Next Article in Journal
From Protein Misfolding to Extracellular Matrix Disorganisation: Understanding Disease Pathology in Rare Skeletal Dysplasias
Previous Article in Journal
Stimulatory Effect of Aluminum in Root Development of Pogostemon cablin: Integration of ROS Homeostasis and Gene Expression Networks
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Portulaca oleracea Extract Modulates Diet-Dependent Neuroplasticity in a Murine Model of MCD-Induced NAFLD and Depression

by
Smaranda Ioana Mitran
1,2,†,
Mădălina Iuliana Muşat
1,3,†,
Cornelia Bejenaru
4,5,*,
George Dan Mogoşanu
4,6,*,
Ianis Kevyn Ştefan Boboc
1,7,
Robertina-Iulia Tudoraşcu
8,
Georgică Târtea
2,
Ovidiu Mircea Zlătian
9,10,
Antonia Blendea
4,5,
Andrei Biţă
4,6,
Adina-Elena Segneanu
11 and
Ludovic Everard Bejenaru
4,6
1
Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
2
Department of Physiology, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
3
Department of Scientific Research Methodology, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
4
Drug Research Center, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
5
Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
6
Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
7
Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
8
Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
9
Department of Microbiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 1 May Avenue, 200638 Craiova, Romania
10
Medical Laboratory, Emergency County Clinical Hospital, 1 Tabaci Street, 200642 Craiova, Romania
11
Institute for Advanced Environmental Research, West University of Timişoara (ICAM–WUT), 4 Oituz Street, 300086 Timişoara, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(20), 10050; https://doi.org/10.3390/ijms262010050
Submission received: 15 September 2025 / Revised: 12 October 2025 / Accepted: 14 October 2025 / Published: 15 October 2025
(This article belongs to the Section Bioactives and Nutraceuticals)

Abstract

Non-alcoholic fatty liver disease (NAFLD) is increasingly recognized as a systemic condition with neuropsychiatric comorbidities, including depression. Growing evidence for the neuroprotective, antidepressant, and anxiolytic potential of Portulaca oleracea (PO) extract, provides a compelling rationale for investigating its effects in the interaction between dietary models of NAFLD and vulnerability to stress-related disorders. Fifty-four 14- to 18-week-old male and female C57BL/6N mice were distributed in two equal groups and fed either a methionine- and choline-deficient diet (MCD) or a methionine- and choline-controlled diet (MC). Subsequently, half of each group was subjected to chronic unpredictable mild stress (CUMS) and PO treatment. MCD caused significant weight loss, whereas MC promoted weight gain. Behaviorally, MCD induced anhedonia- and anxiety-like behaviors, worsened by CUMS. MC diet reduced CUMS-induced anhedonia, though anxiety-like behavior emerged only under stress. Recognition memory was impaired in stressed MCD-fed mice, while MC-fed mice showed enhanced novel object preference. At the cellular level, MCD suppressed hippocampal microglia and caused cortical astrocyte dysfunction, whereas the MC diet promoted cortical neurogenesis potentiated through PO, abolished by chronic stress. These findings underscore the impact of dietary composition on PO’s systemic effects under chronic stress and support a mechanistic link between NAFLD-related dysfunction and depression-like phenotypes.

Graphical Abstract

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) represents one of the most common liver diseases worldwide and a major cause of liver-related mortality and morbidity [1]. It is a metabolic liver disease characterized by excessive accumulation of fat in the liver in the absence of significant alcohol consumption, hepatitis C, medication use, or hereditary disorders [2,3,4]. The illness can progress to more severe forms, such as non-alcoholic steatohepatitis, liver fibrosis, and, in advanced cases, cirrhosis and hepatocellular carcinoma [5,6]. The complexity of predisposing factors (lifestyle—mainly diet, genetic factors, chronic inflammation and mitochondrial dysfunction) [7,8,9] partially explains the involvement of extrahepatic organs (heart, kidney, pancreas, intestine, lung, bone). Being considered a systemic disease [10,11,12,13,14], the interest in the connection between NAFLD and the nervous system has increased in recent years. Since the first formulation of the hypothesis that NAFLD could be independently associated with cognition [15], research related it to bipolar disorder, schizophrenia [16], anxiety, depression, and chronic stress [17,18], cognitive impairments [19], cerebrovascular alterations [20], brain volume reduction [19], and insulin resistance of nervous tissue [21]. There seems to be an interdependent relationship between NAFLD and brain changes: the former aggravates the latter, and neurological treatments, mainly the psychiatric ones, can amplify liver damage [22]. Patients, regardless of mental condition and type of medication, have a higher predisposition to develop NAFLD [23] and thus be prone to all the complications of liver and mental diseases. In the literature, there is a paucity of data regarding innovative treatments that could offer hepatic and neuronal protection in NAFLD. Therefore, finding a treatment that would simultaneously improve liver and brain dysfunction could represent a real breakthrough.
Portulaca oleracea (PO) is the only spontaneous species of the Portulaca genus that grows in Romania [24,25]. It is one of the most useful medicinal plants, acting as diuretic, febrifuge, vermifuge, antiseptic, anti-spasmodic, and has pharmacological effects including analgesic, antibacterial, skeletal muscle relaxant, wound healing [26], radical scavenger [27], and antipyretic [28]. Furthermore, it has been used for mitigating insomnia, headaches [28], seizures [29], and liver inflammation [28,30]. Moreover, PO is known to regulate the lipid [28] and sugar [31] metabolism in animals.
Starting from the existing evidence of PO administration as being able to improve a series of both localized (digestive, renal, cutaneous, muscular, nervous) and systemic (anti-inflammatory, antipyretic, antioxidant, antiseptic) conditions, the present research aimed to study the effects of PO on lipid metabolism, inflammation, and possible correction of neuronal dysfunction in a well-known model of NAFLD induced by a methionine- and choline-deficient diet (MCD) [32], associated or not to a depression model triggered by chronic unpredictable mild stress (CUMS) [33]. As such, PO could represent a reliable, relatively inexpensive, and potentially dual therapeutic strategy for liver and brain diseases.

2. Results

2.1. Clinical and Behavioral Assessments

2.1.1. Methionine- and Choline-Deficient Diet Induces Weight Loss and Increases Vulnerability to Anhedonia-like Behavior, While Methionine- and Choline-Controlled Diet Promotes Weight Gain and Confers Protection Against Chronic Unpredictable Mild Stress-Induced Anhedonia

The two-way analysis of variance (ANOVA) test/analysis performed on animals’ weight revealed differences between sessions (F4.071,166.9 = 45.17, p < 0.0001) and treatments (F12,4 = 12.78, p < 0.0001). A significant interaction was also observed between sessions and treatments (F72,246 = 13.82, p < 0.0001). Post hoc analysis revealed that all animals that were fed an MCD diet exhibited a consistent weekly reduction in body weight until the end of the experiment, regardless of treatment (p < 0.01), whereas mice maintained on the methionine- and choline-controlled diet (MC) showed either stable or increased body weight throughout the experimental period (p < 0.05). No differences in body weight were observed in SHAM mice which were fed a standard diet (p > 0.05). All differences between groups at the end of the experiment were diet-related, with MCD mice exhibiting lower body weight compared to those on the MC diet, regardless of other experimental interventions. Mice in the MCD group exhibited lower body weight (16.00 ± 2.01 g) compared to the MC + CUMS (35.43 ± 4.38 g; p = 0.0144), MC + PO (33.75 ± 3.42 g; p = 0.0057), and MC + CUMS + PO animals (25.25 ± 1.50 g; p = 0.0088). Similarly, animals from the MCD + CUMS group showed reduced body mass (17.37 ± 1.94 g) compared to MC + CUMS (35.43 ± 4.38 g; p = 0.0196), MC + PO (33.75 ± 3.42 g; p = 0.0085), and MC + CUMS + PO mice (25.25 ± 1.50 g; p = 0.0163). In addition, MCD + CUMS + PO mice also presented with lower body mass (15.57 ± 1.20 g) compared with the MC + CUMS + PO group (25.25 ± 1.50 g; p = 0.0013) (Figure 1a).
Sucrose preference analysis using the t-test revealed that mice subjected to the MCD diet combined with the CUMS protocol exhibited a significant increase in anhedonia-like behavior from baseline to the end of the experiment, regardless of the treatment administered. Specifically, the MCD + CUMS group showed a reduction in sucrose preference from 76.96 ± 9.01% to 58.46 ± 3.66% (p = 0.0089, Cohen’s d = 2.68), the MCD + CUMS + vehicle (VEH) group from 87.06 ± 7.79% to 64.88 ± 6.91% (p = 0.0053, Cohen’s d = 3.00), and the MCD + CUMS + PO group from 82.95 ± 7.40% to 64.51 ± 8.42% (p = 0.0024, Cohen’s d = 2.32) (Figure 1b). Mice fed the MCD diet alone did not exhibit differences in sucrose preference (p = 0.2257, Cohen’s d = 0.95). Interestingly, MCD-fed mice that underwent the injection procedure showed a reduced sucrose preference, regardless of whether they received PO (88.29 ± 4.78% to 63.01 ± 9.63%, p = 0.0033, Cohen’s d = 3.32) or VEH (85.32 ± 7.61% to 68.36 ± 8.89%, p = 0.0274, Cohen’s d = 2.04) (Figure 1b).
In the context of MC diet administration, anhedonia-like behavior was not observed in any group, even when combined with the CUMS protocol (p > 0.05), with the exception of the MC + CUMS + PO group, which exhibited a reduction in sucrose preference from 89.18 ± 3.88% to 75.15 ± 10.43% (p = 0.0452, Cohen’s d = 1.78) (Figure 1c).
The Kruskal–Wallis test applied to the percent change in sucrose preference from baseline to post-treatment revealed increased anhedonia-like behavior in all MCD groups: MCD (−16.12 ± 12.46%; p = 0.0427), MCD + CUMS (−23.91 ± 8.34%; p = 0.0134), MCD + VEH (−19.29 ± 8.29%; p = 0.0134), MCD + PO (−28.89 ± 7.63%; p = 0.0181), MCD + CUMS + VEH (−24.90 ± 4.08%; p = 0.0051), MCD + CUMS + PO (−22.15 ± 8.22%; p = 0.0136), compared to SHAM (40.28 ± 68.28%). No differences were observed between MC-fed animals and SHAM controls (p > 0.05) (Figure 1d).

2.1.2. Anxiety-like Behavior Is Induced by the MCD Diet Alone, While MC Diet Induces Anxiety Only Under Chronic Stress Conditions

The open field test (OFT) data revealed increased anxiety-like behavior in mice fed the MCD diet, indicated by a significant reduction in time spent in the center of the arena from 65.73 ± 7.40 s to 19.31 ± 3.51 s (t-test; p < 0.0001, Cohen’s d = 8.00). A similar pattern was observed in the MCD + CUMS group, with a decrease from 146.5 ± 29.47 s to 73.14 ± 10.89 s (t-test; p = 0.0034, Cohen’s d = 3.30). Interestingly, animals from the same groups that received PO treatment did not exhibit enhanced anxiety-like behavior (p > 0.05). However, no significant differences were observed in the VEH-treated groups either (p > 0.05) (Figure 2a).
Regarding the administration of the MC diet, it did not induce anxiety-like behavior on its own (p = 0.1324), but only in combination with the CUMS protocol, as indicated by a reduction in time spent in the center of the arena from 94.78 ± 24.10 s to 57.97 ± 12.20 s (p = 0.0344, Cohen’s d = 1.92). This anxiety-like response was no longer observed in groups that received PO treatment (p > 0.05), nor in VEH-treated animals; although they showed a tendency to anxiety, the reduction in center time from 85.73 ± 34.44 s to 49.04 ± 8.41 s did not reach statistical significance (p = 0.0839, Cohen’s d = 1.46) (Figure 2b).
The exploratory activity during the test was illustrated using representative tracking paths of one animal from each relevant group (Figure 2c).
The percent change in anxiety-like behavior from baseline to post-treatment showed a significant reduction in center time in the MCD group compared to the SHAM group (p = 0.0066), with no significant differences detected among the other groups (p > 0.05) (Figure 2d).

2.1.3. The MCD Diet Impairs Recognition Memory in Chronically Stressed Mice, While the MC Diet Increases Preference for the Novel Object

Analysis of the novel object recognition test (NORT) data revealed a reduction in novel object preference in the MCD + CUMS group at the end of the experiment (40.11 ± 33.78%) compared to baseline (87.51 ± 11.42%) (t-test; p = 0.0376, Cohen’s d = 1.87), with no significant differences recorded for the other groups (Figure 3a).
In contrast, the MC + CUMS animals exhibited a significant increase in novel object preference from baseline (64.00 ± 14.18%) to the end of the experiment (88.43 ± 5.56%) (t-test; p = 0.0203, Cohen’s d = 2.21). A similar pattern was observed in the MC + PO group, where the preference for the novel object increased from 72.19 ± 10.22% to 100% (t-test; p = 0.0016, Cohen’s d = 3.84). No other significant differences were observed (p > 0.05) (Figure 3b).
The exploratory activity during the test was illustrated using representative tracking paths of one animal from each relevant group (Figure 3c).
The analysis of the percent change in novel object preference from baseline to post-treatment revealed a significant reduction in the MCD + CUMS group compared to the MC + CUMS group (Kruskal–Wallis test; p = 0.0132), as well as in the MCD + PO group compared to MC + PO animals (Kruskal–Wallis test; p = 0.0263). No significant differences were found among the remaining groups (p > 0.05) (Figure 3d).

2.2. Immunofluorescent Assessment of Neural and Glial Markers

2.2.1. Portulaca oleracea Treatment Fails to Reverse the Microglial Inhibition Induced by the MCD Diet in the Hippocampus

In the cortex, the analysis revealed differences between treatments (one-way ANOVA; F2,11 = 5.939, p = 0.0178), with MCD + CUMS + PO mice showing reduced ionized calcium binding adaptor molecule 1 (Iba1) density (12,891 ± 1287 cells/mm3) compared to SHAM (16,340 ± 2375 cells/mm3; p = 0.0140). No other differences were observed (p > 0.05) (Figure 4a,b).
We also observed differences between treatments at the hippocampal level (one-way ANOVA; F2,9 = 15.26, p = 0.0013), with MCD mice showing reduced Iba1 density (9809 ± 871.2 cells/mm3) compared to both SHAM (16,351 ± 2380 cells/mm3; p = 0.0037) and MC (17,080 ± 2487 cells/mm3; p = 0.0019) groups. PO treatment did not change this microglial inhibition, as MCD + PO mice still showed reduced Iba1 density (9800 ± 2548 cells/mm3) compared to SHAM (16,351 ± 2380 cells/mm3; p = 0.0225), with one-way ANOVA indicating differences between treatments (F2,9 = 5.907, p = 0.0230) (Figure 4c,d).

2.2.2. The MCD Diet Induces Astrocyte Dysfunction in the Cortex Regardless of Treatment, as Evidenced by Decreased Glial Fibrillary Acidic Protein Expression

Our analysis of glial fibrillary acidic protein (GFAP) expression in the cortex revealed differences between treatments (one-way ANOVA; F2,9 = 6.077, p = 0.0214), with a decreased GFAP-positive signal area in MCD mice (4245 ± 645.0 μm2) compared to both SHAM (8871 ± 3301 μm2; p = 0.0363), and MC (8940 ± 1729 μm2; p = 0.0339) groups (Figure 5a). Additionally, MCD + CUMS animals showed a decreased GFAP signal area (3825 ± 511.5 μm2) compared to SHAM mice (8871 ± 3301 μm2; p = 0.0138), with differences between treatments confirmed by one-way ANOVA (F2,9 = 6.561, p = 0.0175). A similar trend was observed in MCD + VEH (F2,9 = 9.614, p = 0.0058) and MCD + PO (F2,9 = 5.044, p = 0.0339) mice, which also showed reduced astrocytic signal (2281 ± 452.1 μm2, p = 0.0045, and 4103 ± 1310 μm2, p = 0.0356, respectively) compared to the SHAM group. PO treatment did not reverse this trend, as MCD + CUMS + PO mice also showed reduced GFAP signal (4296 ± 1120 μm2) compared to SHAM (p = 0.0108), with differences between groups confirmed by one-way ANOVA (F2,11 = 7.397, p = 0.0092) (Figure 5a,b).
In the hippocampus, we observed significant differences between treatments (one-way ANOVA; F2,9 = 4.305, p = 0.0488), with increased GFAP expression in MCD + CUMS mice (18,588 ± 4279 μm2) compared to MC + CUMS (12,294 ± 1287 μm2; p = 0.0406). This trend was also observed in the MCD + VEH group (25,526 ± 5057 μm2), compared to both SHAM (15,022 ± 2793 μm2; p = 0.0076) and MC + VEH (14.826 ± 2.735 μm2; p = 0.0068). One-way ANOVA also confirmed significant differences among treatments in this case (F2,9 = 11.01, p = 0.0038). No other differences were observed (p > 0.05) (Figure 5c,d).

2.2.3. The MC Diet Combined with PO Can Increase Cortical Neurogenesis, but This Effect Is Not Observed Under Chronic Stress Conditions

In order to assess the differences in terms of cortical neurogenesis, we quantified neuronal nuclei (NeuN)-positive neurons in the mouse cortex. Our analysis revealed differences between treatments (one-way ANOVA; F2,9 = 8.993, p = 0.0071), when comparing SHAM, MCD + PO, and MC + PO groups. Animals treated with PO, and which were fed the MC diet showed an increased number of NeuN-positive cells (2399 ± 129.6 cells/mm2) compared to SHAM (2132 ± 85.64 cells/mm2; p = 0.0317), and MCD + PO mice (2048 ± 143.5 cells/mm2; p = 0.0072). This difference was not observed in stressed groups or in those treated with VEH (p > 0.05) (Figure 6a,b).
No differences were observed between groups in terms of hippocampal NeuN-positive cell counts (Figure 6c,d).

3. Discussion

NAFLD became the most frequent chronic hepatic disease worldwide and its incidence is increasing at an alarming rate [32,33].
The effect of sugar-/lipid-rich and MCD diets on the liver and subsequently the nervous system are well documented [34,35,36,37,38,39,40,41].
Neurological changes associated with hepatic injury have been observed in both animal models [42,43,44,45], and in humans [46,47]. An improved knowledge of the liver–brain axis and of molecules that modulate it would favor a better understanding of the neuroimmune consequences generated by systemic diseases such as NAFLD.
Used as food for thousands of years, especially in Mediterranean and Asian areas, either raw or cooked [48], PO and its constituents have been attributed a series of effects in traditional medicine, such as anti-inflammatory, immunomodulatory, antioxidant, and antidiabetic [28,49,50,51].
In this context and based on data previously observed by our research team, the present study tried to observe the effect of PO in improving hepatic damage and neurological changes in a murine model of MCD-induced NAFLD, with and without chronic stress. The hepatic changes observed in our NAFLD model were consistent with the results previously obtained in our laboratory [17,45,52,53,54] and, at the same time, aligned with the overwhelming data reported in the literature on this well-established mouse model [55,56,57,58].
Our results confirm that the MCD diet induces a significant and progressive decrease in body weight [17,52,59,60,61], and the MC control diet maintains [62,63] or increases it [62,64]. We reproduced the clinical features of human NAFLD, with the predominance of increased weight [65,66,67] by using the MC diet especially conceived by MP Biomedicals (Eschwege, Germany) as a pair-fed control diet for MCD. Through having relatively high content in methionine, this type of diet is more faithful to the metabolic phenotype associated with NAFLD, explaining the differences observed in terms of weight evolution between the experimental groups.
Most studies report no significant sex-related differences in C57BL/6 mice following liver injury [68]. Traditionally, concerns about hormonal cycle variability [69,70] led to the preferential use of males in research [71]. However, recognizing sex as a biological variable has encouraged inclusion of both sexes [72]. In neuroscience, adding females does not increase variability [73] but allows broader conclusions. Aware that certain tests produce sex-dependent effects [74,75], we avoided the tail suspension test, known for gender bias [76]. Instead, to minimize sex and estrous influences, we used a mouse strain and behavioral paradigms less affected by sex, such as the OFT and the sucrose preference test (SPT) [74,76,77].
Behaviorally, we observed that animals which were fed the MCD diet and were exposed to CUMS showed a significant reduction in sucrose preference, presenting anhedonia-like behavior, whether they received PO extract or VEH only. This effect was not present in the MC groups, suggesting that the existence of liver damage increases vulnerability to stress-induced affective disorders. This confirms previous studies, thus strengthening the link between altered liver status and neurobehavioral dysfunction through mechanisms involving the liver–brain axis [17,43,52,78,79,80]. In this direction, our model supports the hypothesis that liver damage may increase susceptibility to depression-like behaviors. On the other hand, this observation supports the theory that dietary methionine and choline have a protective effect on mood [81,82,83,84,85]. However, regardless of the type of diet, and although not statistically significant, it seems that PO provides additional protection, given the disappearance of anxiety-like behavior in all treated groups.
It is noteworthy that the injection procedure appears to induce a decrease in sucrose preference in MCD animals, regardless of whether they receive PO or VEH. This would suggest that repeated injections may constitute a source of additional experimental stress, which confirms the scarce data in the literature showing that invasive manipulations can influence behavioral outcomes [86]. However, when comparing MC with MC–PO, no notable changes were observed. This might suggest that injectable administration does not significantly influence the behavior of the experimental animal in the absence of other stressors, especially since, shortly after the administration of the PO extract, mild abdominal discomfort was observed, which was absent in the case of VEH injection.
Furthermore, given the already published data documenting the intraperitoneal administration of PO in rats and mice with beneficial pharmacological effects including neurophysiological, anti-inflammatory, analgesic, and antioxidative [87,88,89], the effects observed in our study cannot be directly attributed to the PO, but rather to the experimental context. This highlights the importance of including SHAM groups in future behavioral protocols to dissociate the effect of the test substance from that of the procedure.
The OFT test demonstrated that the MCD diet induces anxiety even in the absence of chronic stress, consistent with previous research, while the MC diet generates anxious behavior only under exposure to CUMS stress, highlighting the interaction between liver damage and chronic stressors. PO extract administration did not exacerbate anxiety since the effect was not observed in the treated groups. Moreover, it seems to have protective effects as shown in studies on its antioxidant and neuroprotective properties [90,91,92,93,94]. The same pattern was observed with respect to the administration of VEH, on both MCD and MC foods, confirming the lack of adverse effects of ethanol when administered in 5% concentrations [95]. Thus, the severity of liver damage seems to be a determinant for the vulnerability to anxiety, with PO being able to modulate this relationship. However, future studies on metabolic models closer to human NAFLD and with non-invasive administration are needed to validate the effects.
In accordance with our previous research [17], recognition memory, investigated by the NORT assessment, was compromised by the MCD diet associated with chronic stress, while the MC diet favors the increase in preference for the novel object, suggesting distinct mechanisms depending on the severity of liver damage. These data are consistent with the literature describing MCD as a model of steatohepatitis with neuroinflammation and cognitive deficit [42,96], while metabolic models close to a physiological diet can generate protective adaptations under stress conditions, similar to the phenomenon of “stress inoculation therapy” [97]. PO administration did not correct the deficits induced by the MCD, but potentiated the positive effect of the MC diet, supporting the existence of the liver-brain axis and suggesting that PO can increase cognitive capacity in physiological contexts but is not able to counteract the severe impairment induced by MCD.
At the cellular level, the MCD diet induced hippocampal microglial inhibition and cortical astrocytic dysfunction, phenomena that were not corrected by PO administration. The literature describes the MCD diet as a model that generates severe intestinal and liver inflammation [98,99]. There is very little data on the direct link between MCD, NAFLD, and neuroinflammation (microglia and astrocytes) [43,45,52]. The morphology of hippocampal microglia is altered in the context of MCD associated with chronic stress, even at moderate levels, suggesting correlations between liver damage, behavioral stress, and neuroinflammation [43]. Although previous studies have reported neuroprotective effects of PO by reducing inflammation and oxidative stress in models of ischemia or neuronal toxicity [90,91,92,93,94], our data suggest that, under conditions of severe liver injury, its efficacy is limited. Furthermore, the divergent responses of astrocytes—cortical reduction vs. hippocampal increase—explain why a nonspecific intervention such as PO cannot completely restore glial homeostasis. Thus, the results highlight the need for more targeted therapies to correct neuroglial dysfunctions associated with liver diseases. However, the increase in preference for the novel object in animals that received the MC diet, correlated with the fact that at the cortical level, the same type of diet together with PO can generate neurogenesis, but not under chronic stress, raises the question whether a diversified diet enriched with PO has neurotrophic effects in physiological conditions. Taken together, our data suggest that PO exerts beneficial modulatory effects primarily under moderate metabolic conditions, while its efficacy appears limited in the context of severe liver injury.
It has been shown that humans with higher methionine intake presented improved cognitive function [100]. At the same time, studies on animal models with methionine restriction have shown the same beneficial cognitive effects [101,102], which further supports the possible potentiation of the positive effect or even an improved cognition due to the presence of PO in animals receiving the MC diet. The beneficial effects of PO cannot be attributed to the intake of contained amino acids through treatment, given the very low amounts of methionine in the standardized PO extract, respectively, the undetectable presence of choline. Studies proposing PO as a prophylactic treatment also go in the same direction, with plant extract counteracting induced apoptosis in the striatum of the rat brain [91].
The most recent data in the literature highlighted novel alkaloids obtained from PO as potent anti-inflammatory compounds—olerapyridin at 5 μM significantly reduced interleukin-1 beta (IL-1β) secretion, expression of inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) messenger ribonucleic acid (mRNA) levels. At a higher concentration of 15 μM, it additionally suppressed nitric oxide (NO) production, tumor necrosis factor-alpha (TNF-α) release, COX-2 protein, and iNOS mRNA expression. Another new alkaloid, oleracimine, demonstrated significant anti-inflammatory potential by suppressing NO production in vitro [103,104].
A potential limitation of this study is represented by the absence of a SHAM-injected group which would be necessary to control the potential peritoneal irritant effect of the plant and handling-induced stress.
To date, experimental studies demonstrate neuroprotective effects, reduction in neuronal apoptosis [91], modulation of neurotrophic factors (erythropoietin (EPO), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3)) [105], and functional improvement in various models of toxicity, neurodegeneration, and inflammation [106]. Moreover, most studies have focused on isolated effects on a single system or organ, without exploring its potential impact on systemic interactions, such as those between the liver and the central nervous system. To our knowledge, this is the first study related to the effects of PO on inflammation and neuroplasticity related to the liver–brain axis. Further studies are needed to elucidate the precise mechanisms underpinning the effects of PO in humans.

4. Materials and Methods

4.1. Chemicals and Reagents

The solvents used in this study included acetonitrile, formic acid and ethanol (Merck, Darmstadt, Germany). Ultrapure water was produced using a HALIOS 6 laboratory water system (Neptec, Montabaur, Germany). Borate buffer, hydrochloric acid (HCl), and fluorenylmethyl chloroformate (FMOC) were purchased from Sigma Aldrich (St. Louis, MO, USA).

4.2. Plant Material

The flowering aerial parts of wild-grown P. oleracea were harvested during the summer period (August 2023) from southwest Romania flora (Cârcea Commune, Dolj County, Oltenia Region). The vegetal samples for analysis were deposited in the Herbarium of the Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, Romania (voucher specimen PORT-OLR-2023-1508). Twenty-four hours before processing for extraction and analysis, the plant material was first air-dried and then deposited in brown paper bags, at room temperature (RT), in a cool and dark area. Endangered or protected herbal species are not included in our research.

4.3. Experimental Animals

The study was performed on 14–18 weeks old C57BL/6N male (n = 27) and female (n = 27) mice, housed at 20–23 °C in individual ventilated cages, and with a 12 h light/dark cycle. Food and water were available ad libitum. After being removed from the main colony, and before starting any procedures, the experimental animals were acclimatized for one week to the working laboratory conditions. The mice were obtained from the Animal Facility of the University of Medicine and Pharmacy of Craiova. All experimental protocols and animal care were approved by the Committee for Experimental Animals Wellbeing of the University of Medicine and Pharmacy of Craiova (Protocol Code No. 115/21 March 2024).

4.4. Depressive-like Behavior and Non-Alcoholic Fatty Liver Disease/Non-Alcoholic Steatohepatitis and CUMS Induction

For a subgroup of animals (26 mice, males and females), a non-alcoholic, non-viral hepatitis model for NAFLD was induced by replacing normal pelleted food with a pelleted diet lacking methionine and choline (D20, Figure 7) (MCD) (MP Biomedicals, Eschwege, Germany) [59,107]. A pair-fed control subgroup (24 mice) received pellet food containing precise amounts of methionine and choline, an identical formula in terms of calories with MCD, but with the exact addition of the two nutrients (Methionine/Choline Control diet with 2 g/kg Choline chloride and 3 g/kg D,L-methionine at expense of Sucrose—MC) (MP Biomedicals, Eschwege, Germany). The animals consumed MCD and MC ad libitum for six weeks and were weighed weekly throughout the experiment (Figure 1). After two weeks of pair-fed administration (D24, Figure 7), mice were randomly divided into another two subgroups. One subgroup (26 mice) was additionally submitted for four weeks to a depressive-like behavior protocol induced by CUMS (Table 1) [17]. Stressors were applied daily, one at a time, for four weeks, with no repeated procedure within three days [52,108,109,110,111]. No disturbances of any kind were allowed in the enclosures where the CUMS animals resided. Sham animals received normal pelleted food during the entire experiment, containing 4.56 g/kg methionine and 1.29 g/kg choline (Granulated combined feed. Complete feed for mice, rats and hamsters used for scientific or experimental purposes, Cantacuzino National Institute for Medical-Military Research and Development, Bucharest, Romania).

4.5. Portulaca oleracea Extract Preparation and Administration

4.5.1. Sample Preparation: Extraction, Hydrolysis, and Derivatization

High-performance liquid chromatography (HPLC)-grade acetonitrile and water, along with formic acid, were used for the mobile phase. FMOC was used for derivatization. HCl (6 M) was used for sample hydrolysis. Borate buffer and ethanol were used in the derivatization step.
Plant material (PO) extraction was carried out using an ultrasound-assisted extraction (UAE) method. Finely ground plant material (1 g) was mixed with 10 mL of 70% ethanol. The mixture underwent ultrasonic treatment in a Bandelin Sonorex Digiplus DL 102H (Bandelin electronic GmbH & Co. KG, Berlin, Germany) ultrasound bath (35 kHz, 100 W) for 20 min at 50 °C. The resulting solution was filtered through a 0.22 μm water-wettable polytetrafluoroethene (wwPTFE) syringe filter (Acrodisc, Pall Corporation, Port Washington, NY, USA). The filtered extract was then dried using a Heidolph Laborota 4000 (Heidolph Instruments GmbH & Co. KG, Schwabach, Germany) rotary evaporator.
For methionine analysis, the dried extract residue underwent ultrasound-assisted acid hydrolysis. Based on the optimized method for plant-based proteins, samples were hydrolyzed using 6 M HCl with ultrasound treatment for 30 min at 90 °C. This method utilizes cavitation effects to accelerate the hydrolysis process compared to traditional heating [112]. After hydrolysis, the samples were prepared for derivatization.
Prior to UHPLC injection, the amino acids in the hydrolysate were derivatized using FMOC. Following the principles outlined for FMOC derivatization, the reaction was performed using borate buffer (pH 9.0) and FMOC reagent (5 mM in ethanol), allowing for a short reaction time (5 min) [112]. This attaches the FMOC group (molecular weight (MW) 222.24 g/mol) to the methionine molecule (MW 149.21 g/mol), resulting in FMOC–methionine (MW 371.45 g/mol) (Figure 8a).

4.5.2. UHPLC–MS/PDA Analysis

Chromatographic analysis was performed on a Waters Acquity ARC system equipped with a 2998 Photodiode array (PDA) detector and a Waters QDa mass detector (Waters, Milford, MA, USA). Separation was achieved using a CORTECS C18 column (4.6 mm × 50 mm, 2.7 μm particle size). The mobile phase consisted of (A): water with 0.01% formic acid and (B): acetonitrile with 0.01% formic acid. The gradient elution started at 80% A, decreased linearly to 30% A over 10 min, followed by a 10 min re-equilibration period at initial conditions. The flow rate was maintained at 0.8 mL/min. The column temperature was set to 40 °C, and the autosampler temperature was kept at 8 °C. The injection volume was 10 μL.
PDA detection was performed by monitoring absorbance at 265 nm, characteristic of the FMOC chromophore. Mass spectrometry (MS) detection was conducted using the Waters QDa detector operating in positive electrospray ionization mode. The mass spectrum was scanned over an m/z range of 100–600. For targeted confirmation of FMOC–methionine, single ion recording (SIR) mode was used, monitoring the protonated molecule [M + H]+ at m/z 372. The capillary voltage was set to 0.8 kV, and the cone voltage was 15 V. Quantification was primarily based on the PDA signal at 265 nm, while the MS data served as confirmation of identity.
A calibration curve for L-methionine was constructed using standard solutions prepared from the Sigma Aldrich (St. Louis, MO, USA) reference material. Standards were subjected to the same FMOC derivatization procedure as the samples. The calibration range for the FMOC–methionine standard was: 10 μg/mL, 25 μg/mL, 50 μg/mL, 100 μg/mL, and 200 μg/mL.
The UHPLC–PDA method successfully separated FMOC-derivatized methionine from other components in the ultrasound-assisted acid hydrolysate of the PO extract. Quantification was performed using the calibration curve generated from FMOC-derivatized L-methionine standards, correlating peak area from the PDA detector at 265 nm to concentration. The identity of the methionine peak was confirmed by the Waters QDa mass detector operating in SIR mode, which showed a signal at m/z 372, corresponding to the [M + H]+ ion of FMOC–methionine. Based on the PDA quantification, the concentration of L-methionine in the analyzed PO sample was determined to be 551.963 ± 19.671 mg/100 g of the original plant product (Figure 8b).
Applying the same method to determine the PO composition in choline revealed levels below the detection limit.

4.5.3. Treatment Administration

The treatment groups (MCD + PO, MC + PO, MCD + CUMS + PO, MC + CUMS + PO) were administered PO in a dose of 200 mg extract/kg, in a 5% ethanol solution (2.4 g PO extract in 30 mL 5% ethanol) [113], prepared by the Departments of Botany and Pharmacology, University of Medicine and Pharmacy of Craiova. Mice received intraperitoneal injections with 50 μL solution daily (4 mg PO extract/animal), for a period of three weeks, with its permanent adjustment to the animal’s weight. Based on data available in the specialized literature regarding the amount of PO injected intraperitoneally, the administered dose was chosen, between those which only provide potentiation of the effects of other substances (12.5–100 mg/kg) [114,115], and those which have been shown to cause acute toxicity (1040 mg/kg) and/or 80% lethality (1000 mg/kg) [88]. Control groups (MCD + VEH, MC + VEH, MCD + CUMS + VEH, MC + CUMS + VEH) received the VEH alone (5% ethanol), representing 0.197 g/kg ethanol, less than half the dose at which no effect is observed [95].

4.6. Clinical Evaluation and Behavior Testing

All animals were weighed weekly throughout the experiment. Behavior testing was conducted both at baseline and at the end of the experimental period, using the SPT for anhedonia-like behavior, OFT for anxiety and exploratory behavior, and NORT in order to assess memory [116].
For the SPT test, mice were habituated with the presence of two bottles for four days, both filled with tap water. Then, they were given neither water nor food for 12 h prior to the test. For the test, the bottles have been checked, one filled with tap water and one with sucrose 2% and weighed. After 12 h, the position of the bottles was switched. The consumed volumes of water and sucrose were calculated to assess sucrose preference (% volume of sucrose consumption/total fluid consumption during the test) [44,52].
An open arena (50 cm (length) × 33 cm (width) × 15 cm (height)) and an EthoVision XT 17 (Noldus Information Technology, Leesburg, VA, USA) camera were used for OFT. After being individually placed in the center of the box, the movement of the mice was recorded for 10 min and then analyzed. The anxiety degree of the animal was estimated by comparing the time the mouse spent in the center of the arena to the one in the peripheral parts. After completing the test for one animal, the arena was cleaned with 75% ethanol to remove odors [53,54,117].
The same arena previously described was also used for NORT. Two identical objects were placed inside, and each mouse had 6 min to freely explore them. After one hour spent in the usual cage, the animal was placed again in the arena for another 6 min, this time to explore one object from the two previous ones and a new one. Both “6 min” sessions were recorded using the same camera (as for the OFT) and analyzed in order to calculate the preference (percentage of time spent exploring the new object compared to the total time spent exploring both) [52,53].

4.7. Immunofluorescent Staining of Paraformaldehyde-Fixed Brain Tissue

Following intraperitoneal anesthesia (Ketamine 100 mg/kg, Xylazine 10 mg/kg), animals were transcardially perfused with phosphate-buffered saline (PBS) (Thermo Fisher Scientific, Waltham, MA USA; 10010023), followed by 4% paraformaldehyde (PFA) (Thermo Fisher Scientific; 30525-89-4). The extracted brains were subsequently post-fixed overnight in 4% PFA at 4 °C to reduce microglial activation [118,119,120]. Immunofluorescent stains were performed on 35-μm-thick coronal brain sections, collected into 0.1 M PBS. For immunostaining, brain sections were initially blocked for one hour at RT in PBS containing 0.5% Triton X-100 and 5% horse serum (Thermo Fisher Scientific). This was followed by overnight incubation at 4 °C with the primary antibodies: mouse anti-NeuN monoclonal (Invitrogen, Carlsbad, CA, USA; MA5-33103; 1:500), goat anti-Iba1 (Abcam, Cambridge, UK; ab5076; 1:1000), and rabbit anti-GFAP polyclonal (Dako, Carpinteria, CA, USA; Z0334; 1:1000). After thorough washing, sections were incubated for two hours at RT in the dark with the secondary antibodies: Alexa Fluor 647 donkey anti-mouse (Invitrogen; A31571; 1:1000), Alexa Fluor 488 donkey anti-goat (Invitrogen; A11055; 1:1000), and Alexa Fluor 546 donkey anti-rabbit (Invitrogen; A10040; 1:1000). Finally, the stained sections were mounted and coverslipped using Fluoromount-G with 4′,6-diamidino-2-phenylindole (DAPI) (Thermo Fisher Scientific; 00-4959-52).

4.8. Image Acquisition and Analysis

For analysis, Z-stack images of cortex and hippocampus were taken using the 20× objective of an ApoTome, Axio Imager.Z2 microscope and Zen Software version 2.5 (Carl Zeiss, Jena, Germany). Iba1-positive cells were manually counted using Zen Software. In each image, cells were identified based on morphology. Only clearly labeled cells with defined cell bodies were included in the analysis. The total number of Iba1-positive cells was recorded, and cell density was calculated. NeuN-positive cells were analyzed using QuPath 0.6.0 software (University of Edinburgh, Edinburgh, UK). Detection parameters such as threshold levels, minimum/maximum cell size, and background subtraction were optimized to reduce false positives and improve accuracy. After detection, cell density was also calculated. The GFAP-positive signal area within the cortical and hippocampal regions was quantified for each animal using Fiji ImageJ 2.0.0 and Image-Pro Plus 11 (Media Cybernetics, Bethesda, MD, USA). Image channels were converted to grayscale, and adjustments to intensity, brightness, and contrast were made to enhance signal clarity, reduce background interference, and facilitate accurate quantification. Prior to analysis, acquisition parameters were calibrated to ensure measurement consistency. Following calibration, the signal quantification tools within the software were employed to assess both the intensity of the GFAP signal. For each animal, four representative images from the cortex and four from the hippocampus were analyzed. The values obtained from these individual images were used to calculate the mean for each marker. This mean value was considered the final representative data for each animal in both cortical and hippocampal regions.

4.9. Statistical Analysis

Statistical analyses were conducted using GraphPad 10.3.1 (GraphPad Software, Inc., San Diego, CA, USA) and Microsoft Excel 16.96 (Microsoft Corp., Redmond, WA, USA). The figures were created using Adobe InDesign 20.4.1 (Adobe, San Jose, CA, USA) and App.diagrams.net v27.2.0 (JGraph Ltd., Northampton, UK). Differences in means among the groups were analyzed using t-test, one or two-way ANOVA (Tukey’s multiple comparisons test), with repeated measurements and Geisser–Greenhouse correction, after the data set passed normality testing (Shapiro–Wilk test), and the Kruskal–Wallis test (Dunn’s multiple comparisons test) for non-parametric data. For ANOVA test, Sessions (weekly results) were used as a within-factor, and Treatments (diets or treatments) were considered as a between-factor. All data are presented as mean ± standard deviation (SD), and statistical significance is indicated as follows: * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Effect sizes were interpreted according to Cohen’s conventions, with d > 0.8 indicating large effects, and d > 1.3 reflecting very large effects.

5. Conclusions

Based on our findings, we can state that although PO has well-documented metabolic effects (anti-inflammatory, antidiabetic, hypolipidemic, antioxidant) in preclinical and clinical studies, its impact on neuropsychiatric behavior remains poorly understood. Our results suggest that this plant could interact with the metabolic status and chronic environmental stress, but the interpretation is complicated by confounding factors induced by the method of administration. Therefore, future studies should use less invasive forms of administration (e.g., oral) and metabolic models closer to human NAFLD to clarify the real role of PO in the relationship between liver diseases and vulnerability to affective disorders.
Patients with NAFLD should be concerned with changes in their lifestyle (physical activity, weight loss), along with an early approach to pharmaceutical treatments, including herbal ones, to influence extrahepatic manifestations. Existing data support the metabolic benefits of PO. The added value of this study involves the neurobehavioral component and diet-linked neuroprotective potential of PO, thus providing the foundation for new research avenues.

Author Contributions

Conceptualization, S.I.M., M.I.M. and L.E.B.; methodology, S.I.M., M.I.M., C.B., G.D.M. and L.E.B.; software, M.I.M.; validation, S.I.M., C.B., G.D.M. and L.E.B.; formal analysis, A.B. (Andrei Biţă); investigation, S.I.M., M.I.M., I.K.Ş.B. and A.B. (Antonia Blendea); resources, S.I.M. and L.E.B.; data curation, I.K.Ş.B., R.-I.T., G.T., O.M.Z. and A.B. (Antonia Blendea); writing—original draft preparation, S.I.M., M.I.M. and A.B. (Andrei Biţă); writing—review and editing, S.I.M., M.I.M., C.B., G.D.M., A.-E.S. and L.E.B.; supervision C.B., G.D.M., A.-E.S. and L.E.B.; funding acquisition, S.I.M. and L.E.B. All authors have read and agreed to the published version of the manuscript.

Funding

The Article Processing Charges were funded by the University of Medicine and Pharmacy of Craiova, Romania. This work was supported by Internal Research Project No. 26/725/11/25 July 2024.

Institutional Review Board Statement

The animal study protocol was approved by the Committee for Experimental Animals Wellbeing of the University of Medicine and Pharmacy of Craiova, Romania (Approval No. 115 from 21 March 2024).

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author.

Acknowledgments

The authors would like to thank Bogdan Cătălin and Oltin Pop for their valuable support and constructive advice.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
ANOVAAnalysis of variance
BDNFBrain-derived neurotrophic factor
COX-2Cyclooxygenase-2
CUMSChronic unpredictable mild stress
DDay
DAPI4′,6-Diamidino-2-phenylindole
EPOErythropoietin
FMOCFluorenylmethyl chloroformate
GFAPGlial fibrillary acidic protein
HClHydrochloric acid
HPLCHigh-performance liquid chromatography
Iba1Ionized calcium-binding adaptor molecule 1
IL-1βInterleukin-1 beta
iNOSInducible nitric oxide synthase
MCMethionine- and choline-controlled diet
MCDMethionine- and choline-deficient diet
mRNAMessenger ribonucleic acid
MSMass spectrometry
MWMolecular weight
NAFLDNon-alcoholic fatty liver disease
NeuNNeuronal nuclei
NONitric oxide
NORTNovel object recognition test
NT-3Neurotrophin-3
OFTOpen field test
PBSPhosphate-buffered saline
PDAPhotodiode array
PFAParaformaldehyde
POPortulaca oleracea
RTRoom temperature
SDStandard deviation
SIRSingle ion recording
SPTSucrose preference test
TNF-αTumor necrosis factor-alpha
tRRetention time
UAEUltrasound-assisted extraction
UHPLCUltra-high-performance liquid chromatography
VEHVehicle
wwPTFEWater-wettable polytetrafluoroethene

References

  1. Whalley, S.; Puvanachandra, P.; Desai, A.; Kennedy, H. Hepatology outpatient service provision in secondary care: A study of liver disease incidence and resource costs. Clin. Med. 2007, 7, 119–124. [Google Scholar] [CrossRef]
  2. Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; Romero, D.; Abdelmalek, M.F.; Anstee, Q.M.; Arab, J.P.; et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Hepatology 2023, 78, 1966–1986. [Google Scholar] [CrossRef] [PubMed]
  3. Wong, V.W.; Chan, W.K.; Chitturi, S.; Chawla, Y.; Dan, Y.Y.; Duseja, A.; Fan, J.; Goh, K.L.; Hamaguchi, M.; Hashimoto, E.; et al. Asia-Pacific Working Party on Non-alcoholic Fatty Liver Disease guidelines 2017-Part 1: Definition, risk factors and assessment. J. Gastroenterol. Hepatol. 2018, 33, 70–85. [Google Scholar] [CrossRef]
  4. Younossi, Z.M.; Stepanova, M.; Rafiq, N.; Makhlouf, H.; Younoszai, Z.; Agrawal, R.; Goodman, Z. Pathologic criteria for nonalcoholic steatohepatitis: Interprotocol agreement and ability to predict liver-related mortality. Hepatology 2011, 53, 1874–1882. [Google Scholar] [CrossRef]
  5. Rinella, M.E.; Sanyal, A.J. Management of NAFLD: A stage-based approach. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 196–205. [Google Scholar] [CrossRef]
  6. Adams, L.A.; Sanderson, S.; Lindor, K.D.; Angulo, P. The histological course of nonalcoholic fatty liver disease: A longitudinal study of 103 patients with sequential liver biopsies. J. Hepatol. 2005, 42, 132–138. [Google Scholar] [CrossRef]
  7. Li, L.; Liu, D.-W.; Yan, H.-Y.; Wang, Z.-Y.; Zhao, S.-H.; Wang, B. Obesity is an independent risk factor for non-alcoholic fatty liver disease: Evidence from a meta-analysis of 21 cohort studies. Obes. Rev. 2016, 17, 510–519. [Google Scholar] [CrossRef]
  8. Cho, J.-Y.; Chung, T.-H.; Lim, K.-M.; Park, H.-J.; Jang, J.-M. The impact of weight changes on nonalcoholic Fatty liver disease in adult men with normal weight. Korean J. Fam. Med. 2014, 35, 243–250. [Google Scholar] [CrossRef]
  9. Huh, Y.; Cho, Y.J.; Nam, G.E. Recent Epidemiology and Risk Factors of Nonalcoholic Fatty Liver Disease. J. Obes. Metab. Syndr. 2022, 31, 17–27. [Google Scholar] [CrossRef] [PubMed]
  10. Musso, G.; Gambino, R.; Tabibian, J.H.; Ekstedt, M.; Kechagias, S.; Hamaguchi, M.; Hultcrantz, R.; Hagström, H.; Yoon, S.K.; Charatcharoenwitthaya, P.; et al. Association of non-alcoholic fatty liver disease with chronic kidney disease: A systematic review and meta-analysis. PLoS Med. 2014, 11, e1001680. [Google Scholar] [CrossRef] [PubMed]
  11. Kawasaki, T.; Hashimoto, N.; Kikuchi, T.; Takahashi, H.; Uchiyama, M. The relationship between fatty liver and hyperinsulinemia in obese Japanese children. J. Pediatr. Gastroenterol. Nutr. 1997, 24, 317–321. [Google Scholar] [CrossRef]
  12. Abu-Shanab, A.; Quigley, E.M.M. The role of the gut microbiota in nonalcoholic fatty liver disease. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 691–701. [Google Scholar] [CrossRef]
  13. Miele, L.; Valenza, V.; La Torre, G.; Montalto, M.; Cammarota, G.; Ricci, R.; Mascianà, R.; Forgione, A.; Gabrieli, M.L.; Perotti, G.; et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009, 49, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
  14. Rosato, V.; Masarone, M.; Dallio, M.; Federico, A.; Aglitti, A.; Persico, M. NAFLD and Extra-Hepatic Comorbidities: Current Evidence on a Multi-Organ Metabolic Syndrome. Int. J. Environ. Res. Public Health 2019, 16, 3415. [Google Scholar] [CrossRef]
  15. Seo, S.W.; Gottesman, R.F.; Clark, J.M.; Hernaez, R.; Chang, Y.; Kim, C.; Ha, K.H.; Guallar, E.; Lazo, M. Nonalcoholic fatty liver disease is associated with cognitive function in adults. Neurology 2016, 86, 1136–1142. [Google Scholar] [CrossRef]
  16. Penninx, B.W.J.H.; Lange, S.M.M. Metabolic syndrome in psychiatric patients: Overview, mechanisms, and implications. Dialogues Clin. Neurosci. 2018, 20, 63–73. [Google Scholar] [CrossRef]
  17. Muşat, M.I.; Mitran, S.I.; Udriştoiu, I.; Albu, C.V.; Cătălin, B. The impact of stress on the behavior of C57BL/6 mice with liver injury: A comparative study. Front. Behav. Neurosci. 2024, 18, 1358964. [Google Scholar] [CrossRef] [PubMed]
  18. Ng, C.H.; Xiao, J.; Chew, N.W.S.; Chin, Y.H.; Chan, K.E.; Quek, J.; Lim, W.H.; Tan, D.J.H.; Loke, R.W.K.; Tan, C.; et al. Depression in non-alcoholic fatty liver disease is associated with an increased risk of complications and mortality. Front. Med. 2022, 9, 985803. [Google Scholar] [CrossRef] [PubMed]
  19. Filipović, B.; Marković, O.; Đurić, V.; Filipović, B. Cognitive Changes and Brain Volume Reduction in Patients with Nonalcoholic Fatty Liver Disease. Can. J. Gastroenterol. Hepatol. 2018, 2018, 9638797. [Google Scholar] [CrossRef]
  20. Hadjihambi, A. Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer’s disease? Anal. Biochem. 2022, 636, 114387. [Google Scholar] [CrossRef]
  21. Marušić, M.; Paić, M.; Knobloch, M.; Liberati Pršo, A.-M. NAFLD, Insulin Resistance, and Diabetes Mellitus Type 2. Can. J. Gastroenterol. Hepatol. 2021, 2021, 6613827. [Google Scholar] [CrossRef] [PubMed]
  22. Shaheen, A.A.; Kaplan, G.G.; Sharkey, K.A.; Lethebe, B.C.; Swain, M.G. Impact of major depression and antidepressant use on alcoholic and non-alcoholic fatty liver disease: A population-based study. Liver Int. 2021, 41, 2308–2317. [Google Scholar] [CrossRef] [PubMed]
  23. Jadhav, P.A.; Thomas, A.B.; Chopada, V.M.; Bokaria, P.V.; Deokate, S.B.; Chougule, P.S.; Chavan, P.N.; Chitlange, S.S. Correlation of non-alcoholic fatty liver disease and neurodegenerative disorders. Egypt. Liver J. 2024, 14, 79. [Google Scholar] [CrossRef]
  24. Srivastava, R.; Srivastava, V.; Singh, A. Multipurpose Benefits of an Underexplored Species Purslane (Portulaca oleracea L.): A Critical Review. Environ. Manag. 2023, 72, 309–320. [Google Scholar] [CrossRef]
  25. Segneanu, A.-E.; Vlase, G.; Marin, C.N.; Vlase, T.; Sicoe, C.; Herea, D.D.; Ciocîlteu, M.V.; Bejenaru, L.-E.; Minuti, A.E.; Zară, C.-M.; et al. Wild grown Portulaca oleracea as a novel magnetite based carrier with in vitro antioxidant and cytotoxicity potential. Sci. Rep. 2025, 15, 8694. [Google Scholar] [CrossRef]
  26. Lee, A.S.; Kim, J.S.; Lee, Y.J.; Kang, D.G.; Lee, H.S. Anti-TNF-α activity of Portulaca oleracea in vascular endothelial cells. Int. J. Mol. Sci. 2012, 13, 5628–5644. [Google Scholar] [CrossRef]
  27. Askari, V.R.; Rezaee, S.A.; Abnous, K.; Iranshahi, M.; Boskabady, M.H. The influence of hydro-ethanolic extract of Portulaca oleracea L. on Th1/Th2 balance in isolated human lymphocytes. J. Ethnopharmacol. 2016, 194, 1112–1121. [Google Scholar] [CrossRef]
  28. Iranshahy, M.; Javadi, B.; Iranshahi, M.; Jahanbakhsh, S.P.; Mahyari, S.; Hassani, F.V.; Karimi, G. A review of traditional uses, phytochemistry and pharmacology of Portulaca oleracea L. J. Ethnopharmacol. 2017, 205, 158–172. [Google Scholar] [CrossRef]
  29. Shakeri, F.; Boskabady, M.H. A review of the relaxant effect of various medicinal plants on tracheal smooth muscle, their possible mechanism(s) and potency. J. Ethnopharmacol. 2015, 175, 528–548. [Google Scholar] [CrossRef] [PubMed]
  30. Chan, K.; Islam, M.W.; Kamil, M.; Radhakrishnan, R.; Zakaria, M.N.; Habibullah, M.; Attas, A. The analgesic and anti-inflammatory effects of Portulaca oleracea L. subsp. sativa (Haw.) Celak. J. Ethnopharmacol. 2000, 73, 445–451. [Google Scholar] [CrossRef] [PubMed]
  31. Bai, Y.; Zang, X.; Ma, J.; Xu, G. Anti-Diabetic Effect of Portulaca oleracea L. Polysaccharide and its Mechanism in Diabetic Rats. Int. J. Mol. Sci. 2016, 17, 1201. [Google Scholar] [CrossRef] [PubMed]
  32. Martinou, E.; Pericleous, M.; Stefanova, I.; Kaur, V.; Angelidi, A.M. Diagnostic Modalities of Non-Alcoholic Fatty Liver Disease: From Biochemical Biomarkers to Multi-Omics Non-Invasive Approaches. Diagnostics 2022, 12, 407. [Google Scholar] [CrossRef]
  33. Riazi, K.; Azhari, H.; Charette, J.H.; Underwood, F.E.; King, J.A.; Afshar, E.E.; Swain, M.G.; Congly, S.E.; Kaplan, G.G.; Shaheen, A.A. The prevalence and incidence of NAFLD worldwide: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2022, 7, 851–861. [Google Scholar] [CrossRef]
  34. Du, H.; Zhou, Y.; Wang, J.; Bai, X.; Tao, B.; Chen, M. High-fat Fructose diet induces neuroinflammation and anxiety-like behaviors by modulating liver–brain axis communication. Psychopharmacology 2025, 1–14. [Google Scholar] [CrossRef]
  35. Kim, D.-G.; Krenz, A.; Toussaint, L.E.; Maurer, K.J.; Robinson, S.-A.; Yan, A.; Torres, L.; Bynoe, M.S. Non-alcoholic fatty liver disease induces signs of Alzheimer’s disease (AD) in wild-type mice and accelerates pathological signs of AD in an AD model. J. Neuroinflamm. 2016, 13, 1. [Google Scholar] [CrossRef] [PubMed]
  36. Kaya, E.; Yılmaz, Y. Association of Metabolic Dysfunction-Associated Fatty Liver Disease with Cognitive Impairment and All-Cause Dementia: A Comprehensive Review. Turk. J. Gastroenterol. 2024, 35, 76–82. [Google Scholar] [CrossRef]
  37. Haley, A.P.; Knight-Scott, J.; Caillaud, M.; Gallagher, I.; Park, J.; Li, Y.; Wang, T.; Tanaka, H.; Browning, J.D. Low carbohydrate and low-calorie diets reduce liver fat and lower brain glutamate and myo-inositol levels in patients with Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Metab. Brain Dis. 2025, 40, 199. [Google Scholar] [CrossRef]
  38. Lau, J.K.; Zhang, X.; Yu, J. Animal models of non-alcoholic fatty liver disease: Current perspectives and recent advances. J. Pathol. 2017, 241, 36–44. [Google Scholar] [CrossRef]
  39. Soret, P.-A.; Magusto, J.; Housset, C.; Gautheron, J. In Vitro and In Vivo Models of Non-Alcoholic Fatty Liver Disease: A Critical Appraisal. J. Clin. Med. 2020, 10, 36. [Google Scholar] [CrossRef] [PubMed]
  40. Yamada, T.; Obata, A.; Kashiwagi, Y.; Rokugawa, T.; Matsushima, S.; Hamada, T.; Watabe, H.; Abe, K. Gd–EOB–DTPA-enhanced–MR imaging in the inflammation stage of nonalcoholic steatohepatitis (NASH) in mice. Magn. Reson. Imaging 2016, 34, 724–729. [Google Scholar] [CrossRef]
  41. Marcolin, E.; Forgiarini, L.F.; Tieppo, J.; Dias, A.S.; Freitas, L.A.R.; Marroni, N.P. Methionine- and choline-deficient diet induces hepatic changes characteristic of non-alcoholic steatohepatitis. Arq. Gastroenterol. 2011, 48, 72–79. [Google Scholar] [CrossRef] [PubMed]
  42. Kim, J.W.; Hahn, K.R.; Yoo, D.Y.; Jung, H.Y.; Hwang, I.K.; Seong, J.K.; Yoon, Y.S. Methionine-Choline Deprivation Impairs Adult Hippocampal Neurogenesis in C57BL/6 Mice. J. Med. Food. 2019, 22, 344–354. [Google Scholar] [CrossRef]
  43. Nedelea, G.; Muşat, M.I.; Mitran, S.I.; Ciorbagiu, M.C.; Cătălin, B. Morphological Differences in Hippocampal Microglia in C57BL/6N Mice with Liver Injury and Depressive-Like Behavior. Curr. Health Sci. J. 2024, 50, 577–584. [Google Scholar] [CrossRef]
  44. Nedelea, G.; Muşat, M.I.; Buican-Chirea, A.C.; Ciorbagiu, M.C.; Cătălin, B. Depressive-Like Behavior and Liver Damage Generate Behavioral and Cortical Microglial Morphological Differences in Mice. Curr. Health Sci. J. 2024, 50, 546–555. [Google Scholar] [CrossRef]
  45. Nedelea, G.; Muşat, M.I.; Mitran, S.I.; Ciorbagiu, M.C.; Cătălin, B. Acute liver damage generates age independent microglia morphology changes in mice. Rom. J. Morphol. Embryol. 2024, 65, 679–685. [Google Scholar] [CrossRef] [PubMed]
  46. Cho, I.Y.; Chang, Y.; Sung, E.; Kang, J.-H.; Wild, S.H.; Byrne, C.D.; Shin, H.; Ryu, S. Depression and increased risk of non-alcoholic fatty liver disease in individuals with obesity. Epidemiol. Psychiatr. Sci. 2021, 30, e23. [Google Scholar] [CrossRef]
  47. Moretti, R.; Caruso, P.; Gazzin, S. Non-alcoholic fatty liver disease and neurological defects. Ann. Hepatol. 2019, 18, 563–570. [Google Scholar] [CrossRef] [PubMed]
  48. Zhou, Y.-X.; Xin, H.-L.; Rahman, K.; Wang, S.-J.; Peng, C.; Zhang, H. Portulaca oleracea L.: A review of phytochemistry and pharmacological effects. Biomed. Res. Int. 2015, 2015, 925631. [Google Scholar] [CrossRef]
  49. Zhao, C.-Q.; Zhou, Y.; Ping, J.; Xu, L.-M. Traditional Chinese medicine for treatment of liver diseases: Progress, challenges and opportunities. J. Integr. Med. 2014, 12, 401–408. [Google Scholar] [CrossRef]
  50. Liu, X.-Y.; Shen, Z.-Y.; Wu, B.; Xia, S.-J.; Huang, J.-H.; Chen, W.-H.; Ying, J. Study on changes of protein phosphorylation of p65, IκBα and IκBε in lymphocytes of rats in progress of aging and interventional effect of Epimedium flavonoids. Zhongguo Zhong Yao Za Zhi 2008, 33, 73–76. [Google Scholar]
  51. El-Sayed, M.I. Effects of Portulaca oleracea L. seeds in treatment of type-2 diabetes mellitus patients as adjunctive and alternative therapy. J. Ethnopharmacol. 2011, 137, 643–651. [Google Scholar] [CrossRef]
  52. Muşat, M.I.; Ifrim-Predoi, A.-M.; Mitran, S.I.; Osiac, E.; Cătălin, B. The Behavioral and Neuroinflammatory Impact of Ketamine in a Murine Model of Depression and Liver Damage. Int. J. Mol. Sci. 2025, 26, 3558. [Google Scholar] [CrossRef] [PubMed]
  53. Morega, S.; Gresita, A.; Mitran, S.I.; Musat, M.I.; Boboc, I.K.S.; Gheorman, V.; Udristoiu, I.; Albu, C.V.; Streba, C.T.; Catalin, B.; et al. Cerebrolysin Use in Patients with Liver Damage—A Translational Study. Life 2022, 12, 1791. [Google Scholar] [CrossRef]
  54. Morega, S.; Cătălin, B.; Simionescu, C.E.; Sapalidis, K.; Rogoveanu, I. Cerebrolysin Prevents Brain Injury in a Mouse Model of Liver Damage. Brain Sci. 2021, 11, 1622. [Google Scholar] [CrossRef]
  55. Rinella, M.E.; Green, R.M. The methionine-choline deficient dietary model of steatohepatitis does not exhibit insulin resistance. J. Hepatol. 2004, 40, 47–51. [Google Scholar] [CrossRef] [PubMed]
  56. Hansen, H.H.; Feigh, M.; Veidal, S.S.; Rigbolt, K.T.; Vrang, N.; Fosgerau, K. Mouse models of nonalcoholic steatohepatitis in preclinical drug development. Drug Discov. Today 2017, 22, 1707–1718. [Google Scholar] [CrossRef]
  57. Rinella, M.E.; Elias, M.S.; Smolak, R.R.; Fu, T.; Borensztajn, J.; Green, R.M. Mechanisms of hepatic steatosis in mice fed a lipogenic methionine choline-deficient diet. J. Lipid Res. 2008, 49, 1068–1076. [Google Scholar] [CrossRef]
  58. Sims, F.H. Methionine and choline deficiency in the rat with special reference to the pregnant state. Br. J. Exp. Pathol. 1951, 32, 481–492. [Google Scholar]
  59. Rizki, G.; Arnaboldi, L.; Gabrielli, B.; Yan, J.; Lee, G.S.; Ng, R.K.; Turner, S.M.; Badger, T.M.; Pitas, R.E.; Maher, J.J. Mice fed a lipogenic methionine-choline-deficient diet develop hypermetabolism coincident with hepatic suppression of SCD-1. J. Lipid Res. 2006, 47, 2280–2290. [Google Scholar] [CrossRef]
  60. Ning, K.; Lu, K.; Chen, Q.; Guo, Z.; Du, X.; Riaz, F.; Feng, L.; Fu, Y.; Yin, C.; Zhang, F.; et al. Epigallocatechin Gallate Protects Mice against Methionine-Choline-Deficient-Diet-Induced Nonalcoholic Steatohepatitis by Improving Gut Microbiota to Attenuate Hepatic Injury and Regulate Metabolism. ACS Omega 2020, 5, 20800–20809. [Google Scholar] [CrossRef] [PubMed]
  61. Zhu, B.; Li, H.; Lu, B.; Guo, X.; Wu, C.; Wang, F.; Li, Q.; Xie, L.; Glaser, S.; Francis, H.; et al. Indole supplementation ameliorates MCD-induced NASH in mice. J. Nutr. Biochem. 2022, 107, 109041, Erratum in: J. Nutr. Biochem. 2024, 133, 109705. https://doi.org/10.1016/j.jnutbio.2024.109705. [Google Scholar] [CrossRef]
  62. Chiba, T.; Suzuki, S.; Sato, Y.; Itoh, T.; Umegaki, K. Evaluation of Methionine Content in a High-Fat and Choline-Deficient Diet on Body Weight Gain and the Development of Non-Alcoholic Steatohepatitis in Mice. PLoS ONE 2016, 11, e0164191. [Google Scholar] [CrossRef]
  63. Matsumoto, M.; Hada, N.; Sakamaki, Y.; Uno, A.; Shiga, T.; Tanaka, C.; Ito, T.; Katsume, A.; Sudoh, M. An improved mouse model that rapidly develops fibrosis in non-alcoholic steatohepatitis. Int. J. Exp. Pathol. 2013, 94, 93–103. [Google Scholar] [CrossRef] [PubMed]
  64. Brown-Borg, H.M. Reduced growth hormone signaling and methionine restriction: Interventions that improve metabolic health and extend life span. Ann. N. Y. Acad. Sci. 2016, 1363, 40–49. [Google Scholar] [CrossRef] [PubMed]
  65. Fan, J.-G.; Kim, S.-U.; Wong, V.W.-S. New trends on obesity and NAFLD in Asia. J. Hepatol. 2017, 67, 862–873. [Google Scholar] [CrossRef] [PubMed]
  66. Mundi, M.S.; Velapati, S.; Patel, J.; Kellogg, T.A.; Abu Dayyeh, B.K.; Hurt, R.T. Evolution of NAFLD and Its Management. Nutr. Clin. Pract. 2020, 35, 72–84. [Google Scholar] [CrossRef]
  67. Aleksandrova, K.; Stelmach-Mardas, M.; Schlesinger, S. Obesity and Liver Cancer. Recent Results Cancer Res. 2016, 208, 177–198. [Google Scholar] [CrossRef]
  68. Zhu, L.-L.; Shen, J.-D.; Wang, B.-Y.; Lu, S.-F.; Ming, B.; Xu, E.-P.; Li, Y.-C.; Fang, X.-Y. Depressive-like behaviors are induced by chronic liver injury in male and female mice. Neurosci. Lett. 2020, 718, 134750. [Google Scholar] [CrossRef]
  69. Romeo, R.D.; Mueller, A.; Sisti, H.M.; Ogawa, S.; McEwen, B.S.; Brake, W.G. Anxiety and fear behaviors in adult male and female C57BL/6 mice are modulated by maternal separation. Horm. Behav. 2003, 43, 561–567. [Google Scholar] [CrossRef]
  70. Maguire, J.L.; Stell, B.M.; Rafizadeh, M.; Mody, I. Ovarian cycle-linked changes in GABAA receptors mediating tonic inhibition alter seizure susceptibility and anxiety. Nat. Neurosci. 2005, 8, 797–804. [Google Scholar] [CrossRef]
  71. Shansky, R.M. Are hormones a “female problem” for animal research? Science 2019, 364, 825–826. [Google Scholar] [CrossRef]
  72. Clayton, J.A.; Collins, F.S. Policy: NIH to balance sex in cell and animal studies. Nature 2014, 509, 282–283. [Google Scholar] [CrossRef]
  73. Beery, A.K. Inclusion of females does not increase variability in rodent research studies. Curr. Opin. Behav. Sci. 2018, 23, 143–149. [Google Scholar] [CrossRef] [PubMed]
  74. Meziane, H.; Ouagazzal, A.-M.; Aubert, L.; Wietrzych, M.; Krezel, W. Estrous cycle effects on behavior of C57BL/6J and BALB/cByJ female mice: Implications for phenotyping strategies. Genes Brain Behav. 2007, 6, 192–200. [Google Scholar] [CrossRef] [PubMed]
  75. Nisbett, K.E.; Gonzalez, L.A.; Teruel, M.; Carter, C.S.; Vendruscolo, L.F.; Ragozzino, M.E.; Koob, G.F. Sex and hormonal status influence the anxiolytic-like effect of oxytocin in mice. Neurobiol. Stress 2023, 26, 100567. [Google Scholar] [CrossRef] [PubMed]
  76. Tsao, C.-H.; Wu, K.-Y.; Su, N.C.; Edwards, A.; Huang, G.-J. The influence of sex difference on behavior and adult hippocampal neurogenesis in C57BL/6 mice. Sci. Rep. 2023, 13, 17297. [Google Scholar] [CrossRef]
  77. Zeng, P.-Y.; Tsai, Y.-H.; Lee, C.-L.; Ma, Y.-K.; Kuo, T.-H. Minimal influence of estrous cycle on studies of female mouse behaviors. Front. Mol. Neurosci. 2023, 16, 1146109. [Google Scholar] [CrossRef]
  78. Nguyen, H.H.; Swain, M.G. Avenues within the gut–liver–brain axis linking chronic liver disease and symptoms. Front. Neurosci. 2023, 17, 1171253. [Google Scholar] [CrossRef]
  79. Yan, M.; Man, S.; Sun, B.; Ma, L.; Guo, L.; Huang, L.; Gao, W. Gut liver brain axis in diseases: The implications for therapeutic interventions. Signal Transduct. Target Ther. 2023, 8, 443. [Google Scholar] [CrossRef]
  80. De Cól, J.P.; de Lima, E.P.; Pompeu, F.M.; Cressoni Araújo, A.; de Alvares Goulart, R.; Bechara, M.D.; Laurindo, L.F.; Méndez-Sánchez, N.; Barbalho, S.M. Underlying Mechanisms behind the Brain–Gut–Liver Axis and Metabolic-Associated Fatty Liver Disease (MAFLD): An Update. Int. J. Mol. Sci. 2024, 25, 3694. [Google Scholar] [CrossRef]
  81. Bilen, M.; Ibrahim, P.; Barmo, N.; Abou Haidar, E.; Karnib, N.; El Hayek, L.; Khalifeh, M.; Jabre, V.; Houbeika, R.; Stephan, J.S.; et al. Methionine mediates resilience to chronic social defeat stress by epigenetic regulation of NMDA receptor subunit expression. Psychopharmacology 2020, 237, 3007–3020. [Google Scholar] [CrossRef]
  82. Sarkisova, K.Y.; Gabova, A.V.; Fedosova, E.A.; Shatskova, A.B.; Narkevich, V.B.; Kudrin, V.S. Antidepressant and Anxiolytic Effects of L-Methionine in the WAG/Rij Rat Model of Depression Comorbid with Absence Epilepsy. Int. J. Mol. Sci. 2023, 24, 12425. [Google Scholar] [CrossRef]
  83. Li, J.; Kang, X.; Zhang, L.; Luo, J.; Zhang, D. Dietary choline is inversely associated with depressive symptoms: A cross-sectional study of the National Health and Nutrition Examination Survey (NHANES) 2011 to 2018. J. Affect. Disord. 2022, 301, 23–29. [Google Scholar] [CrossRef]
  84. Kazeminejad, S.; Moradmand, Z.; Shahdadian, F.; Hajhashemy, Z.; Rouhani, P.; Saneei, P. Dietary choline and betaine intake in relation to psychological disorders in adults. Br. J. Nutr. 2025, 133, 1431–1438. [Google Scholar] [CrossRef]
  85. Bekdash, R.A. Neuroprotective Effects of Choline and Other Methyl Donors. Nutrients 2019, 11, 2995. [Google Scholar] [CrossRef] [PubMed]
  86. Li, Q.; Zhao, B.; Li, W.; He, Y.; Tang, X.; Zhang, T.; Zhong, Z.; Pan, Q.; Zhang, Y. Effects of repeated drug administration on behaviors in normal mice and fluoxetine efficacy in chronic unpredictable mild stress mice. Biochem. Biophys. Res. Commun. 2022, 615, 36–42. [Google Scholar] [CrossRef]
  87. Radhakrishnan, R.; Zakaria, M.N.; Islam, M.W.; Chen, H.B.; Kamil, M.; Chan, K.; Al-Attas, A. Neuropharmacological actions of Portulaca oleraceae L v. sativa (Hawk). J. Ethnopharmacol. 2001, 76, 171–176. [Google Scholar] [CrossRef]
  88. Parry, O.; Okwuasaba, F.K.; Ejike, C. Skeletal muscle relaxant action of an aqueous extract of Portulaca oleracea in the rat. J. Ethnopharmacol. 1987, 19, 247–253. [Google Scholar] [CrossRef] [PubMed]
  89. Forouzanfar, F.; Hosseinzadeh, H.; Khorrami, M.B.; Asgharzade, S.; Rakhshandeh, H. Attenuating Effect of Portulaca oleracea Extract on Chronic Constriction Injury Induced Neuropathic Pain in Rats: An Evidence of Anti-oxidative and Anti-inflammatory Effects. CNS Neurol. Disord. Drug Targets 2019, 18, 342–349. [Google Scholar] [CrossRef] [PubMed]
  90. Rahimi, V.B.; Ajam, F.; Rakhshandeh, H.; Askari, V.R. A Pharmacological Review on Portulaca oleracea L.: Focusing on Anti-Inflammatory, Anti-Oxidant, Immuno-Modulatory and Antitumor Activities. J. Pharmacopunct. 2019, 22, 7–15. [Google Scholar] [CrossRef]
  91. Abdel Moneim, A.E. The neuroprotective effects of purslane (Portulaca oleracea) on rotenone-induced biochemical changes and apoptosis in brain of rat. CNS Neurol. Disord. Drug Targets 2013, 12, 830–841. [Google Scholar] [CrossRef] [PubMed]
  92. Wanyin, W.; Liwei, D.; Lin, J.; Hailiang, X.; Changquan, L.; Min, L. Ethanol extract of Portulaca oleracea L. protects against hypoxia-induced neuro damage through modulating endogenous erythropoietin expression. J. Nutr. Biochem. 2012, 23, 385–391. [Google Scholar] [CrossRef]
  93. Wang, C.Q.; Yang, G.Q. Betacyanins from Portulaca oleracea L. ameliorate cognition deficits and attenuate oxidative damage induced by D-galactose in the brains of senescent mice. Phytomedicine 2010, 17, 527–532. [Google Scholar] [CrossRef]
  94. Yang, Z.; Zhang, D.; Ren, J.; Yang, M.; Li, S. Acetylcholinesterase inhibitory activity of the total alkaloid from traditional Chinese herbal medicine for treating Alzheimer’s disease. Med. Chem. Res. 2012, 21, 734–738. [Google Scholar] [CrossRef]
  95. Groblewski, P.A.; Bax, L.S.; Cunningham, C.L. Reference-dose place conditioning with ethanol in mice: Empirical and theoretical analysis. Psychopharmacology 2008, 201, 97–106. [Google Scholar] [CrossRef]
  96. Li, X.; Cheng, X.; Wang, X.; Liu, Q.; Ma, H.; Li, M. Dyslipidemic Diet Induces Mobilization of Peripheral Neutrophils and Monocytes That Exacerbate Hemorrhagic Brain Injury and Neuroinflammation. Front. Cell. Neurosci. 2020, 14, 154. [Google Scholar] [CrossRef]
  97. Brockhurst, J.; Cheleuitte-Nieves, C.; Buckmaster, C.L.; Schatzberg, A.F.; Lyons, D.M. Stress inoculation modeled in mice. Transl. Psychiatry 2015, 5, e537. [Google Scholar] [CrossRef]
  98. Matthews, D.R.; Li, H.; Zhou, J.; Li, Q.; Glaser, S.; Francis, H.; Alpini, G.; Wu, C. Methionine- and Choline-Deficient Diet-Induced Nonalcoholic Steatohepatitis Is Associated with Increased Intestinal Inflammation. Am. J. Pathol. 2021, 191, 1743–1753. [Google Scholar] [CrossRef]
  99. Gautam, J.; Aggarwal, H.; Kumari, D.; Gupta, S.K.; Kumar, Y.; Dikshit, M. A methionine-choline-deficient diet induces nonalcoholic steatohepatitis and alters the lipidome, metabolome, and gut microbiome profile in the C57BL/6J mouse. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2024, 1869, 159545. [Google Scholar] [CrossRef]
  100. Sun, X.; Li, Z.; Chen, Y.; Xu, T.; Shu, J.; Shi, L.; Shi, Z. Interactive Effects of Methionine and Lead Intake on Cognitive Function among Chinese Adults. Nutrients 2022, 14, 4561. [Google Scholar] [CrossRef]
  101. Lail, H.; Mabb, A.M.; Parent, M.B.; Pinheiro, F.; Wanders, D. Effects of Dietary Methionine Restriction on Cognition in Mice. Nutrients 2023, 15, 4950. [Google Scholar] [CrossRef]
  102. Xi, Y.; Zhang, Y.; Zhou, Y.; Liu, Q.; Chen, X.; Liu, X.; Grune, T.; Shi, L.; Hou, M.; Liu, Z. Effects of methionine intake on cognitive function in mild cognitive impairment patients and APP/PS1 Alzheimer’s Disease model mice: Role of the cystathionine-β-synthase/H2S pathway. Redox Biol. 2023, 59, 102595. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, X.; Zhao, Y.; Liu, J.; Zhang, H.; Yao, J.; Liu, J.; Niu, Y.; Ying, X. The anti-inflammatory mechanism of a new skeleton alkaloid isolated from Portulaca oleracea L. Fitoterapia 2025, 185, 106760. [Google Scholar] [CrossRef]
  104. Zhang, X.; Jiu, J.; Wei, W.; Yao, J.; He, F.; Ying, X. A new alkaloid and five known compounds from Portulaca oleracea L. and their anti-inflammatory activities. Nat. Prod. Res. 2025, 29, 1–9. [Google Scholar] [CrossRef]
  105. Wang, W.; Gu, L.; Dong, L.; Wang, X.; Ling, C.; Li, M. Protective effect of Portulaca oleracea extracts on hypoxic nerve tissue and its mechanism. Asia Pac. J. Clin. Nutr. 2007, 16, 227–233. [Google Scholar]
  106. Martins, W.B.; Rodrigues, S.A.; Silva, H.K.; Dantas, C.G.; Júnior, W.L.; Filho, L.X.; Cardoso, J.C.; Gomes, M.Z. Neuroprotective effect of Portulaca oleracea extracts against 6-hydroxydopamine-induced lesion of dopaminergic neurons. An. Acad. Bras. Cienc. 2016, 88, 1439–1450. [Google Scholar] [CrossRef]
  107. Itagaki, H.; Shimizu, K.; Morikawa, S.; Ogawa, K.; Ezaki, T. Morphological and functional characterization of non-alcoholic fatty liver disease induced by a methionine-choline-deficient diet in C57BL/6 mice. Int. J. Clin. Exp. Pathol. 2013, 6, 2683–2696. [Google Scholar]
  108. Wang, X.-D.; Yang, G.; Bai, Y.; Feng, Y.-P.; Li, H. The behavioral study on the interactive aggravation between pruritus and depression. Brain Behav. 2018, 8, e00964. [Google Scholar] [CrossRef]
  109. Pałucha-Poniewiera, A.; Podkowa, K.; Rafało-Ulińska, A. The group II mGlu receptor antagonist LY341495 induces a rapid antidepressant-like effect and enhances the effect of ketamine in the chronic unpredictable mild stress model of depression in C57BL/6J mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 109, 110239. [Google Scholar] [CrossRef]
  110. Herselman, M.F.; Bobrovskaya, L. The Effects of Chronic Unpredictable Mild Stress and Semi-Pure Diets on the Brain, Gut and Adrenal Medulla in C57BL6 Mice. Int. J. Mol. Sci. 2023, 24, 14618. [Google Scholar] [CrossRef]
  111. Li, Z.; Wang, Q.; Zhang, Z.; Guo, Y.; Sun, M.; Li, L.; He, W. Chrysin alleviated CUMS-induced depressive-like behaviors in mice via directly targeting Fyn. J. Funct. Foods 2023, 106, 105603. [Google Scholar] [CrossRef]
  112. Custodio-Mendoza, J.A.; Pokorski, P.; Aktaş, H.; Kurek, M.A. Rapid and efficient high-performance liquid chromatography-ultraviolet determination of total amino acids in protein isolates by ultrasound-assisted acid hydrolysis. Ultrason. Sonochem. 2024, 111, 107082. [Google Scholar] [CrossRef]
  113. Thackaberry, E.A.; Wang, X.; Schweiger, M.; Messick, K.; Valle, N.; Dean, B.; Sambrone, A.; Bowman, T.; Xie, M. Solvent-based formulations for intravenous mouse pharmacokinetic studies: Tolerability and recommended solvent dose limits. Xenobiotica 2014, 44, 235–241. [Google Scholar] [CrossRef]
  114. Jalali, J.; Ghasemzadeh Rahbardar, M. Ameliorative effects of Portulaca oleracea L. (purslane) and its active constituents on nervous system disorders: A review. Iran. J. Basic Med. Sci. 2023, 26, 2–12. [Google Scholar] [CrossRef]
  115. Hamedi, S.; Forouzanfar, F.; Rakhshandeh, H.; Arian, A. Hypnotic Effect of Portulaca oleracea on Pentobarbital-Induced Sleep in Mice. Curr. Drug Discov. Technol. 2019, 16, 198–203. [Google Scholar] [CrossRef]
  116. Muşat, M.I.; Cătălin, B.; Hadjiargyrou, M.; Popa-Wagner, A.; Greşiţă, A. Advancing Post-Stroke Depression Research: Insights from Murine Models and Behavioral Analyses. Life 2024, 14, 1110. [Google Scholar] [CrossRef]
  117. Boboc, I.K.S.; Cojocaru, A.; Nedelea, G.; Catalin, B.; Bogdan, M.; Calina, D. Chronic Administration of Ion Channel Blockers Impact Microglia Morphology and Function in a Murine Model of Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 14474. [Google Scholar] [CrossRef]
  118. Cătălin, B.; Stopper, L.; Bălşeanu, T.-A.; Scheller, A. The in situ morphology of microglia is highly sensitive to the mode of tissue fixation. J. Chem. Neuroanat. 2017, 86, 59–66. [Google Scholar] [CrossRef]
  119. Godeanu, S.; Muşat, M.I.; Scheller, A.; Osiac, E.; Cătălin, B. Minimal differences observed when comparing the morphological profiling of microglia obtained by confocal laser scanning and optical sectioning microscopy. Front. Neuroanat. 2025, 18, 1507140. [Google Scholar] [CrossRef]
  120. Godeanu, S.; Clarke, D.; Stopper, L.; Deftu, A.-F.; Popa-Wagner, A.; Bălşeanu, A.T.; Scheller, A.; Catalin, B. Microglial morphology in the somatosensory cortex across lifespan. A quantitative study. Dev. Dyn. 2023, 252, 1113–1129. [Google Scholar] [CrossRef]
Figure 1. (a) Body weight evolution during the experiment. Anhedonia-like behavior assessed by Sucrose Preference Test in mice which were fed (b) MCD or (c) MC diet. (d) Diet-dependent comparison of percent change in sucrose preference. The plots show mean values ± SD, * p < 0.05, ** p < 0.01. CUMS: Chronic unpredictable mild stress; MC: Methionine- and choline-controlled diet; MCD: Methionine- and choline-deficient diet; PO: Portulaca oleracea; SD: Standard deviation; VEH: Vehicle.
Figure 1. (a) Body weight evolution during the experiment. Anhedonia-like behavior assessed by Sucrose Preference Test in mice which were fed (b) MCD or (c) MC diet. (d) Diet-dependent comparison of percent change in sucrose preference. The plots show mean values ± SD, * p < 0.05, ** p < 0.01. CUMS: Chronic unpredictable mild stress; MC: Methionine- and choline-controlled diet; MCD: Methionine- and choline-deficient diet; PO: Portulaca oleracea; SD: Standard deviation; VEH: Vehicle.
Ijms 26 10050 g001
Figure 2. Anxiety-like behavior using the open field test in mice which were fed either (a) an MCD or (b) an MC diet. (c) Representative images depicting trajectories of animals within the testing arena at baseline and after treatment. (d) Diet-dependent comparison of percent changes in anxiety-like behavior. The plots show mean values ± SD, * p < 0.05, ** p < 0.01, and **** p < 0.0001.
Figure 2. Anxiety-like behavior using the open field test in mice which were fed either (a) an MCD or (b) an MC diet. (c) Representative images depicting trajectories of animals within the testing arena at baseline and after treatment. (d) Diet-dependent comparison of percent changes in anxiety-like behavior. The plots show mean values ± SD, * p < 0.05, ** p < 0.01, and **** p < 0.0001.
Ijms 26 10050 g002
Figure 3. Memory performance assessed using the Novel Object Preference test in mice which were fed either (a) MCD or (b) MC diet. (c) Representative images depicting exploration trajectories within the testing arena at baseline and following treatment. (d) Diet-dependent analysis of percent changes in novel object preference. The plots show mean values ± SD, * p < 0.05, ** p < 0.01.
Figure 3. Memory performance assessed using the Novel Object Preference test in mice which were fed either (a) MCD or (b) MC diet. (c) Representative images depicting exploration trajectories within the testing arena at baseline and following treatment. (d) Diet-dependent analysis of percent changes in novel object preference. The plots show mean values ± SD, * p < 0.05, ** p < 0.01.
Ijms 26 10050 g003
Figure 4. Quantification of Iba1-positive microglia in (a) the cortex, and in (c) the hippocampus from mice included in the study groups. Representative microscopic images for the Iba1 positivity in the cortex (b) and in the hippocampus (d). Microglia were labeled with Iba1 (green) and cell nuclei were labeled with DAPI (blue). The graphs show mean values ± SD, * p < 0.05, ** p < 0.01. Scale bars 50 μm. DAPI: 4′,6-Diamidino-2-phenylindole; Iba1: Ionized calcium binding adaptor molecule 1.
Figure 4. Quantification of Iba1-positive microglia in (a) the cortex, and in (c) the hippocampus from mice included in the study groups. Representative microscopic images for the Iba1 positivity in the cortex (b) and in the hippocampus (d). Microglia were labeled with Iba1 (green) and cell nuclei were labeled with DAPI (blue). The graphs show mean values ± SD, * p < 0.05, ** p < 0.01. Scale bars 50 μm. DAPI: 4′,6-Diamidino-2-phenylindole; Iba1: Ionized calcium binding adaptor molecule 1.
Ijms 26 10050 g004
Figure 5. Comparison of GFAP-positive signal in (a) the cortex and in (c) the hippocampus of mice included in the study groups. Representative microscopic images depicting GFAP positivity in the cortex (b) and in the hippocampus (d). Astrocytes were labeled with GFAP (yellow), and cell nuclei with DAPI (blue). The plots show mean values ± SD, * p < 0.05, ** p < 0.01. Scale bars 50 μm. GFAP: Glial fibrillary acidic protein.
Figure 5. Comparison of GFAP-positive signal in (a) the cortex and in (c) the hippocampus of mice included in the study groups. Representative microscopic images depicting GFAP positivity in the cortex (b) and in the hippocampus (d). Astrocytes were labeled with GFAP (yellow), and cell nuclei with DAPI (blue). The plots show mean values ± SD, * p < 0.05, ** p < 0.01. Scale bars 50 μm. GFAP: Glial fibrillary acidic protein.
Ijms 26 10050 g005
Figure 6. Comparison of (a) cortical and (c) hippocampal neurogenesis by quantitative morphometry of immunofluorescent stains for NeuN in mouse brains. Representative microscopic images depicting NeuN positivity in the cortex (b) and in the hippocampus (d). Neurons were labeled with NeuN (red), and cell nuclei were labeled with DAPI (blue). The plots show mean values ± SD, * p < 0.05, ** p < 0.01. Scale bars 50 μm. NeuN: Neuronal nuclei.
Figure 6. Comparison of (a) cortical and (c) hippocampal neurogenesis by quantitative morphometry of immunofluorescent stains for NeuN in mouse brains. Representative microscopic images depicting NeuN positivity in the cortex (b) and in the hippocampus (d). Neurons were labeled with NeuN (red), and cell nuclei were labeled with DAPI (blue). The plots show mean values ± SD, * p < 0.05, ** p < 0.01. Scale bars 50 μm. NeuN: Neuronal nuclei.
Ijms 26 10050 g006
Figure 7. Experimental design. The diagram shows the stages of the study and their sequence. D: Day.
Figure 7. Experimental design. The diagram shows the stages of the study and their sequence. D: Day.
Ijms 26 10050 g007
Figure 8. Portulaca oleracea extract preparation. (a) Representative UHPLC–PDA chromatogram (detection at 265 nm) showing the separation of FMOC-derivatized methionine standard. The peak corresponding to unreacted FMOC reagent (or its hydrolysis product) is observed at a tR of 5.512 min, while the peak for FMOC–methionine elutes at tR 7.127 min. (b) Positive ion electrospray mass spectrum obtained for the peak identified as FMOC–methionine in (a). The spectrum confirms the identity of the derivatized complex, showing the characteristic protonated molecular ion [M + H]+ for FMOC–methionine at m/z 372. FMOC: Fluorenylmethyl chloroformate; tR: Retention time; UHPLC–PDA: Ultra-high-performance liquid chromatography–Photodiode array.
Figure 8. Portulaca oleracea extract preparation. (a) Representative UHPLC–PDA chromatogram (detection at 265 nm) showing the separation of FMOC-derivatized methionine standard. The peak corresponding to unreacted FMOC reagent (or its hydrolysis product) is observed at a tR of 5.512 min, while the peak for FMOC–methionine elutes at tR 7.127 min. (b) Positive ion electrospray mass spectrum obtained for the peak identified as FMOC–methionine in (a). The spectrum confirms the identity of the derivatized complex, showing the characteristic protonated molecular ion [M + H]+ for FMOC–methionine at m/z 372. FMOC: Fluorenylmethyl chloroformate; tR: Retention time; UHPLC–PDA: Ultra-high-performance liquid chromatography–Photodiode array.
Ijms 26 10050 g008
Table 1. Experimental groups and number of animals used in the study.
Table 1. Experimental groups and number of animals used in the study.
GroupNo. of Animals
SHAM4
MCD4
MC4
MCD + CUMS4
MC + CUMS4
MCD + VEH4
MC + VEH4
MCD + PO4
MC + PO4
MCD + CUMS + VEH4
MC + CUMS + VEH4
MCD + CUMS + PO6
MC + CUMS + PO4
CUMS: Chronic unpredictable mild stress; MC: Methionine- and choline-controlled diet; MCD: Methionine- and choline-deficient diet; PO: Portulaca oleracea; VEH: Vehicle.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mitran, S.I.; Muşat, M.I.; Bejenaru, C.; Mogoşanu, G.D.; Boboc, I.K.Ş.; Tudoraşcu, R.-I.; Târtea, G.; Zlătian, O.M.; Blendea, A.; Biţă, A.; et al. Portulaca oleracea Extract Modulates Diet-Dependent Neuroplasticity in a Murine Model of MCD-Induced NAFLD and Depression. Int. J. Mol. Sci. 2025, 26, 10050. https://doi.org/10.3390/ijms262010050

AMA Style

Mitran SI, Muşat MI, Bejenaru C, Mogoşanu GD, Boboc IKŞ, Tudoraşcu R-I, Târtea G, Zlătian OM, Blendea A, Biţă A, et al. Portulaca oleracea Extract Modulates Diet-Dependent Neuroplasticity in a Murine Model of MCD-Induced NAFLD and Depression. International Journal of Molecular Sciences. 2025; 26(20):10050. https://doi.org/10.3390/ijms262010050

Chicago/Turabian Style

Mitran, Smaranda Ioana, Mădălina Iuliana Muşat, Cornelia Bejenaru, George Dan Mogoşanu, Ianis Kevyn Ştefan Boboc, Robertina-Iulia Tudoraşcu, Georgică Târtea, Ovidiu Mircea Zlătian, Antonia Blendea, Andrei Biţă, and et al. 2025. "Portulaca oleracea Extract Modulates Diet-Dependent Neuroplasticity in a Murine Model of MCD-Induced NAFLD and Depression" International Journal of Molecular Sciences 26, no. 20: 10050. https://doi.org/10.3390/ijms262010050

APA Style

Mitran, S. I., Muşat, M. I., Bejenaru, C., Mogoşanu, G. D., Boboc, I. K. Ş., Tudoraşcu, R.-I., Târtea, G., Zlătian, O. M., Blendea, A., Biţă, A., Segneanu, A.-E., & Bejenaru, L. E. (2025). Portulaca oleracea Extract Modulates Diet-Dependent Neuroplasticity in a Murine Model of MCD-Induced NAFLD and Depression. International Journal of Molecular Sciences, 26(20), 10050. https://doi.org/10.3390/ijms262010050

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

Article Metrics

Back to TopTop