 ± 37.1 ng/mL to 698.0 ± 75.9 ng/mL, p < 0.0001), TNBS (98.6 ± 17.7 ng/mL to 739.8 ± 59.4 ng/mL, p < 0.0001), MS (81.4 ± 24.2 ng/mL to 684.8 ± 34.6 ng/mL, p < 0.0001), and MS + TNBS (271.6 ± 112.5 ng/mL to 732.1 ± 94.7 ng/mL, p < 0.01).

Discussion

Hyperactive behaviors dominantly in male animals after MS + TNBS

This is the first study to show that a combination of early-life stress and gut inflammation may specifically lead to hyperactive behaviors in male animals. The EPM is believed to be a task that evaluates animals exposed to unconditioned fear or the anxiolytic properties of drugs [44, 63]. However, the idea that an increased presence of animals in the open arms of the EPM can be used as an index of impulsivity has been proposed by Ueno et al. [64]. There are several distinct animal models of ADHD, including spontaneously hypertensive rats (SHRs) [65] and stroke-prone spontaneously hypertensive (SHRSP) rats [66]. SHR and Wistar Kyoto rats display less anxiety-related behavior in the EPM than Sprague–Dawley rats [67]. SHRSP rats display higher motor activity, impulsivity, inattention, and dopaminergic hypofunction and decreased 5-hydroxytryptamine in the brain than SHRs [64]. Entries into open arms in the EPM task are also remarkably increased in SHRSP rats [64]. The percentage of time spent in open arms by male rats with MS + TNBS in this study (37.4 ± 3.4%) is higher than that of SHRSP rats in the report of Ueno et al. [64]. Therefore, it is natural to assume that a combination of early-life stress and gut inflammation may be one of the risk factors for comorbid ADHD and IBS in males.

Relevance of hyperactive behaviors and combination of recovered gut inflammation and early-life stress in animals and humans

The male:female sex ratio in ADHD children is approximately 9:1 [68, 69]. Inherent or genetic components related to the Y chromosome probably play important roles in the pathogenesis of ADHD [70]. It is of great interest to note distinct abnormal behavior predominantly in males after combined early trauma and gut inflammation. In even SHRSP rats, the most widely used model of ADHD, female rats also spend more time in open arms [64]. Therefore, at least for time spent in open arms, early trauma combined with gut inflammation seems to better reflect the features of hyperactive behavior. We originally expected that the combination of early trauma and gut inflammation in female rats would induce anxiety-like behavior, which is well known to be comorbid with IBS [34]. However, ADHD is also comorbid with IBS [35]. We previously reported that IBS patients show hypoactivity of the right dorsolateral prefrontal cortex during tasks needed cognitive flexibility [71]. There is a report that children with right hemisphere damage or dysfunction show symptoms of ADHD [72]. Therefore, the combination of early trauma and previous colonic inflammation may synergistically impair inhibitory function of the forebrain against the limbic brain in male animals, resulting in hyperactive behavior.

Dysregulation of homeostasis in rats after MS + TNBS

Hyperactive behavior and anxiety/depression are not mutually exclusive phenomena. The rate of comorbid psychiatric and learning problems, including anxiety and depression, ranges from 12 to 60% in patients with ADHD [52]. Both increased (hyperactive) and decreased (anxiety) time spent in the open arms of the EPM imply externally oriented and dysregulated non-homeostatic behaviors. Thus, an elevated basal plasma ACTH in female rats exposed to MS + TNBS may reflect interoception-oriented dysregulation of homeostasis. This may in part explain the female predominance of IBS in western countries with high levels of stress.

HPA axis in female rats after MS + TNBS

Early trauma [25] or previous gut inflammation [37] increases IBS risk and IBS patients show elevated ACTH [41]. In addition, elevated plasma ACTH correlates well with plasma interleukin-6 in IBS patients [73]. Neonatal rats exposed to MS also demonstrate exaggerated activation of the HPA axis [47, 48]. Reduced expression of hippocampal glucocorticoid receptors caused by negative feedback to the paraventricular nucleus of the hypothalamus [57] due to DNA methylation [58, 74] has been shown to be one of the causes of elevated plasma ACTH. Shanks et al. [75] reported that exposure of neonatal rats to a low dose of endotoxin causes long-lasting changes in the activity of the HPA axis and an elevated mean serum corticosterone concentration due to increased corticosterone pulse frequency and amplitude. Moreover, estrogen and progesterone likely play some sex role in the mechanism of IBS [49]. Because elevated plasma ACTH was selectively seen in the female rats of our study, the same mechanism is suggested to alter the HPA axis.

HPA axis in male rats after MS + TNBS

In the present study, the same levels of plasma ACTH and corticosterone were detected in male rats exposed to early trauma, previous inflammation, or their combination at baseline or at visceral stimulation. In contrast, an earlier study [74] showed an increased ACTH and corticosterone response to restraint stress in male rats treated by a low-licking mother. We cannot simply compare our data with those of earlier studies because early experience and gut manipulation are different each other. CRH and HPA axis function are not always parallel to exploratory behavior [44]. Therefore, our present data are not surprising and we cannot rule out the potential role of the CRH system in stress response and behavior after MS + TNBS in male rats.

Strength

There are some strengths in this study. The first strength is that both male and female rats were examined. Most previous studies were limited to male rats. However, female rats seem to be apparent hyperactive more frequently than male rats. We investigate hyperactive behaviors through a method directly compared with behaviors in male control rats and female control rats. These phenomena may reflect alternate anxiety symptoms in female rats, as discussed above, but the precise mechanism needs to be addressed. Second, the menstrual cycles of the female rats were precisely determined. Because visceral sensation and emotion depend on the menstrual cycle [49, 50, 60], the influence of this cycle on the results was excluded.

Limitation

There are some limitations to this study. The first limitation is that neither anxiety-like behaviors in female rats nor deactivation of the HPA axis in male rats after CRD was observed after MS + TNBS. Rather than that, plasma ACTH response to CRD was blunted in female rats after MS + TNBS. This is partially different from original hypothesis. However, it is concordant with basal increase in plasma ACTH in female rats after MS + TNBS and an earlier study with blunted responses of the HPA axis after chronic stress [76]. The second limitation is that molecular events inside the rats’ brains could not be examined. The detection of dopaminergic function or the 5-hydroxytryptaminergic molecule as well as modification of ADHD-like behavior using methylphenidate [64] is the future direction of this model. Third, the immune function of the gut has not yet been determined. In addition, whether these phenomena are specific to gut inflammation or found in other organs is unknown. A recent study indicated that immature rats with early-life stress have impaired intestinal barrier function that permits increased penetration of antigens and/or microbeads into the body [32]. They also reported that CRH antagonists may play a role in the colonic mucosal changes induced by MS. Visceral afferent nerve terminals have been shown to be sensitized by degranulated mast cells after acute stress [36]. Additionally, activation of enterochromaffin cells has also been suggested to play a role in this sensitization [31]. Exposure of neonatal rats to endotoxin has long-lasting effects on immune system regulation, including increased sensitivity of lymphocytes to stress-induced suppression of proliferation and remarkable protection against adjuvant-induced arthritis. Collins et al. [39] reported that stress caused a greater increase in myeloperoxidase activity in rats administered TNBS for 6 weeks than in controls, even though the plasma corticosterone levels were similar between the two groups. Moreover, previous colitis renders the colon more susceptible to the effects of stress on enteric nerve function and increases inflammation parameters in response to stress [39]. Therefore, molecular and immunological changes in the gut as well as the specificity of previous gut inflammation on behavioral or neuroendocrine changes should be addressed in a future study.

Conclusions

Our hypothesis that MS and gut inflammation synergistically induce hyperactivity in male rats but not in female rats was supported by our findings. In contrast, MS and gut inflammation synergistically induce exaggerated secretion of plasma ACTH after CRD in female rats. This study supports the hypothesis that there is a potent and long-lasting effect of neonatal exposure that can program major changes in the development of neuroendocrine, immunological regulatory, and behavioral characteristics.

Availability of data and materials

Datasets are available upon reasonable request.

Abbreviations

ACTH:

Adrenocorticotropic hormone

ADHD:

Attention-deficit/hyperactivity disorder

CRD:

Colorectal distention

EPM:

Elevated plus-maze

IBS:

Irritable bowel syndrome

MS:

Maternal separation

PND:

Post-natal day

TNBS:

Trinitrobenzene-sulfonic acid

References

  1. Levine S. Maternal and environmental influences on the adrenocortical response to stress in weanling rats. Science. 1967;156:258–60.

    Article  CAS  PubMed  Google Scholar 

  2. Francis DD, Champagne FA, Liu D, Meaney MJ. Maternal care, gene expression, and the development of individual differences in stress reactivity. Ann NY Acad Sci. 1999;896:66–84.

    Article  CAS  PubMed  Google Scholar 

  3. Sánchez MM, Ladd CO, Plotsky PM. Early adverse experience as a developmental risk factor for later psychopathology: evidencrom rodent and primate models. Dev Psychopathol. 2001;13:419–49.

    Article  PubMed  Google Scholar 

  4. Avishai-Eliner S, Brunson KL, Sandman CA, Baram TZ. Stressed-out, or in (utero)? Trends Neurosci. 2002;25:518–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Plotsky PM, Thrivikraman KV, Nemeroff CB, Caldji C, Sharma S, Meaney MJ. Long-term consequences of neonatal rearing on central corticotropin-releasing factor systems in adult male rat offspring. Neuropsychopharmacology. 2005;30:2192–204.

    Article  CAS  PubMed  Google Scholar 

  6. Colorado RA, Shumake J, Conejo NM, Gonzalez-Pardo H, Gonzalez-Lima F. Effects of maternal separation, early handling, and standard facility rearing on orienting and impulsive behavior of adolescent rats. Behav Processes. 2006;71:51–8.

    Article  PubMed  Google Scholar 

  7. Roceri M, Hendriks W, Racagni G, Ellenbroek BA, Riva MA. Early maternal deprivation reduces the expression of BDNF and NMDA receptor subunits in rat hippocampus. Mol Psychiatry. 2002;7:609–16.

    Article  CAS  PubMed  Google Scholar 

  8. Kuhn CM, Schanberg SM. Responses to maternal separatuion : mechanisms and mediators. Int J Dev Neurosci. 1998;16:261–70.

    Article  CAS  PubMed  Google Scholar 

  9. Barna I, Bálint E, Baranyi J, Bakos N, Makara GB, Haller J. Gender-specific effect of maternal deprivation on anxiety and corticotropin-releasing hormone mRNA expression in rats. Brain Res Bull. 2003;62:85–91.

    Article  CAS  PubMed  Google Scholar 

  10. Stone EA, Bonnet KA, Hofer MA. Survival and development of maternally deprived rats: role of body temprerature. Psychosom Med. 1976;38:242–9.

    Article  CAS  PubMed  Google Scholar 

  11. Janus K. Effects of early separation of young rats from the mother on their open-field behavior. Physiol Behav. 1987;40:711–5.

    Article  CAS  PubMed  Google Scholar 

  12. Matthews K, Robbins TW. Early experience as a determinant of adult behavioral responses to reward: the effects of repeated maternal separation in rat. Neurosci Biobehav Rev. 2003;27:45–55.

    Article  PubMed  Google Scholar 

  13. Penke Z, Felszeghy K, Femette B, Sage D, Nyakas C, Bulrlet A. Postnatal maternal deprivation produces long-lasting modifications of the stress response, feeding and stress-related behaviour in the rat. Eur J Neurosci. 2001;14:747–55.

    Article  CAS  PubMed  Google Scholar 

  14. Arnold JL, Siviy SM. Effects of neonatal handling and maternal separation on rough-and-tumble play in the rat. Dev Psychobiol. 2002;41:2005–15.

    Article  Google Scholar 

  15. Braun K, Kremz P, Weltzel W, Wagner T, Poeggle G. Influence of parental deprivation on the behavioral development in Octodon degus: modulation by maternal vocalizations. Dev Psychobiol. 2003;42:237–45.

    Article  PubMed  Google Scholar 

  16. Kaneko WM, Riley EP, Ehlers CL. Behavioral and electrophysiological effects of early repeated maternal separation. Depression. 1994;2:43–53.

    Article  Google Scholar 

  17. von Hoersten S, Dimitrijevic M, Markovior BM, Jankovic BD. Effect of early experience on behavior and immune response in the rat. Physiol Behav. 1993;54:931–40.

    Article  Google Scholar 

  18. Gonzalez-Lima F. Cortical and limbic systems mediating the predisposition to attention deficit and hyperactivity. In: Larimer MP, editor. Attention deficit and hyperactivity disorder research. New York: Nova Science; 2005. p. 1–18.

    Google Scholar 

  19. Jimenez-Vasquez PA, Mathe AA, Thomas JD, Riley EP, Ehlers CL. Early maternal separation alters neuropeptide Y concentrations in selected brain regions in adult rats. Dev Brain Res. 2001;131:149–52.

    Article  CAS  Google Scholar 

  20. Lacy BE, Mearin F, Chang L, Chey WD, Lembo AJ, Simren M, Spiller R. Bowel disorders. Gastroenterology. 2016;150:1393–407.

    Article  Google Scholar 

  21. Fukudo S, Nomura T, Muranaka M, Taguchi F. Brain-gut response to stress and cholinergic stimulation in irritable bowel syndrome. A preliminary study J Clin Gastroenteol. 1993;17:133–41.

    CAS  Google Scholar 

  22. Mayer EA, Naliboff BD, Chang L, Coutinho SVV. Stress and irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2001;280:G519-524.

    Article  CAS  PubMed  Google Scholar 

  23. Shinozaki M, Fukudo S, Hongo M, Shimosegawa T, Sasaki D, Matsueda K, Harasawa S, Miura S, Mine T, Kaneko H, Arakawa T, Haruma K, Torii A, Azuma T, Miwa H, Fukunaga M, Handa M, Kitamori S, Miwa T, IBS Club. High prevalence of irritable bowel syndrome in medical out-patients in Japan. J Clin Gastroenterol. 2008;42:1010–6.

    Article  PubMed  Google Scholar 

  24. Whitehead WE, Crowell MD, Robinson JC, Heller BR, Schuster MM. Effects of stressful life events on bowel symptoms: subjects with irritable bowel syndrome compared with subjects without bowel dysfunction. Gut. 1992;33:825–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Drossman DA. Personality and psychological factors in the irritable bowel syndrome. Gastroeterol Clin Biol. 1990;14:49–53.

    Google Scholar 

  26. Barreau F, Ferrier L, Fioramonti J, Bueno L. Neonatal maternal deprivation triggers long term alterations in colonic epithelial barrier and mucosal immunity in rats. Gut. 2004;53:501–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Barreau F, Cartier C, Ferrier L, Fioramonti J, Bueno L. Nerve growth factor mediates alterations of colonic sensitivity and mucosal barrier induced by neonatal stress in rats. Gastroenterology. 2004;127:524–34.

    Article  CAS  PubMed  Google Scholar 

  28. Murray CD, Flynn J, Ratcliffe L, Jacyna MR, Kamm MA, Emmanuel AV. Effect of acute physical and psychological stress on gut autonomic interaction in irritable bowel syndrome. Gastroenterology. 2004;127:1695–703.

    Article  PubMed  Google Scholar 

  29. Coutinho SV, Plotsky PM, Sablad M, Miller JC, Zhou H, Bayati AI, McRoberts JA, Mayer EA. Neonatal maternal separation alters stress-induced responses to viscerosomatic nociceptive stimuli in rat. Am J Physiol Gastrointest Liver Phisiol. 2002;282:G307-316.

    Article  CAS  Google Scholar 

  30. Rosztóczy A, Fioramonti J, Jármay K, Barreau F, Wittmann T, Buéno L. Influence of sex and experimental protocol on the effect of maternal deprivation on rectal sensitivity to distention in the adult rat. Neurogastroenterol Motil. 2003;15:679–86.

    Article  PubMed  Google Scholar 

  31. Söderholm JD, Yates DA, Gareau MG, Yang PC, MacQueen G, Perdue MH. Neonatal maternal separation predisposes adult rats to colonic barrier dysfunction in response to mild stress. Am J Physiol. 2002;283:G1257-1263.

    Google Scholar 

  32. Gareau MG, Jury J, Yang PC, MacQueen G, Perdue MH. Neonatal maternal separation causes colonic dysfunction in rat pups including impaired host resistance. Pediatr Res. 2006;59:83–8.

    Article  PubMed  Google Scholar 

  33. Accarie A, Vanuytsel T. Animal models for functional gastrointestinal disorders. Front Psychiatry. 2020;11:509681.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Levy RL, Olden KW, Naliboff BD, Bradley LA, Francisconi C, Drossman DA, Creed F. Psychosocial aspects of the functional gastrointestinal disorders. Gastroenterology. 2006;130:1447–58.

    Article  PubMed  Google Scholar 

  35. Kedem S, Yust-Katz S, Carter D, Levi Z, Kedem R, Dickstein A, Daher S, Katz LH. Attention deficit hyperactivity disorder and gastrointestinal morbidity in a large cohort of young adults. World J Gastroenterol. 2020;26:6626–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Fukudo S. Role of corticotropin-releasing hormone in irritable bowel syndrome and intestinal inflammation. J Gastroenterol. 2007;42(Suppl 17):48–51.

    Article  CAS  PubMed  Google Scholar 

  37. Gwee KA, Graham JC, McKendrick MW, Collins SM, Marshall JS, Walters SJ, Read NW. Psychometric scores and persistence of irritable bowel after infectious diarrhoea. Lancet. 1996;347:150–3.

    Article  CAS  PubMed  Google Scholar 

  38. Spiller R, Garsed K. Postinfectious irritable bowel syndrome. Gastroenterology. 2009;136:1979–88.

    Article  PubMed  Google Scholar 

  39. Collins SM, Mchugh K, Jacobson K, Khan I, Riddell R, Murase K, Weingarten HP. Previous inflammation alters the response of the rat colon to stress. Gastroenterology. 1996;111:1509–15.

    Article  CAS  PubMed  Google Scholar 

  40. Jacobson K, McHugh K, Collins SM. The mechanism of altered neural function in a rat model of acute colitis. Gastroenterology. 1997;112:156–62.

    Article  CAS  PubMed  Google Scholar 

  41. Fukudo S, Nomura T, Hongo M. Impact of corticotropin-releasing hormone on gastrointestinal motility and adrenocorticortropic hormone in normal controls and patients with irritable bowel syndrome. Gut. 1998;42:845–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sagami Y, Shimada Y, Tayama J, Nomura T, Satake M, Endo Y, Shoji T, Karahashi K, Hongo M, Fukudo S. Effect of a corticotropin-releasing hormone receptor antagonist on colonic sensory and motor function in patients with irritable bowel syndrome. Gut. 2004;53:958–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tayama J, Sagami Y, Shimada Y, Hongo M, Fukudo S. Effect of alphahelical CRH on quantitative electroencephalogram in patients with irritable bowel syndrome. Neurogastroenterol Motil. 2007;19:471–83.

    Article  CAS  PubMed  Google Scholar 

  44. Saito K, Kasai T, Nagura Y, Ito H, Kanazawa M, Fukudo S. Corticotropin-releasing hormone receptor 1 antagonist blocks brain-gut activation induced by colonic distention in rats. Gastroenterology. 2005;129:1533–43.

    Article  CAS  PubMed  Google Scholar 

  45. Saito-Nakaya K, Hasegawa R, Nagura Y, Ito H, Fukudo S. Corticotropin-releasing hormone receptor 1 antagonist blocks colonic hypersensitivity induced by a combination of inflammation and repetitive colorectal distension. Neurogastroenterol Motil. 2008;20:1147–56.

    Article  CAS  PubMed  Google Scholar 

  46. Maillot C, Million M, Wei JY, Gauthier A, Taché Y. Peripheral corticotropin-releasing factor and stress-stimulated colonic motor activity involved type 1 receptor in rats. Gastroenterology. 2000;119:1569–79.

    Article  CAS  PubMed  Google Scholar 

  47. Aborelius L, Owens MJ, Plotsky PM, Nemeroff CB. The role of corticotropin-releasing factor in depression and anxiety disorders. J Endocrinol. 1999;160:1–12.

    Article  Google Scholar 

  48. Newport DJ, Stowe ZN, Nemeroff CB. Parental depression: animal models of an adverse life event. Am J Psychiatry. 2002;159:1265–83.

    Article  PubMed  Google Scholar 

  49. Fukudo S. Sex and gender in irritable bowel syndrome. J Gastroenterol. 2006;41:608–10.

    Article  PubMed  Google Scholar 

  50. Chang L, Toner BB, Fukudo S, Guthrie E, Locke GR, Norton NJ, Sperber AD. Gender, age, society, culture, and the patient’s perspective in the functional gastrointestinal disorders. Gastroenterology. 2006;130:1435–46.

    Article  PubMed  Google Scholar 

  51. Weissman MM, Olfson M. Depression in women: implications for health care research. Science. 1995;269:799–801.

    Article  CAS  PubMed  Google Scholar 

  52. Furman L. What is attention-deficit hyperactivity disorder (ADHD)? J Child Neurol. 2005;20:994–1002.

    Article  PubMed  Google Scholar 

  53. Slotten HA, Kalinichev M, Hagan JJ, Marsden CA, Fone KCF. Long-lasting changes in behavioural and neuroendocrine indices in the rat following maternal separation: Gender-dependent effects. Brain Res. 2006;1097:123–32.

    Article  CAS  PubMed  Google Scholar 

  54. Lehmann J, Pryce CR, Bettschen D, Felden J. The maternal separation paradigm and adult emotionality and cognition in male and female Wistar rats. Pharmacol Biochem Behav. 1999;64:705–15.

    Article  CAS  PubMed  Google Scholar 

  55. Wigger A, Neumann ID. Periodic maternal deprivation induces gender-dependent alterations in behavioral and neuroendocrine responses to emotional stress in adult rats. Physiol Behav. 1999;66:293–302.

    Article  CAS  PubMed  Google Scholar 

  56. Farady MM, O’Donoghue VA, Grunberg NE. Effects of nicotine and stress on locomotion in Spraugue-Dawely and long-Evans male and female rats. Pharmacol Biochem Behav. 2003;74:325–33.

    Article  Google Scholar 

  57. Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D, Freedman A, Sharma S, Perason D, Plotsky PM, Meaney MJ. Maternal care, hippocampal glucocorticoid receptors, and hypothalamic-pituitary-adrenal responses to stress. Science. 1997;277:1659–62.

    Article  CAS  PubMed  Google Scholar 

  58. Liu D, Caldji C, Msarma S, Plotsky PM, Meaney MJ. Influence of neonatal rearing conditions on stress-induced adrenocorticotropin responses and norepinepherine release in the hypothalamic paraventricular nucleus. J Neuroendocrinol. 2000;12:5–12.

    Article  PubMed  Google Scholar 

  59. Coutinho SV, Plotsky PM, Sablad M, Miller JC, Zhou H, Bayati A, McRoberts JA, Mayre EA. Neonatal maternal separation alters stress-induced responses to viscerosomatic nociceptive stimuli in rat. Am J Physiol Gastrointest Liver Physiol. 2002;282:G307-316.

    Article  CAS  PubMed  Google Scholar 

  60. Chang L, Heitkemper MM. Gender differences in irritable bowel syndrome. Gastroenterology. 2002;123:1686–701.

    Article  PubMed  Google Scholar 

  61. Becker JB, Arnold AP, Berkley KJ, Blaustein JD, Eckel LA, Hampson E, Herman JP, Marts S, Sadee W, Steiner M, Taylor J, Young E. Strategies and methods for research on sex differences in brain and behavior. Endocrinology. 2005;146:1650–73.

    Article  CAS  PubMed  Google Scholar 

  62. Coutinho SV, Meller ST, Gebhart GF. Intracolonic zymosan produces visceral hyperalgesia in the rat that is mediated by spinal NMDA and non-NNDA receptors. Brain Res. 1996;736:7–15.

    Article  CAS  PubMed  Google Scholar 

  63. Saito K, Kanazawa M, Fukudo S. Colorectal distention induces hippocampal noradrenaline release in rat: an in vivo microdialysis study. Brain Res. 2002;947:146–9.

    Article  CAS  PubMed  Google Scholar 

  64. Ueno K, Togashi H, Mori K, Matsumoto M, Ohashi S, Hoshino A, Fujita T, Saito H, Minami M, Yoshioka M. Behavioural and pharmacological relevance of stroke-prone spontaneously hypertensive rats as an animal model of a developmental disorder. Behav Pharmacol. 2002;13:1–13.

    Article  PubMed  Google Scholar 

  65. Okamoto K, Aoki K. Development of a strain of spontaneously hyperactive rats. Jpn Circ J. 1963;27:282–93.

    Article  CAS  PubMed  Google Scholar 

  66. Okamoto K, Yamori Y, Nagaoka A. Establishment of the stroke-prone spontaneously hypertensive rat (SHR). Circ Res. 1974;34(35 Suppl):143–53.

    Google Scholar 

  67. Ferguson SA, Cada AM. Spatial learning/memory and social and no social behaviors in the spontaneously hypertensive, Wister-Kyoto and Sprague-Dawley rat strains. Pharmacol Biochem Behav. 2004;77:583–94.

    Article  CAS  PubMed  Google Scholar 

  68. Lahey BB, Applegate B, Biederman J, Greenhill L, Hynd GW, Barkley RA, Newcorn J, Jensen P, Richters J, Garfinkel B, Kerdyk L, Frick PJ, Ollendick T, Perez D, Hart EL, Waldman I, Shaffer D. DSM-IV filed trials for attention deficit hyperactivity disorder in children and adolescents. Am J Psychiat. 1994;151:1673–85.

    Article  CAS  PubMed  Google Scholar 

  69. Taylor E. Clinical foundation of hyperactivity research. Behav Brain Res. 1998;94:11–24.

    Article  CAS  PubMed  Google Scholar 

  70. Barkley RA. Attention-deficit hyperactivity disorder. Sci Am. 1998;297:43–50.

    Google Scholar 

  71. Aizawa E, Sato Y, Kochiyama T, Saito N, Izumiyama M, Morishita J, Kanazawa M, Shima K, Mushiake H, Hongo M, Fukudo S. Altered cognitive function of prefrontal cortex during error feedback in patients with irritable bowel syndrome, based on fMRI and dynamic causal modeling. Gastroenterology. 2012;143:1188–98.

    Article  PubMed  Google Scholar 

  72. Voeller KK. Right-hemisphere deficit syndrome in children. Am J Psychiatry. 1986;143:1004–9.

    Article  CAS  PubMed  Google Scholar 

  73. Dinan TG, Quigley PW, Ahmed SM, Scully P, O’Brien S, O’Mahony L, O’Mahony S, Shanahan F, Keeling PW. Hypothalamic-pituitary-gut axis dysregulation in irritable bowel syndrome: plasma cytokines as apotential biomarker? Gastroenterology. 2006;130:304–11.

    Article  CAS  PubMed  Google Scholar 

  74. Weaver ICG, Cervoni N, Champagne FA, Alessio ACD, Sharma S, Seckl JR, Dymov S, Szyf M, Meaney M. Epigenetic programming by maternal behavior. Nat Neurosci. 2004;8:847–54.

    Article  Google Scholar 

  75. Shanks N, Windle RJ, Perks PA, Harbuz MS, Jessop DS, Ingram CD, Lightman SL. Early-life exposure to endotoxin alters hypothalamic-pituitary-adrenal function and predisposition to inflammation. Proc Nat Acad Sci U S A. 2000;97(5):645–56.

    Google Scholar 

  76. Simpkiss JL, Devine DP. Responses of the HPA axis after chronic variable stress: effects of novel and familiar stressors. Neuro Endocrinol Lett. 2003;24:97–103.

    PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank members of Department of Behavioral Medicine for their assistance and support throughout the study. We also appreciate the help of the members of Tohoku University Animals Laboratory.

Funding

This research was supported by a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology, from the Ministry of Health, Welfare and Labor of Japan, and from the SRF. The sponsors had no role in the study design, in the collection, analysis, or interpretation of data, in the writing of the report, or in the decision to submit the article for publication.

Author information

Authors and Affiliations

Authors

Contributions

FS contributed to the experimental design and general guidance. MK provided general support. KN assisted in the preparation of the paper. RK provided experimental support. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Shin Fukudo.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Animal Ethics Committee of Tohoku University School of Medicine.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hasegawa, R., Saito-Nakaya, K., Gu, L. et al. Maternal separation and TNBS-induced gut inflammation synergistically alter the sexually differentiated stress response in rats. BioPsychoSocial Med 17, 7 (2023). https://doi.org/10.1186/s13030-022-00258-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13030-022-00258-x

Keywords