Next Article in Journal
Analysis of Intestinal Bacterial Microbiota in Individuals with and without Chronic Low Back Pain
Previous Article in Journal
Lonicera japonica Thunb. Ethanol Extract Exerts a Protective Effect on Normal Human Gastric Epithelial Cells by Modulating the Activity of Tumor-Necrosis-Factor-α-Induced Inflammatory Cyclooxygenase 2/Prostaglandin E2 and Matrix Metalloproteinase 9
Previous Article in Special Issue
A Synopsis of Biomarkers in Glioblastoma: Past and Present
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ghrelin/GHSR System in Depressive Disorder: Pathologic Roles and Therapeutic Implications

1
School of Biological Sciences, Jining Medical University, Jining 272067, China
2
School of Clinical Medicine, Jining Medical University, Jining 272067, China
3
Neurobiology Institute, Jining Medical University, Jining 272067, China
4
Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
5
Institute of Precision Medicine, Jining Medical University, Jining 272067, China
*
Authors to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2024, 46(7), 7324-7338; https://doi.org/10.3390/cimb46070434
Submission received: 29 May 2024 / Revised: 3 July 2024 / Accepted: 8 July 2024 / Published: 10 July 2024
(This article belongs to the Special Issue Molecular Genetics and Genomics in Brain Disorders)

Abstract

:
Depression is the most common chronic mental illness and is characterized by low mood, insomnia, and affective disorders. However, its pathologic mechanisms remain unclear. Numerous studies have suggested that the ghrelin/GHSR system may be involved in the pathophysiologic process of depression. Ghrelin plays a dual role in experimental animals, increasing depressed behavior and decreasing anxiety. By combining several neuropeptides and traditional neurotransmitter systems to construct neural networks, this hormone modifies signals connected to depression. The present review focuses on the role of ghrelin in neuritogenesis, astrocyte protection, inflammatory factor production, and endocrine disruption in depression. Furthermore, ghrelin/GHSR can activate multiple signaling pathways, including cAMP/CREB/BDNF, PI3K/Akt, Jak2/STAT3, and p38-MAPK, to produce antidepressant effects, given which it is expected to become a potential therapeutic target for the treatment of depression.

1. Introduction

Growth-hormone-releasing peptide (ghrelin) is a multifunctional 28-amino-acid peptide hormone which is mainly released from the stomach but is also expressed in a number of other tissues, including the pancreas and kidneys [1]. Ghrelin’s serine 3 (Ser3) is n-octastylated, a modification that allows for the cycling of both the acylated and des-acylated forms of ghrelin [2]. Ghrelin can bind to central growth-hormone-releasing peptide receptor (GHSR), which is widely found in the central nervous system (CNS) and peripheral tissues, and has two isoforms, GHSR1a and GHSR1b [3]. Ghrelin mainly binds to GHSR1a and exerts a variety of physiological and behavioral modulatory effects, such as the regulation of glucose homeostasis, the regulation of metabolism and energy homeostasis, the modulation of blood pressure, and renal protection [4,5,6]. GHSR1b contains 289 amino acids and is a splice variant and a dominantly inactivated form of GHSR1a [7]. Due to the absence of the sixth and seventh transmembrane chains, it is unable to bind ghrelin and does not have signaling ability. There is much evidence that GHSR1b does not bind ghrelin, but it can heterodimerize with GHSR1a to interfere with GHSR1a function [8]. In an experiment to study the expression of the two GHSR forms cloned from black seabream in HEK293 cells, GHSR1b was found to inhibit the signaling activity of the GHSR1a-mediated increase in intracellular Ca2+ concentration [9]. GHSR1a has also been shown to form mixed heterodimers with dopamine receptor subtype 2 (D2R) [10], serotonin 2c receptor (5-HT2cR) [11], and orexin 1 receptor (OX1R) [12], and the dimers can fine-tune its activity.
Depression is a prevalent severe chronic mental illness which is characterized by episodes of depressed mood, appetite disorders, sleep disturbances, bipolar disorder, and loss of interest and pleasure. The above-mentioned disorders may be attributed to decreased size and function of the hippocampus and amygdala in the limbic brain regions [13]. With the rise in survival pressures in contemporary society and the dramatic increase in prevalence and incidence in different parts of the country, depression is emerging as a complex disorder present in all parts of the population, including the pregnant population, the adolescent population, and the elderly population [14,15]. Although the exact pathogenesis of the disorder remains obscure, a large body of evidence strongly suggests that stress and genetic predisposition are collectively involved in the progression of the disorder [16]. The pathophysiologic processes of depression include inflammation in the brain, reduced neurogenesis, monoamine neurotransmitter changes, and endocrine abnormalities [17,18,19]. However, the interrelationships among these processes remain unclear. Currently, several classes of drugs are available for the treatment of major depressive disorder (MDD) [20], and most of them exert their antidepressant effects by enhancing monoaminergic function, such as typical monoamine oxidase inhibitors (MAOIs) and selective 5-hydroxytryptamine reuptake inhibitors (SSRIs) [21]. In addition, scientists are now working on research beyond typical monoamine targets and pathways to develop antidepressants with novel mechanisms of action [22].
However, the efficacy and severe side effects of these antidepressants remain major issues to be addressed [23], and there are no therapeutic candidates shown to completely eliminate disease progression. In line with ghrelin’s effects on the CNS, there is increasing evidence for an important role of ghrelin in depression [24,25,26]. The majority of research has elucidated the anti-depressive mechanisms of ghrelin. Conversely, a few studies have reported a detrimental effect as ghrelin itself induces depressive tendencies and patterns in animal models. Here, we attempted to elucidate the aforementioned link between ghrelin and the onset as well as the treatment of depression and to elucidate the mechanisms and pathways that may be involved in the development of depression. We performed such an explanatory study by conducting a comprehensive review of the existing literature (reviews and research articles) through keyword searches.

2. Mechanisms of Ghrelin/GHSR System in Depressive Disorder

Numerous studies have shown that the ghrelin/GHSR system inhibits key pathways and mechanisms in the development of depression that have been found to correlate with certain factors, transmitters, and cells in the body in a variety of models of depression, which we will describe in more detail in the following paragraphs.

2.1. Links between Monoamine Neurotransmitter Receptors and Ghrelin

The monoamine hypothesis, long considered to be the most common hypothesis for depression pathogenesis, is centered on the fact that the concentration of monoamines (5-hydroxytryptamine, norepinephrine, and dopamine) in the synaptic gap is reduced in depressive states [27]. These monoamine neurotransmitters may influence ghrelin’s neuropathology. It was found that in neurons co-expressing D1R and GHSR, ghrelin can amplify dopamine signaling by activating GHSR to enhance its mediated downstream pathways [28]. Moreover, the mRNA expression of dopaminergic receptors was reduced in the amygdala and dorsal raphe nucleus of GHSR1a−/− mice [29], suggesting the potential regulation of dopamine neuron production by the ghrelin/GHSR system. In addition to this, another study showed that the central 5-hydroxytryptamine system is a target of ghrelin [29], and increased mRNA expression of some 5-hydroxytryptamine receptors in the amygdala and the dorsal nucleus of the intermediate suture was found in mice acutely centrally administered with ghrelin. Conversely, unlike the ghrelin/GHSR system described above, which regulates the expression of monoamine neurotransmitters, norepinephrine has been shown to stimulate ghrelin secretion in mouse cells [30], and the depletion of catecholamine-secreting neurons can also inhibit fasting-induced ghrelin secretion [31]. Moreover, GHSR1a differentially inhibits presynaptic voltage-gated calcium channels (CaV2.1 and CaV2.2) via the Gi/o- or Gq-dependent pathway, which reduce GABA release from hypothalamic neurons [32], contributing to neuronal activation through the disinhibition of postsynaptic neurons. In conclusion, there are many unknown connections between the monoamine system and the ghrelin/GHSR system, and more research is needed to determine whether monoamine receptors can be used as targets in the treatment of depression.

2.2. The Ghrelin/GHSR System Mediates the Inflammatory Response to Depression

In the CNS, IL-1β stimulates microglia and astrocytes to produce other cytokines, such as IL-6 and TNF-α, to promote inflammation in the brain [33]. Furthermore, pro-inflammatory cytokines such as IL-1β and TNF-α have been reported to play an important role in the onset of depression [34], and an increase in pro-inflammatory cytokines contributes to the development of depression [17]. Moreover, IL-6 is the most persistently elevated cytokine in the blood of MDD patients; so, it may serve as a predictive biomarker and a potential target for the treatment of depression in humans [35]. These experimental conclusions show that the role of pro-inflammatory cytokines of the CNS in the pathogenesis of depression cannot be ignored.
It is well-documented that serum ghrelin concentration is positively correlated with inflammation. For instance, serum ghrelin concentrations were higher in rats with severe pancreatitis, arthritis, and acute colitis. Nonetheless, rheumatoid arthritis patients and arthritis rats had lower levels of serum ghrelin concentrations [36]. This discrepancy may be due to unpredictable reasons, such as rat nutritional status or experimental conditions, making it difficult to analyze the effect of inflammation on serum ghrelin levels. However, it is undeniable that ghrelin levels are elevated in inflammation, as demonstrated in most experiments. On the whole, the causal relationship between inflammation and changes in serum ghrelin concentrations requires further study, although the effects of various conditions cannot be ruled out. Pursuant to the existing data, there are many reports indicating that elevated serum ghrelin concentrations are the result of inflammation. Interestingly, experiments have found that elevated ghrelin levels are involved in regulating inflammatory response, downregulating neutrophil transport and the number of pro-inflammatory cytokines, significantly reducing cerebral ischemic injury, and improving neurobehavioral function [37]. The same study also reported that exogenous ghrelin inhibits the endothelial cell production of IL-1, IL-6, and IL-8 by regulating the release of pro-inflammatory cytokines, which play an important role in the pathologic process of depression [38]. In addition, GHSR and ghrelin are expressed in human T lymphocytes and monocytes, and ghrelin specifically inhibits the expression of pro-inflammatory cytokines via GHSR [39]. In addition, two other investigations displayed decreased expression of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL6 in rats with GHSR gene knock-out [40]. Thus, there is increasing evidence of the mediating role of the ghrelin/GHSR system in regulating pro-inflammatory cytokine release events in depressive disorders.
On the other hand, we can note that inflammation reduces neuroplasticity by downregulating brain-derived neurotrophic factor (BDNF), which may be the basis of the pathophysiology of depression [41,42]. IL-6 has been shown to be a reliable positive predictor of BDNF in patients with melancholic MDD [43]. However, the correlation between inflammation and BDNF requires further study. Moreover, the ghrelin/GHSR system can also produce antidepressant effects through the regulation of BDNF, which we will elaborate on below.

2.3. The Ghrelin/GHSR System Promotes Neurogenesis in Depression

Numerous clinical studies have shown that depression is closely associated with decreased size and function of the hippocampus and amygdala in limbic brain regions, and that patients with depression have reduced hippocampal volume and decreased neurogenesis [44,45], where neurogenesis is closely related to the treatment of depression. Studies have found that ablation of hippocampal nerve genesis in mice impairs the efficacy of antidepressants [18]. In animal experiments, antidepressant drugs promote hippocampal neurogenesis. Similarly, alterations in adult hippocampal neurogenesis mediate the effects of antidepressants, and chronic administration of the latter enhances the former [46].
As a neuropeptide, ghrelin exhibits strong neuroprotective effects. In rat cortical neuronal damage induced by hypoxia and hypoglycemia [47], ghrelin inhibited the neuronal damage process, and the protective effect disappeared after administration of a GHSR-specific inhibitor. In addition to this, a reduction in oligodendrocyte and neuronal apoptosis was found in an experiment in which ghrelin was administered after spinal cord injury in rats [48]. Most notably, the antidepressant effects exerted by ghrelin in vivo can be dependent on its neuroprotective effects. The published literature shows that ghrelin directly increases hippocampal neurogenesis in the treatment of depression [49]. Further studies have identified hippocampal neuroprotection as the primary mechanism by which a stress-induced increase in ghrelin protects the organism from the stress-induced worsening of associated depression [50]. In addition to the protective effects of ghrelin on hippocampal neurons against depression, the activation of catecholaminergic neurons has also been identified as a possible mechanism contributing to the antidepressant effects of ghrelin [51]. Although numerous studies have demonstrated that ghrelin exerts its antidepressant effects through neuroprotection, the mechanisms involved are currently not well understood. It has been found that in the hippocampus, ghrelin is able to cross the BBB and bind to GHSR1a to improve cognitive function and enhance hippocampal neurogenesis [52], and it can enhance LTP [53]. Ghrelin also directly induces the proliferation and differentiation of adult neural progenitor cells in the hippocampal subgranular zone. The number of progenitor cells was reduced in the GHSR1a KO mouse compared with the wild-type controls [49]. In addition to this, another report concluded that ghrelin provides neuroprotection through the activation of AMPK and enhances the clearance of damaged mitochondria [54], and it has also been demonstrated that ghrelin mediates neuroprotection through the inhibition of glial cell activation and the release of pro-inflammatory mediators [55]. In addition to the above-mentioned related mechanisms, it has been experimentally confirmed that BDNF plays an important role in the pathophysiology of depression [56]. The antidepressant effect exerted by ghrelin by regulating the relevant expression of BDNF is also a hot topic of research.

2.4. The Regulation of Astrocyte Physiology by the Ghrelin/GHSR System

Analysis of post-mortem tissue from patients with affective disorders has revealed a decreased number of astrocytes in several brain areas [57]. In addition, chronic unpredictable mild stress (CUMS)-induced cellular, metabolic and behavioral deficits can be reversed with antidepressants, which promotes glial cell Glu clearance [58]. Another investigation reported that the long-term administration of the antidepressant fluoxetine reversed the stress-induced decline in the number of hippocampal glial cells in tree shrews, and the relevance of the structural plasticity of astrocytes in stress and therapeutic support with antidepressants was proposed [59]. These results all suggest that antidepressant-mediated changes in astrocytes may be key to their effects and that these cells may play an active role in brain function that is connected to the process of depression.
Traditionally, astrocytes have often been thought of as brain glue, a class of cells that only provide metabolic and functional support to neurons. However, with the discovery of various neurotransmitter receptors and channels on the astrocyte membrane, our understanding of the function of astrocytes in the nervous system has fundamentally changed [60]. A large number of studies have found that the most prominent neurotransmitter receptors expressed on astrocyte membranes are GPCRs, including metabotropic glutamate receptors, adrenergic receptors, GABAergic receptors, cholinergic receptors, histaminic receptors, dopaminergic receptors, and neurotrophic factor receptors [61,62,63]. Upon activation of these receptors by neurotransmitters released by presynaptic neurons, astrocytes can undergo “gliotransduction” [64], releasing gliotransmitters to feed back on neuronal excitability and synaptic transmission [65,66]. Thus, astrocytes are now considered to be active participants in neuronal communication.
GHSR1a has been shown to be expressed in astrocytes in the arcuate nucleus of the hypothalamus [67] and the dentate gyrus of the hippocampus [68]. Therefore, exploring whether ghrelin is associated with astrocytes will be very interesting. A study found that the astrocytoma cell line C6 could respond to GHRP-6 by upregulating GHSR1a levels, increasing the activation of the PI3K/Akt pathway and its own proliferation [69]. Moreover, this effect can be inhibited by D-Lys3-GHRP-6, an antagonist of GHSR1a [68]; so, ghrelin may exert neuroprotective effects by stimulating astrocyte proliferation through GHSR1a to increase the expression of the PI3K/Akt pathway. However, another study found that ghrelin did not show a promotive effect on astrocyte proliferation and that it reversed the activation and accumulation of astrocytes in hippocampal neurodegeneration following hippocampal excitotoxicity injury induced by sea manate [55]. This difference may be related to the activation of astrocytes and the increase in the concentrated release of pro-inflammatory cytokines in inflammatory and related conditions and the triggering of neuroinflammation [70]. In conclusion, the specific mechanisms associated with ghrelin and astrocytes need to be investigated more thoroughly, as ghrelin may have a neuroprotective role in the development of depression through the regulation of astrocyte physiological activity by GHSR1a.

2.5. The Role of the Ghrelin/GHSR System in Endocrine Disruption in Depression

Depression has long been recognized as having a correlation with endocrine disruption, with the overactivity of the hypothalamic–pituitary–adrenal (HPA) axis being the most common trait [71]. In addition, the hypothalamic–pituitary–thyroid (HPT) axis and the hypothalamic–pituitary–gonadal (HPG) axis are also disturbed in depressed patients [72,73]. Therefore, changes in the levels of related hormones in the body can provide ideas for research on the treatment of depression. For example, testosterone levels are reduced in men suffering from depression [74], and the vulnerability of perimenopausal women to depression is associated with changes in estrogen [75]. Then, we can consider testosterone and estrogen to be targets for further research on antidepressants.
There is growing evidence that ghrelin is involved in the regulation of endocrine disruption in depression. For example, ghrelin can activate hypophysiotropic corticotropin-releasing factor (CRF) neurons and the HPA axis via inhibition of local GABAergic tone, in an arcuate nucleus (ARC)-independent manner [76,77]. Sarah et al. found that ghrelin suppressed anxiety after acute stress by stimulating the HPA axis at the level of the anterior pituitary [78]. However, des-acyl ghrelin administration has differential effects on the HPA axis and anxiety-like behavior in ghrelin-oacyltransferase (GOAT) KO mice and ghrelin KO mice [79]. Des-acyl ghrelin produces anxiolytic effect under stress, but GOAT KO mice showed anxiety-like behavior under stressed conditions. Ghrelin also affects the activity of the HPT axis by decreasing thyroid-stimulating hormone and increasing free thyroxine in the plasma [80]. In addition, the β1-adrenergic receptor blocker atenolol exacerbated depression-like behaviors in chronic social defeat stress (CSDS) mice by attenuating the increase in plasma acylgrowth factor-releasing peptide [81]. In conclusion, the relevant role of ghrelin in endocrine disruption in depressed patients requires more research.

3. Signaling Pathways Induced by Ghrelin/GHSR1a System in Depression

3.1. cAMP/CREB/BDNF Signaling Pathway

Neurodegenerative and neuropsychiatric disorders can be caused by inadequate supply of neurotrophic factors [82]. Among them, BDNF, as a member of the neurotrophic protein family, plays extremely important roles in promoting neuronal growth, survival, and differentiation; in synaptic transmission; in enhancing central plasticity [83,84,85]; and in slowing down depressive progression. Relatedly, ghrelin increases total BDNF mRNA expression in the mouse hippocampus and synthesizes different kinds of BDNF mRNAs by acting on different promoters in rats of different ages [86]. Different BDNF transcripts exhibit different subcellular localization that selectively shape the proximal and distal compartments of the cytosol or dendrites [87] and play an important role in increasing neuronal plasticity [88].
Impaired cAMP signaling occurs in patients with major depression. In addition, in the hippocampus and prefrontal cortex of patients, the levels of BDNF, CREB, and p-CREB are significantly reduced [89,90,91,92], and so are the levels of BDNF mRNA in peripheral monocytes in this population [93]. Interestingly, the central administration of ghrelin normalized hippocampal BDNF levels [94]. In addition, exogenous ghrelin can improve depressive behavior by upregulating CREB signaling through the activation of ghrelin receptors and the cAMP/PKA signaling pathway and by increasing BDNF expression downstream [24]. Thus, ghrelin-induced increases in BDNF in the hippocampus involve the activation of the GHSR1a/cAMP/PKA/CREB signaling pathway (Figure 1).

3.2. p38-MAPK Signaling Pathway

Several studies have demonstrated that p38-MAPK is activated in response to various stressful stimuli and is involved in the pathologic process of depression [95,96]. p38-MAPK can be activated by interferon and lipopolysaccharide to upregulate the expression of the depression-related gene IDO [97,98], and it can also exacerbate esophageal cancer-associated depression by directly enhancing the expression of the IDO gene [99]. Therefore, the expression status of p38-MAPK-pathway-related substances could be a powerful tool for depression monitoring, while p38-MAPK itself could be a target for antidepressant research. In addition, the activation of the p38-MAPK pathway phosphorylates glucocorticoid receptor (GR), whose phosphorylation is associated with reduced glucocorticoid sensitivity [100], which may be closely related to the glucocorticoid resistance exhibited by depressed patients [101]. In contrast, ghrelin treatment in rats activates GHSR1a and decreases p38-MAPK phosphorylation, which, in turn, increases the GR levels [102]. Furthermore, no significant increase in the phosphorylation of p38 by CSDS in vector-treated mice was observed after ghrelin treatment. Additionally, hippocampal GHSR-deficient mice showed higher levels of p38 phosphorylation than control mice, suggesting that ghrelin may also mediate antidepressant mechanisms by inhibiting the p38-MAPK signaling pathway in the hippocampus [103]. Interestingly, social failure stress produced depression-like behavior in wild-type mice, but the selective deletion of p38-MAPK in the serotonergic neurons of the nucleus dorsalis of the mouse middle suture protected the mice under stress induction [104]. This suggests that p38-MAPK has the ability to specifically regulate selected downstream targets; thus, the role played by this pathway in antidepressant disorders requires further investigation.

3.3. PI3K/Akt Signaling Pathway

Depression is also closely related to neurogenic hypoplasia [105], and PI3K/Akt is thought to be an important signal for the proliferation of adult hippocampal progenitor cells [106]. Akt can exert its utility in controlling cellular proliferation by activating the phosphorylation of its downstream targets, GSK-3β, mTOR, and p70S6K, where GSK-3β is a pro-apoptotic protein whose activity plays an important role in neuropathology and psychiatric disorders [107]. β-Catenin, as a transcription factor regulated by GSK-3β, undergoes nuclear translocation under conditions of GSK-3β inactivation [108], which is an indispensable step in its role in promoting cell survival. In addition, downstream of PI3K/Akt, the phosphorylation of mTOR and p70S6K also promotes the proliferation of neural stem cells [109]. Interestingly, ghrelin could induce hippocampal neural stem cell (NSC) proliferation by activating the PI3K/Akt signaling pathway by binding to GHSR1a, and the stimulatory effects of ghrelin on GSK-3β, mTOR, and p70S6K phosphorylation were significantly inhibited by treatment with GHSR1a-specific antagonist D-Lys-3-GHRP-6 [110]. Furthermore, it has been demonstrated that ghrelin enhances the nuclear translocation of β-catenin, which, in turn, contributes to its anti-apoptotic effects [111]. Therefore, we may hypothesize that ghrelin may promote neuronal cell proliferation through the activation of the PI3K/Akt pathway and subsequently play a role in the treatment of depression. Moreover, autophagy plays an important role in maintaining neuronal stem cells and adult neuronal plasticity, while ghrelin can stimulate autophagy by inhibiting the PI3K/AKT/MTOR signaling pathway [112]. However, an experiment in a mouse model of corticosterone-induced depression found that overactive neuronal autophagy depleted BDNF and impaired adult hippocampal neurogenesis [84]. Therefore, it remains to be investigated whether ghrelin can regulate autophagy homeostasis in vivo through the PI3K/AKT/MTOR pathway and promote neurogenesis.

3.4. Jak2/STAT3 Signaling Pathway

The Jak2/STAT3 signaling pathway, like the aforementioned PI3K/Akt signaling pathway, has also been shown to play an important role in neuroprotection. Unlike single pathways that act independently, one study found that resveratrol may exert neuronal protection by indirectly upregulating the PI3K/Akt/mTOR pathway through the activation of Jak2/STAT3 [113]. As shown in Figure 1, the exposure of rat hippocampal NSCs to the Jak2/STAT3 inhibitor cucurbitacin I significantly blocked the proliferative effects of ghrelin on NSCs [110]. Thus, ghrelin could also be potent through the activation of the Jak2/STAT3 pathway. Furthermore, this pathway not only plays an active role in neuroprotection and regeneration, but its role in neuroinflammation has also been shown to be promoted. It has been found that the inhibition of the Jak2/STAT3 pathway ameliorates neuroinflammation [114] and reduces neuronal senescence by suppressing the inflammatory response [115]. Since depression is closely related to decreased neuronal genesis and the upregulation of inflammatory factors and given that the activation of the Jak2/STAT3 pathway has been found to have completely opposite effects in these two aspects, whether this pathway can be a therapeutic target for depression needs to be investigated at a deeper level in terms of its multifaceted mechanisms. The study of whether ghrelin regulates neuroinflammation when activating the Jak2/STAT3 pathway will be an important basis for comprehensively determining whether ghrelin can be used as a target for the treatment of depression.

4. Ghrelin/GHSR as a Therapeutic Target for Depressive Disorder

Many neuropeptides have been reported as targets for depression treatment. For example, nonselective glycopeptide receptor agonists (galnon) can exert antidepressant effects in preclinical models of depression [116,117]. VP antagonists have shown similar antidepressant behavior in preclinical studies [118,119]. Similarly, many studies have shown that ghrelin can be used as a powerful tool in the treatment of depression. It has been reported that ghrelin produces antidepressant effects in estrogen-deficient mice [94] and that it may also counteract depressive symptoms caused by chronic stress [120]. In addition, many substances that exert antidepressant effects by modulating ghrelin/GHSR expression have been identified. For example, paeoniflorin (PF) significantly increased the expression of GHSR1a to mediate antidepressant effects [121], and the GHSR inhibitor JMV29282259 blocked the saffronin-induced expression of neuroplasticity-related proteins [122]. Here, we briefly summarized the correlation between the ghrelin/GHSR system and depression, as well as the research methodology, as shown in Table 1. Thus, the ghrelin/GHSR system has many potent functions in the defense against depression-like symptoms. However, further studies are needed because of the stable antidepressant behavioral effects of neuropeptides expressed in various tests and the two-sided nature of ghrelin’s effects on depression in different models [123]. Recently, liver-expressed antimicrobial peptide 2 (LEAP2) was reported as an endogenous antagonist of GHSR and produced in the liver and small intestine [124,125]. Whether LEAP2 is involved in the development of depression needs further study.

5. Conclusions and Future Direction

There is an ongoing debate about the role of ghrelin in depression. The majority of studies suggest that ghrelin has antidepressant effects, with few studies indicating depressogenic effects. Indeed, ghrelin/GHSR can exert antidepressant and neuroprotective effects by triggering multiple signaling pathways, including cAMP/CREB/BDNF, PI3K/Akt, Jak2/STAT3, and p38-MAPK. Moreover, GHSR1a can also form dimers with other GPCRs to exert antidepressant effects. Therefore, the ghrelin/GHSR system is becoming a new target for the treatment of depression. However, there are two-sided claims on ghrelin’s antidepressant potency according to different experiments; for instance, ghrelin had no antidepressant effect on young rats, and the drug’s neuropharmacology differed in adolescents and adults. Future experiments should, therefore, focus on investigating the link between the pathogenesis of ghrelin and depression in different depressive groups, where the development of receptor-biased drugs is also a good strategy. Moreover, more studies are necessary to determine the extent to which central and peripheral ghrelin signaling are functionally interconnected, as this understanding is crucial to the development of new ghrelin-based therapeutic agents. Finally, promising findings from animal studies necessitate further human-based research to ascertain the extent to which such results can be applied to human disorders.

Author Contributions

Writing—original draft preparation, X.P. and Y.G.; writing—review and editing, J.C. and B.J.; supervision, K.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation of Shandong Province (ZR2020MH148) and the University Student Innovative Training Program of Jining Medical University (cx2023072z). And The APC was funded by ZR2020MH148.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

BBB, blood–brain barrier; BDNF, brain-derived neurotrophic factor; CAO, coronary artery occlusion; CDK, cyclin-dependent kinases; CNS, central nervous system; CREB, cAMP response element-binding protein; CSDS, chronic social defeat stress; CUMS, chronic unpredictable mild stress; DDR, disturbed diurnal rhythm; EPM, elevated plus maze; FST, forced swimming test; GFAP, glial fibrillary acidic protein; GHSR, growth-hormone-releasing peptide receptor; GSK-3β, glycogen synthase kinase 3β; GR, glucocorticoid receptor; HFD, high-fat diet; HPA, hypothalamic–pituitary–adrenal; HPT, hypothalamic–pituitary–thyroid; HPG, hypothalamic–pituitary–gonadal; Iba-1, ionized calcium-binding adapter molecule 1; LH, luteinizing hormone; MDD, major depressive disorder; mPFC, medial prefrontal cortex; mTOR, mammalian target of rapamycin; NSCs, neural stem cells; OFT, open field test; OX1R, orexin 1 receptor; OB, olfactory bulbectomy surgery; p70S6K, p70 ribosomal protein S6 kinase; RS, restraint stress; SIT, social interaction test; SPT, sucrose preference test; TST, tail suspension test.

References

  1. Gnanapavan, S.; Kola, B.; Bustin, S.A.; Morris, D.G.; McGee, P.; Fairclough, P.; Bhattacharya, S.; Carpenter, R.; Grossman, A.B.; Korbonits, M. The Tissue Distribution of the mRNA of Ghrelin and Subtypes of Its Receptor, GHS-R, in Humans. J. Clin. Endocrinol. Metab. 2002, 87, 2988–2991. [Google Scholar] [CrossRef] [PubMed]
  2. Kojima, M.; Kangawa, K. Ghrelin: Structure and Function. Physiol. Rev. 2005, 85, 495–522. [Google Scholar] [CrossRef] [PubMed]
  3. Albarrán-Zeckler, R.G.; Smith, R.G. The Ghrelin Receptors (GHS-R1a and GHS-R1b). In The Ghrelin System; S. Karger AG: Basel, Switzerland, 2013; pp. 5–15. [Google Scholar]
  4. Sun, Y.; Asnicar, M.; Smith, R.G. Central and Peripheral Roles of Ghrelin on Glucose Homeostasis. Neuroendocrinology 2007, 86, 215–228. [Google Scholar] [CrossRef] [PubMed]
  5. Gortan Cappellari, G.; Barazzoni, R. Ghrelin forms in the modulation of energy balance and metabolism. Eat. Weight Disord.-Stud. Anorex. Bulim. Obes. 2018, 24, 997–1013. [Google Scholar] [CrossRef] [PubMed]
  6. Vinci, M.C.; Fujimura, K.; Wakino, S.; Minakuchi, H.; Hasegawa, K.; Hosoya, K.; Komatsu, M.; Kaneko, Y.; Shinozuka, K.; Washida, N.; et al. Ghrelin Protects against Renal Damages Induced by Angiotensin-II via an Antioxidative Stress Mechanism in Mice. PLoS ONE 2014, 9, e94373. [Google Scholar] [CrossRef]
  7. Laviano, A.; Molfino, A.; Rianda, S.; Rossi Fanelli, F. The growth hormone secretagogue receptor (Ghs-R). Curr. Pharm. Des. 2012, 18, 4749–4754. [Google Scholar] [CrossRef] [PubMed]
  8. Leung, P.-K.; Chow, K.B.S.; Lau, P.-N.; Chu, K.-M.; Chan, C.-B.; Cheng, C.H.K.; Wise, H. The truncated ghrelin receptor polypeptide (GHS-R1b) acts as a dominant-negative mutant of the ghrelin receptor. Cell. Signal. 2007, 19, 1011–1022. [Google Scholar] [CrossRef]
  9. Chan, C.-B.; Leung, P.-K.; Wise, H.; Cheng, C.H.K. Signal transduction mechanism of the seabream growth hormone secretagogue receptor. FEBS Lett. 2004, 577, 147–153. [Google Scholar] [CrossRef]
  10. Tang, T.-T.; Bi, M.-X.; Diao, M.-N.; Zhang, X.-Y.; Chen, L.; Xiao, X.; Jiao, Q.; Chen, X.; Yan, C.-L.; Du, X.-X.; et al. Quinpirole ameliorates nigral dopaminergic neuron damage in Parkinson’s disease mouse model through activating GHS-R1a/D(2)R heterodimers. Acta Pharmacol. Sin. 2023, 44, 1564–1575. [Google Scholar] [CrossRef]
  11. Liu, X.; Lan, X.; Zhang, X.; Ye, H.; Shen, L.; Hu, M.; Chen, X.; Zheng, M.; Weston-Green, K.; Jin, T.; et al. Olanzapine attenuates 5-HT2cR and GHSR1a interaction to increase orexigenic hypothalamic NPY: Implications for neuronal molecular mechanism of metabolic side effects of antipsychotics. Behav. Brain Res. 2024, 463, 114885. [Google Scholar] [CrossRef]
  12. Xue, Q.; Bai, B.; Ji, B.; Chen, X.; Wang, C.; Wang, P.; Yang, C.; Zhang, R.; Jiang, Y.; Pan, Y.; et al. Ghrelin through GHSR1a and OX1R Heterodimers Reveals a Gαs-cAMP-cAMP Response Element Binding Protein Signaling Pathway in Vitro. Front. Mol. Neurosci. 2018, 11, 245. [Google Scholar] [CrossRef] [PubMed]
  13. Duman, R.S.; Voleti, B. Signaling pathways underlying the pathophysiology and treatment of depression: Novel mechanisms for rapid-acting agents. Trends Neurosci. 2012, 35, 47–56. [Google Scholar] [CrossRef] [PubMed]
  14. Hauenstein, E.J. Depression in adolescence. J. Obstet. Gynecol. Neonatal Nurs. 2003, 32, 239–248. [Google Scholar] [CrossRef] [PubMed]
  15. Alexopoulos, G.S. Depression in the elderly. Lancet 2005, 365, 1961–1970. [Google Scholar] [CrossRef] [PubMed]
  16. Park, C.; Rosenblat, J.D.; Brietzke, E.; Pan, Z.; Lee, Y.; Cao, B.; Zuckerman, H.; Kalantarova, A.; McIntyre, R.S. Stress, epigenetics and depression: A systematic review. Neurosci. Biobehav. Rev. 2019, 102, 139–152. [Google Scholar] [CrossRef] [PubMed]
  17. Felger, J.C.; Lotrich, F.E. Inflammatory cytokines in depression: Neurobiological mechanisms and therapeutic implications. Neuroscience 2013, 246, 199–229. [Google Scholar] [CrossRef] [PubMed]
  18. MacQueen, G.; Frodl, T. The hippocampus in major depression: Evidence for the convergence of the bench and bedside in psychiatric research? Mol. Psychiatry 2010, 16, 252–264. [Google Scholar] [CrossRef] [PubMed]
  19. Salvat-Pujol, N.; Labad, J.; Urretavizcaya, M.; de Arriba-Arnau, A.; Segalàs, C.; Real, E.; Ferrer, A.; Crespo, J.M.; Jiménez-Murcia, S.; Soriano-Mas, C.; et al. Hypothalamic-pituitary-adrenal axis activity and cognition in major depression: The role of remission status. Psychoneuroendocrinology 2017, 76, 38–48. [Google Scholar] [CrossRef]
  20. Nierenberg, A.A. Current perspectives on the diagnosis and treatment of major depressive disorder. Am. J. Manag. Care 2001, 7, S353–S366. [Google Scholar]
  21. Pruckner, N.; Holthoff-Detto, V. Antidepressant pharmacotherapy in old-age depression—A review and clinical approach. Eur. J. Clin. Pharmacol. 2017, 73, 661–667. [Google Scholar] [CrossRef]
  22. Gonda, X.; Dome, P.; Neill, J.C.; Tarazi, F.I. Novel antidepressant drugs: Beyond monoamine targets. CNS Spectr. 2021, 28, 6–15. [Google Scholar] [CrossRef]
  23. Hashimoto, K.; Naudet, F.; Maria, A.S.; Falissard, B. Antidepressant Response in Major Depressive Disorder: A Meta-Regression Comparison of Randomized Controlled Trials and Observational Studies. PLoS ONE 2011, 6, e20811. [Google Scholar] [CrossRef]
  24. Li, Y.-H.; Liu, Q.-X.; Wang, J.-S.; Xiang, H.; Zhang, R.-F.; Huang, C.-Q. Ghrelin improves cognition via activation of the cAMP- CREB signalling pathway in depressed male C57BL/6J mice. Int. J. Neurosci. 2023, 133, 1233–1241. [Google Scholar] [CrossRef]
  25. Bianconi, S.; Poretti, M.B.; Rodríguez, P.; Maestri, G.; Rodríguez, P.E.; de Barioglio, S.R.; Schiöth, H.B.; Carlini, V.P. Ghrelin restores memory impairment following olfactory bulbectomy in mice by activating hippocampal NMDA1 and MAPK1 gene expression. Behav. Brain Res. 2021, 410, 113341. [Google Scholar] [CrossRef]
  26. Guo, L.; Niu, M.; Yang, J.; Li, L.; Liu, S.; Sun, Y.; Zhou, Z.; Zhou, Y. GHS-R1a Deficiency Alleviates Depression-Related Behaviors After Chronic Social Defeat Stress. Front. Neurosci. 2019, 13, 364. [Google Scholar] [CrossRef]
  27. Hirschfeld, R.M. History and evolution of the monoamine hypothesis of depression. J. Clin. Psychiatry 2000, 61 (Suppl. S6), 4–6. [Google Scholar]
  28. Jiang, H.; Betancourt, L.; Smith, R.G. Ghrelin Amplifies Dopamine Signaling by Cross Talk Involving Formation of Growth Hormone Secretagogue Receptor/Dopamine Receptor Subtype 1 Heterodimers. Mol. Endocrinol. 2006, 20, 1772–1785. [Google Scholar] [CrossRef]
  29. Hansson, C.; Alvarez-Crespo, M.; Taube, M.; Skibicka, K.P.; Schmidt, L.; Karlsson-Lindahl, L.; Egecioglu, E.; Nissbrandt, H.; Dickson, S.L. Influence of ghrelin on the central serotonergic signaling system in mice. Neuropharmacology 2014, 79, 498–505. [Google Scholar] [CrossRef]
  30. Sakata, I.; Gong, Z.; Ikenoya, C.; Takemi, S.; Sakai, T. The study of ghrelin secretion and acyl-modification using mice and ghrelinoma cell lines. Endocr. J. 2017, 64, S27–S29. [Google Scholar] [CrossRef]
  31. Zhao, T.J.; Sakata, I.; Li, R.L.; Liang, G.; Richardson, J.A.; Brown, M.S.; Goldstein, J.L.; Zigman, J.M. Ghrelin secretion stimulated by {beta}1-adrenergic receptors in cultured ghrelinoma cells and in fasted mice. Proc. Natl. Acad. Sci. USA 2010, 107, 15868–15873. [Google Scholar] [CrossRef]
  32. López Soto, E.J.; Agosti, F.; Cabral, A.; Mustafa, E.R.; Damonte, V.M.; Gandini, M.A.; Rodríguez, S.; Castrogiovanni, D.; Felix, R.; Perelló, M.; et al. Constitutive and ghrelin-dependent GHSR1a activation impairs CaV2.1 and CaV2.2 currents in hypothalamic neurons. J. Gen. Physiol. 2015, 146, 205–219. [Google Scholar] [CrossRef]
  33. Granado, M.; Priego, T.; Martín, A.I.; Villanúa, M.Á.; López-Calderón, A. Anti-inflammatory effect of the ghrelin agonist growth hormone-releasing peptide-2 (GHRP-2) in arthritic rats. Am. J. Physiol.-Endocrinol. Metab. 2005, 288, E486–E492. [Google Scholar] [CrossRef]
  34. Schiepers, O.J.G.; Wichers, M.C.; Maes, M. Cytokines and major depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2005, 29, 201–217. [Google Scholar] [CrossRef]
  35. Hodes, G.E.; Ménard, C.; Russo, S.J. Integrating Interleukin-6 into depression diagnosis and treatment. Neurobiol. Stress 2016, 4, 15–22. [Google Scholar] [CrossRef]
  36. Otero, M. Chronic inflammation modulates ghrelin levels in humans and rats. Rheumatology 2003, 43, 306–310. [Google Scholar] [CrossRef]
  37. Cheyuo, C.; Wu, R.; Zhou, M.; Jacob, A.; Coppa, G.; Wang, P. Ghrelin Suppresses Inflammation and Neuronal Nitric Oxide Synthase in Focal Cerebral Ischemia Via the Vagus Nerve. Shock 2011, 35, 258–265. [Google Scholar] [CrossRef]
  38. Waseem, T.; Duxbury, M.; Ito, H.; Ashley, S.W.; Robinson, M.K. Exogenous ghrelin modulates release of pro-inflammatory and anti-inflammatory cytokines in LPS-stimulated macrophages through distinct signaling pathways. Surgery 2008, 143, 334–342. [Google Scholar] [CrossRef]
  39. Dixit, V.D.; Schaffer, E.M.; Pyle, R.S.; Collins, G.D.; Sakthivel, S.K.; Palaniappan, R.; Lillard, J.W.; Taub, D.D. Ghrelin inhibits leptin- and activation-induced proinflammatory cytokine expression by human monocytes and T cells. J. Clin. Investig. 2004, 114, 57–66. [Google Scholar] [CrossRef]
  40. Lin, L.; Lee, J.H.; Buras, E.D.; Yu, K.; Wang, R.; Smith, C.W.; Wu, H.; Sheikh-Hamad, D.; Sun, Y. Ghrelin receptor regulates adipose tissue inflammation in aging. Aging 2016, 8, 178–191. [Google Scholar] [CrossRef]
  41. Krishnan, V.; Nestler, E.J. The molecular neurobiology of depression. Nature 2008, 455, 894–902. [Google Scholar] [CrossRef]
  42. Schmidt, H.D.; Shelton, R.C.; Duman, R.S. Functional Biomarkers of Depression: Diagnosis, Treatment, and Pathophysiology. Neuropsychopharmacology 2011, 36, 2375–2394. [Google Scholar] [CrossRef]
  43. Patas, K.; Penninx, B.W.J.H.; Bus, B.A.A.; Vogelzangs, N.; Molendijk, M.L.; Elzinga, B.M.; Bosker, F.J.; Oude Voshaar, R.C. Association between serum brain-derived neurotrophic factor and plasma interleukin-6 in major depressive disorder with melancholic features. Brain Behav. Immun. 2014, 36, 71–79. [Google Scholar] [CrossRef]
  44. Berman, R.M.; Cappiello, A.; Anand, A.; Oren, D.A.; Heninger, G.R.; Charney, D.S.; Krystal, J.H. Antidepressant effects of ketamine in depressed patients. Biol. Psychiatry 2000, 47, 351–354. [Google Scholar] [CrossRef]
  45. Zarate, C.A., Jr.; Singh, J.B.; Carlson, P.J.; Brutsche, N.E.; Ameli, R.; Luckenbaugh, D.A.; Charney, D.S.; Manji, H.K. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch. Gen. Psychiatry 2006, 63, 856–864. [Google Scholar] [CrossRef]
  46. Li, N.; Lee, B.; Liu, R.-J.; Banasr, M.; Dwyer, J.M.; Iwata, M.; Li, X.-Y.; Aghajanian, G.; Duman, R.S. mTOR-Dependent Synapse Formation Underlies the Rapid Antidepressant Effects of NMDA Antagonists. Science 2010, 329, 959–964. [Google Scholar] [CrossRef]
  47. Park, S.; Kim, H.; Lee, D.; Ju, S.; Seo, S.; Lee, D.H.; Kim, E.; Chung, H. Ghrelin Inhibits Apoptosis in Hypothalamic Neuronal Cells during Oxygen-Glucose Deprivation. Endocrinology 2007, 148, 148–159. [Google Scholar] [CrossRef]
  48. Lee, J.Y.; Chung, H.; Yoo, Y.S.; Oh, Y.J.; Oh, T.H.; Park, S.; Yune, T.Y. Inhibition of Apoptotic Cell Death by Ghrelin Improves Functional Recovery after Spinal Cord Injury. Endocrinology 2010, 151, 3815–3826. [Google Scholar] [CrossRef]
  49. Li, E.; Chung, H.; Kim, Y.; Kim, D.H.; Ryu, J.H.; Sato, T.; Kojima, M.; Park, S. Ghrelin directly stimulates adult hippocampal neurogenesis: Implications for learning and memory. Endocr. J. 2013, 60, 781–789. [Google Scholar] [CrossRef]
  50. Walker, A.K.; Rivera, P.D.; Wang, Q.; Chuang, J.C.; Tran, S.; Osborne-Lawrence, S.; Estill, S.J.; Starwalt, R.; Huntington, P.; Morlock, L.; et al. The P7C3 class of neuroprotective compounds exerts antidepressant efficacy in mice by increasing hippocampal neurogenesis. Mol. Psychiatry 2014, 20, 500–508. [Google Scholar] [CrossRef]
  51. Chuang, J.-C.; Perello, M.; Sakata, I.; Osborne-Lawrence, S.; Savitt, J.M.; Lutter, M.; Zigman, J.M. Ghrelin mediates stress-induced food-reward behavior in mice. J. Clin. Investig. 2011, 121, 2684–2692. [Google Scholar] [CrossRef]
  52. Cuellar, J.N.; Isokawa, M. Ghrelin-induced activation of cAMP signal transduction and its negative regulation by endocannabinoids in the hippocampus. Neuropharmacology 2011, 60, 842–851. [Google Scholar] [CrossRef]
  53. Diano, S.; Farr, S.A.; Benoit, S.C.; McNay, E.C.; da Silva, I.; Horvath, B.; Gaskin, F.S.; Nonaka, N.; Jaeger, L.B.; Banks, W.A.; et al. Ghrelin controls hippocampal spine synapse density and memory performance. Nat. Neurosci. 2006, 9, 381–388. [Google Scholar] [CrossRef]
  54. Bayliss, J.A.; Andrews, Z.B. Ghrelin is neuroprotective in Parkinson’s disease: Molecular mechanisms of metabolic neuroprotection. Ther. Adv. Endocrinol. Metab. 2013, 4, 25–36. [Google Scholar] [CrossRef]
  55. Lee, J.; Lim, E.; Kim, Y.; Li, E.; Park, S. Ghrelin attenuates kainic acid-induced neuronal cell death in the mouse hippocampus. J. Endocrinol. 2010, 205, 263–270. [Google Scholar] [CrossRef]
  56. Fornaro, M.; Escelsior, A.; Rocchi, G.; Conio, B.; Magioncalda, P.; Marozzi, V.; Presta, A.; Sterlini, B.; Contini, P.; Amore, M.; et al. BDNF plasma levels variations in major depressed patients receiving duloxetine. Neurol. Sci. 2014, 36, 729–734. [Google Scholar] [CrossRef]
  57. Choudary, P.V.; Molnar, M.; Evans, S.J.; Tomita, H.; Li, J.Z.; Vawter, M.P.; Myers, R.M.; Bunney, W.E., Jr.; Akil, H.; Watson, S.J.; et al. Altered cortical glutamatergic and GABAergic signal transmission with glial involvement in depression. Proc. Natl. Acad. Sci. USA 2005, 102, 15653–15658. [Google Scholar] [CrossRef]
  58. Banasr, M.; Chowdhury, G.M.I.; Terwilliger, R.; Newton, S.S.; Duman, R.S.; Behar, K.L.; Sanacora, G. Glial pathology in an animal model of depression: Reversal of stress-induced cellular, metabolic and behavioral deficits by the glutamate-modulating drug riluzole. Mol. Psychiatry 2008, 15, 501–511. [Google Scholar] [CrossRef]
  59. Czéh, B.; Simon, M.; Schmelting, B.; Hiemke, C.; Fuchs, E. Astroglial Plasticity in the Hippocampus is Affected by Chronic Psychosocial Stress and Concomitant Fluoxetine Treatment. Neuropsychopharmacology 2005, 31, 1616–1626. [Google Scholar] [CrossRef]
  60. Volterra, A.; Meldolesi, J. Astrocytes, from brain glue to communication elements: The revolution continues. Nat. Rev. Neurosci. 2005, 6, 626–640. [Google Scholar] [CrossRef]
  61. Rose, C.R.; Blum, R.; Pichler, B.; Lepier, A.; Kafitz, K.W.; Konnerth, A. Truncated TrkB-T1 mediates neurotrophin-evoked calcium signalling in glia cells. Nature 2003, 426, 74–78. [Google Scholar] [CrossRef]
  62. Zhang, J.M.; Wang, H.K.; Ye, C.Q.; Ge, W.; Chen, Y.; Jiang, Z.L.; Wu, C.P.; Poo, M.M.; Duan, S. ATP released by astrocytes mediates glutamatergic activity-dependent heterosynaptic suppression. Neuron 2003, 40, 971–982. [Google Scholar] [CrossRef]
  63. Araque, A.; Martín, E.D.; Perea, G.; Arellano, J.I.; Buño, W. Synaptically released acetylcholine evokes Ca2+ elevations in astrocytes in hippocampal slices. J. Neurosci. Off. J. Soc. Neurosci. 2002, 22, 2443–2450. [Google Scholar] [CrossRef]
  64. Bezzi, P.; Volterra, A. A neuron-glia signalling network in the active brain. Curr. Opin. Neurobiol. 2001, 11, 387–394. [Google Scholar] [CrossRef]
  65. Halassa, M.M.; Haydon, P.G. Integrated Brain Circuits: Astrocytic Networks Modulate Neuronal Activity and Behavior. Annu. Rev. Physiol. 2010, 72, 335–355. [Google Scholar] [CrossRef]
  66. Araque, A.; Parpura, V.; Sanzgiri, R.P.; Haydon, P.G. Tripartite synapses: Glia, the unacknowledged partner. Trends Neurosci. 1999, 22, 208–215. [Google Scholar] [CrossRef]
  67. Fuente-Martín, E.; García-Cáceres, C.; Argente-Arizón, P.; Díaz, F.; Granado, M.; Freire-Regatillo, A.; Castro-González, D.; Ceballos, M.L.; Frago, L.M.; Dickson, S.L.; et al. Ghrelin Regulates Glucose and Glutamate Transporters in Hypothalamic Astrocytes. Sci. Rep. 2016, 6, 23673. [Google Scholar] [CrossRef]
  68. Baquedano, E.; Chowen, J.A.; Argente, J.; Frago, L.M. Differential effects of GH and GH-releasing peptide-6 on astrocytes. J. Endocrinol. 2013, 218, 263–274. [Google Scholar] [CrossRef]
  69. Dixit, V.D.; Weeraratna, A.T.; Yang, H.; Bertak, D.; Cooper-Jenkins, A.; Riggins, G.J.; Eberhart, C.G.; Taub, D.D. Ghrelin and the Growth Hormone Secretagogue Receptor Constitute a Novel Autocrine Pathway in Astrocytoma Motility. J. Biol. Chem. 2006, 281, 16681–16690. [Google Scholar] [CrossRef]
  70. Fischer, R.; Maier, O. Interrelation of Oxidative Stress and Inflammation in Neurodegenerative Disease: Role of TNF. Oxidative Med. Cell. Longev. 2015, 2015, 610813. [Google Scholar] [CrossRef]
  71. Pariante, C.M.; Lightman, S.L. The HPA axis in major depression: Classical theories and new developments. Trends Neurosci. 2008, 31, 464–468. [Google Scholar] [CrossRef] [PubMed]
  72. Vandoolaeghe, E.; Maes, M.; Vandevyvere, J.; Neels, H. Hypothalamic-pituitary-thyroid-axis function in treatment resistant depression. J. Affect. Disord. 1997, 43, 143–150. [Google Scholar] [CrossRef]
  73. Bartalena, L.; Placidi, G.F.; Martino, E.; Falcone, M.; Pellegrini, L.; Dell’Osso, L.; Pacchiarotti, A.; Pinchera, A. Nocturnal serum thyrotropin (TSH) surge and the TSH response to TSH-releasing hormone: Dissociated behavior in untreated depressives. J. Clin. Endocrinol. Metab. 1990, 71, 650–655. [Google Scholar] [CrossRef] [PubMed]
  74. Fischer, S.; Ehlert, U.; Amiel Castro, R. Hormones of the hypothalamic-pituitary-gonadal (HPG) axis in male depressive disorders—A systematic review and meta-analysis. Front. Neuroendocrinol. 2019, 55, 100792. [Google Scholar] [CrossRef]
  75. Han, Y.; Gu, S.; Li, Y.; Qian, X.; Wang, F.; Huang, J.H. Neuroendocrine pathogenesis of perimenopausal depression. Front. Psychiatry 2023, 14, 1162501. [Google Scholar] [CrossRef]
  76. Cabral, A.; Suescun, O.; Zigman, J.M.; Perello, M. Ghrelin indirectly activates hypophysiotropic CRF neurons in rodents. PLoS ONE 2012, 7, e31462. [Google Scholar] [CrossRef]
  77. Cabral, A.; Portiansky, E.; Sánchez-Jaramillo, E.; Zigman, J.M.; Perello, M. Ghrelin activates hypophysiotropic corticotropin-releasing factor neurons independently of the arcuate nucleus. Psychoneuroendocrinology 2016, 67, 27–39. [Google Scholar] [CrossRef]
  78. Spencer, S.J.; Xu, L.; Clarke, M.A.; Lemus, M.; Reichenbach, A.; Geenen, B.; Kozicz, T.; Andrews, Z.B. Ghrelin regulates the hypothalamic-pituitary-adrenal axis and restricts anxiety after acute stress. Biol. Psychiatry 2012, 72, 457–465. [Google Scholar] [CrossRef] [PubMed]
  79. Stark, R.; Santos, V.V.; Geenen, B.; Cabral, A.; Dinan, T.; Bayliss, J.A.; Lockie, S.H.; Reichenbach, A.; Lemus, M.B.; Perello, M.; et al. Des-Acyl Ghrelin and Ghrelin O-Acyltransferase Regulate Hypothalamic-Pituitary-Adrenal Axis Activation and Anxiety in Response to Acute Stress. Endocrinology 2016, 157, 3946–3957. [Google Scholar] [CrossRef]
  80. Kluge, M.; Riedl, S.; Uhr, M.; Schmidt, D.; Zhang, X.; Yassouridis, A.; Steiger, A. Ghrelin affects the hypothalamus–pituitary–thyroid axis in humans by increasing free thyroxine and decreasing TSH in plasma. Eur. J. Endocrinol. 2010, 162, 1059–1065. [Google Scholar] [CrossRef]
  81. Gupta, D.; Chuang, J.-C.; Mani, B.K.; Shankar, K.; Rodriguez, J.A.; Osborne-Lawrence, S.; Metzger, N.P.; Zigman, J.M. β1-adrenergic receptors mediate plasma acyl-ghrelin elevation and depressive-like behavior induced by chronic psychosocial stress. Neuropsychopharmacology 2019, 44, 1319–1327. [Google Scholar] [CrossRef]
  82. Zuccato, C.; Cattaneo, E. Brain-derived neurotrophic factor in neurodegenerative diseases. Nat. Rev. Neurol. 2009, 5, 311–322. [Google Scholar] [CrossRef]
  83. Keifer, J. Regulation of AMPAR trafficking in synaptic plasticity by BDNF and the impact of neurodegenerative disease. J. Neurosci. Res. 2022, 100, 979–991. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, K.; Wang, F.; Zhai, M.; He, M.; Hu, Y.; Feng, L.; Li, Y.; Yang, J.; Wu, C. Hyperactive neuronal autophagy depletes BDNF and impairs adult hippocampal neurogenesis in a corticosterone-induced mouse model of depression. Theranostics 2023, 13, 1059–1075. [Google Scholar] [CrossRef]
  85. Liu, S.; Tao, G.; Zhou, C.; Wang, Q.; Wang, W.; Fei, X. Ketamine inhibits neuronal differentiation by regulating brain-derived neurotrophic factor (BDNF) signaling. Toxicol In Vitro 2021, 72, 105091. [Google Scholar] [CrossRef]
  86. Perea Vega, M.L.; Sanchez, M.S.; Fernández, G.; Paglini, M.G.; Martin, M.; de Barioglio, S.R. Ghrelin treatment leads to dendritic spine remodeling in hippocampal neurons and increases the expression of specific BDNF-mRNA species. Neurobiol. Learn. Mem. 2021, 179, 107409. [Google Scholar] [CrossRef]
  87. Baj, G.; Leone, E.; Chao, M.V.; Tongiorgi, E. Spatial segregation of BDNF transcripts enables BDNF to differentially shape distinct dendritic compartments. Proc. Natl. Acad. Sci. USA 2011, 108, 16813–16818. [Google Scholar] [CrossRef] [PubMed]
  88. Aid, T.; Kazantseva, A.; Piirsoo, M.; Palm, K.; Timmusk, T. Mouse and rat BDNF gene structure and expression revisited. J. Neurosci. Res. 2007, 85, 525–535. [Google Scholar] [CrossRef] [PubMed]
  89. Banerjee, R.; Ghosh, A.K.; Ghosh, B.; Bhattacharyya, S.; Mondal, A.C. Decreased mRNA and Protein Expression of BDNF, NGF, and their Receptors in the Hippocampus from Suicide: An Analysis in Human Postmortem Brain. Clin. Med. Insights Pathol. 2013, 6, CPath-S12530. [Google Scholar] [CrossRef]
  90. Paska, A.V.; Zupanc, T.; Pregelj, P. The role of brain-derived neurotrophic factor in the pathophysiology of suicidal behavior. Psychiatr. Danub. 2013, 25 (Suppl. S2), S341–S344. [Google Scholar]
  91. Dowlatshahi, D.; MacQueen, G.M.; Wang, J.F.; Young, L.T. Increased temporal cortex CREB concentrations and antidepressant treatment in major depression. Lancet 1998, 352, 1754–1755. [Google Scholar] [CrossRef] [PubMed]
  92. Nakagawa, S.; Kim, J.E.; Lee, R.; Malberg, J.E.; Chen, J.; Steffen, C.; Zhang, Y.J.; Nestler, E.J.; Duman, R.S. Regulation of neurogenesis in adult mouse hippocampus by cAMP and the cAMP response element-binding protein. J. Neurosci. 2002, 22, 3673–3682. [Google Scholar] [CrossRef]
  93. Lee, B.H.; Kim, Y.K. BDNF mRNA expression of peripheral blood mononuclear cells was decreased in depressive patients who had or had not recently attempted suicide. J. Affect. Disord. 2010, 125, 369–373. [Google Scholar] [CrossRef] [PubMed]
  94. Fan, J.; Li, B.J.; Wang, X.F.; Zhong, L.L.; Cui, R.J. Ghrelin produces antidepressant-like effect in the estrogen deficient mice. Oncotarget 2017, 8, 58964–58973. [Google Scholar] [CrossRef]
  95. Lizama, C.; Lagos, C.F.; Lagos-Cabré, R.; Cantuarias, L.; Rivera, F.; Huenchuñir, P.; Pérez-Acle, T.; Carrión, F.; Moreno, R.D. Calpain inhibitors prevent p38 MAPK activation and germ cell apoptosis after heat stress in pubertal rat testes. J. Cell. Physiol. 2009, 221, 296–305. [Google Scholar] [CrossRef]
  96. Peng, Z.; Wang, H.; Zhang, R.; Chen, Y.; Xue, F.; Nie, H.; Chen, Y.; Wu, D.; Wang, Y.; Wang, H.; et al. Gastrodin ameliorates anxiety-like behaviors and inhibits IL-1beta level and p38 MAPK phosphorylation of hippocampus in the rat model of posttraumatic stress disorder. Physiol. Res. 2013, 62, 537–545. [Google Scholar] [CrossRef] [PubMed]
  97. Zoga, M.; Oulis, P.; Chatzipanagiotou, S.; Masdrakis, V.G.; Pliatsika, P.; Boufidou, F.; Foteli, S.; Soldatos, C.R.; Nikolaou, C.; Papageorgiou, C. Indoleamine 2,3-dioxygenase and immune changes under antidepressive treatment in major depression in females. In Vivo 2014, 28, 633–638. [Google Scholar]
  98. Zhou, W.; Dantzer, R.; Budac, D.P.; Walker, A.K.; Mao-Ying, Q.L.; Lee, A.W.; Heijnen, C.J.; Kavelaars, A. Peripheral indoleamine 2,3-dioxygenase 1 is required for comorbid depression-like behavior but does not contribute to neuropathic pain in mice. Brain Behav. Immun. 2015, 46, 147–153. [Google Scholar] [CrossRef]
  99. Cheng, Y.; Qiao, Z.; Dang, C.; Zhou, B.; Li, S.; Zhang, W.; Jiang, J.; Song, Y.; Zhang, J.; Diao, D. p38 predicts depression and poor outcome in esophageal cancer. Oncol. Lett. 2017, 14, 7241–7249. [Google Scholar] [CrossRef]
  100. Irusen, E.; Matthews, J.G.; Takahashi, A.; Barnes, P.J.; Chung, K.F.; Adcock, I.M. p38 Mitogen-activated protein kinase-induced glucocorticoid receptor phosphorylation reduces its activity: Role in steroid-insensitive asthma. J. Allergy Clin. Immunol. 2002, 109, 649–657. [Google Scholar] [CrossRef]
  101. Holsboer, F. The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 2000, 23, 477–501. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, C.; Huang, J.; Li, H.; Yang, Z.; Zeng, Y.; Liu, J.; Hao, Y.; Li, R. Ghrelin accelerates wound healing through GHS-R1a-mediated MAPK-NF-κB/GR signaling pathways in combined radiation and burn injury in rats. Sci. Rep. 2016, 6, 27499. [Google Scholar] [CrossRef] [PubMed]
  103. Han, Q.Q.; Huang, H.J.; Wang, Y.L.; Yang, L.; Pilot, A.; Zhu, X.C.; Yu, R.; Wang, J.; Chen, X.R.; Liu, Q.; et al. Ghrelin exhibited antidepressant and anxiolytic effect via the p38-MAPK signaling pathway in hippocampus. Prog. Neuropsychopharmacol. Biol. Psychiatry 2019, 93, 11–20. [Google Scholar] [CrossRef] [PubMed]
  104. Bruchas, M.R.; Schindler, A.G.; Shankar, H.; Messinger, D.I.; Miyatake, M.; Land, B.B.; Lemos, J.C.; Hagan, C.E.; Neumaier, J.F.; Quintana, A.; et al. Selective p38α MAPK deletion in serotonergic neurons produces stress resilience in models of depression and addiction. Neuron 2011, 71, 498–511. [Google Scholar] [CrossRef] [PubMed]
  105. Schoenfeld, T.J.; Cameron, H.A. Adult neurogenesis and mental illness. Neuropsychopharmacology 2015, 40, 113–128. [Google Scholar] [CrossRef] [PubMed]
  106. Peltier, J.; O’Neill, A.; Schaffer, D.V. PI3K/Akt and CREB regulate adult neural hippocampal progenitor proliferation and differentiation. Dev. Neurobiol. 2007, 67, 1348–1361. [Google Scholar] [CrossRef] [PubMed]
  107. Doble, B.W.; Woodgett, J.R. GSK-3: Tricks of the trade for a multi-tasking kinase. J. Cell Sci. 2003, 116, 1175–1186. [Google Scholar] [CrossRef] [PubMed]
  108. Hart, M.J.; de los Santos, R.; Albert, I.N.; Rubinfeld, B.; Polakis, P. Downregulation of beta-catenin by human Axin and its association with the APC tumor suppressor, beta-catenin and GSK3 beta. Curr. Biol. 1998, 8, 573–581. [Google Scholar] [CrossRef] [PubMed]
  109. Ryu, J.K.; Choi, H.B.; Hatori, K.; Heisel, R.L.; Pelech, S.L.; McLarnon, J.G.; Kim, S.U. Adenosine triphosphate induces proliferation of human neural stem cells: Role of calcium and p70 ribosomal protein S6 kinase. J. Neurosci. Res. 2003, 72, 352–362. [Google Scholar] [CrossRef] [PubMed]
  110. Chung, H.; Li, E.; Kim, Y.; Kim, S.; Park, S. Multiple signaling pathways mediate ghrelin-induced proliferation of hippocampal neural stem cells. J. Endocrinol. 2013, 218, 49–59. [Google Scholar] [CrossRef]
  111. Chung, H.; Seo, S.; Moon, M.; Park, S. Phosphatidylinositol-3-kinase/Akt/glycogen synthase kinase-3β and ERK1/2 pathways mediate protective effects of acylated and unacylated ghrelin against oxygen–glucose deprivation-induced apoptosis in primary rat cortical neuronal cells. J. Endocrinol. 2008, 198, 511–521. [Google Scholar] [CrossRef]
  112. Ferreira-Marques, M.; Carvalho, A.; Cavadas, C.; Aveleira, C.A. PI3K/AKT/MTOR and ERK1/2-MAPK signaling pathways are involved in autophagy stimulation induced by caloric restriction or caloric restriction mimetics in cortical neurons. Aging 2021, 13, 7872–7882. [Google Scholar] [CrossRef] [PubMed]
  113. Hou, Y.; Wang, K.; Wan, W.; Cheng, Y.; Pu, X.; Ye, X. Resveratrol provides neuroprotection by regulating the JAK2/STAT3/PI3K/AKT/mTOR pathway after stroke in rats. Genes Dis. 2018, 5, 245–255. [Google Scholar] [CrossRef]
  114. Zhu, H.; Jian, Z.; Zhong, Y.; Ye, Y.; Zhang, Y.; Hu, X.; Pu, B.; Gu, L.; Xiong, X. Janus Kinase Inhibition Ameliorates Ischemic Stroke Injury and Neuroinflammation through Reducing NLRP3 Inflammasome Activation via JAK2/STAT3 Pathway Inhibition. Front. Immunol. 2021, 12, 714943. [Google Scholar] [CrossRef]
  115. Zhang, W.; Xu, M.; Chen, F.; Su, Y.; Yu, M.; Xing, L.; Chang, Y.; Yan, T. Targeting the JAK2-STAT3 pathway to inhibit cGAS-STING activation improves neuronal senescence after ischemic stroke. Exp. Neurol. 2023, 368, 114474. [Google Scholar] [CrossRef] [PubMed]
  116. Ring, R.H.; Malberg, J.E.; Potestio, L.; Ping, J.; Boikess, S.; Luo, B.; Schechter, L.E.; Rizzo, S.; Rahman, Z.; Rosenzweig-Lipson, S. Anxiolytic-like activity of oxytocin in male mice: Behavioral and autonomic evidence, therapeutic implications. Psychopharmacology 2006, 185, 218–225. [Google Scholar] [CrossRef]
  117. Weissman, M.M.; Klerman, G.L. Sex differences and the epidemiology of depression. Arch. Gen. Psychiatry 1977, 34, 98–111. [Google Scholar] [CrossRef] [PubMed]
  118. Bleickardt, C.J.; Mullins, D.E.; Macsweeney, C.P.; Werner, B.J.; Pond, A.J.; Guzzi, M.F.; Martin, F.D.; Varty, G.B.; Hodgson, R.A. Characterization of the V1a antagonist, JNJ-17308616, in rodent models of anxiety-like behavior. Psychopharmacology 2009, 202, 711–718. [Google Scholar] [CrossRef]
  119. Serradeil-Le Gal, C.; Wagnon, J., 3rd; Tonnerre, B.; Roux, R.; Garcia, G.; Griebel, G.; Aulombard, A. An overview of SSR149415, a selective nonpeptide vasopressin V(1b) receptor antagonist for the treatment of stress-related disorders. CNS Drug Rev. 2005, 11, 53–68. [Google Scholar] [CrossRef]
  120. Lutter, M.; Sakata, I.; Osborne-Lawrence, S.; Rovinsky, S.A.; Anderson, J.G.; Jung, S.; Birnbaum, S.; Yanagisawa, M.; Elmquist, J.K.; Nestler, E.J.; et al. The orexigenic hormone ghrelin defends against depressive symptoms of chronic stress. Nat. Neurosci. 2008, 11, 752–753. [Google Scholar] [CrossRef]
  121. Zhang, Y.; Zhu, M.Z.; Qin, X.H.; Zeng, Y.N.; Zhu, X.H. The Ghrelin/Growth Hormone Secretagogue Receptor System Is Involved in the Rapid and Sustained Antidepressant-like Effect of Paeoniflorin. Front. Neurosci. 2021, 15, 631424. [Google Scholar] [CrossRef] [PubMed]
  122. Wu, R.; Xiao, D.; Shan, X.; Dong, Y.; Tao, W.W. Rapid and Prolonged Antidepressant-like Effect of Crocin Is Associated with GHSR-Mediated Hippocampal Plasticity-related Proteins in Mice Exposed to Prenatal Stress. ACS Chem. Neurosci. 2020, 11, 1159–1170. [Google Scholar] [CrossRef] [PubMed]
  123. Rana, T.; Behl, T.; Sehgal, A.; Singh, S.; Sharma, N.; Abdeen, A.; Ibrahim, S.F.; Mani, V.; Iqbal, M.S.; Bhatia, S.; et al. Exploring the role of neuropeptides in depression and anxiety. Prog. Neuropsychopharmacol. Biol. Psychiatry 2022, 114, 110478. [Google Scholar] [CrossRef] [PubMed]
  124. Ge, X.; Yang, H.; Bednarek, M.A.; Galon-Tilleman, H.; Chen, P.; Chen, M.; Lichtman, J.S.; Wang, Y.; Dalmas, O.; Yin, Y.; et al. LEAP2 Is an Endogenous Antagonist of the Ghrelin Receptor. Cell Metab. 2018, 27, 461–469.e6. [Google Scholar] [CrossRef] [PubMed]
  125. Céline, M.K.; Agustina, C.; Franco, B.; Julien, G.; Sonia, C.; Marjorie, D.; Sophie, M.; Séverine, D.; Sébastien, D.; Sylvie, P.-R.; et al. N-Terminal Liver-Expressed Antimicrobial Peptide 2 (LEAP2) Region Exhibits Inverse Agonist Activity toward the Ghrelin Receptor. J. Med. Chem. 2018, 62, 965–973. [Google Scholar] [CrossRef]
  126. Jackson, T.M.; Ostrowski, T.D.; Middlemas, D.S. Intracerebroventricular Ghrelin Administration Increases Depressive-like Behavior in Male Juvenile Rats. Front. Behav. Neurosci. 2019, 13, 77. [Google Scholar] [CrossRef] [PubMed]
  127. Hansson, C.; Haage, D.; Taube, M.; Egecioglu, E.; Salomé, N.; Dickson, S.L. Central administration of ghrelin alters emotional responses in rats: Behavioural, electrophysiological and molecular evidence. Neuroscience 2011, 180, 201–211. [Google Scholar] [CrossRef]
  128. Jensen, M.; Ratner, C.; Rudenko, O.; Christiansen, S.H.; Skov, L.J.; Hundahl, C.; Woldbye, D.P.; Holst, B. Anxiolytic-like Effects of Increased Ghrelin Receptor Signaling in the Amygdala. Int. J. Neuropsychopharmacol. 2016, 19, pyv123. [Google Scholar] [CrossRef] [PubMed]
  129. Mahbod, P.; Smith, E.P.; Fitzgerald, M.E.; Morano, R.L.; Packard, B.A.; Ghosal, S.; Scheimann, J.R.; Perez-Tilve, D.; Herman, J.P.; Tong, J. Desacyl Ghrelin Decreases Anxiety-like Behavior in Male Mice. Endocrinology 2018, 159, 388–399. [Google Scholar] [CrossRef]
  130. Pawar, G.R.; Agrawal, Y.O.; Nakhate, K.T.; Patil, C.R.; Sharma, C.; Ojha, S.; Mahajan, U.B.; Goyal, S.N. Ghrelin alleviates depression-like behaviour in rats subjected to high-fat diet and diurnal rhythm disturbance. Am. J. Transl. Res. 2022, 14, 7098–7108. [Google Scholar]
  131. Sun, N.; Mei, Y.; Hu, Z.; Xing, W.; Lv, K.; Hu, N.; Zhang, T.; Wang, D. Ghrelin attenuates depressive-like behavior, heart failure, and neuroinflammation in postmyocardial infarction rat model. Eur. J. Pharmacol. 2021, 901, 174096. [Google Scholar] [CrossRef] [PubMed]
  132. Huang, H.J.; Chen, X.R.; Han, Q.Q.; Wang, J.; Pilot, A.; Yu, R.; Liu, Q.; Li, B.; Wu, G.C.; Wang, Y.Q.; et al. The protective effects of Ghrelin/GHSR on hippocampal neurogenesis in CUMS mice. Neuropharmacology 2019, 155, 31–43. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The antidepressant potential of ghrelin, which exerts neuroprotective effects by promoting neuronal proliferation and neurotrophic factor production in the brain. Ghrelin can inhibit apoptosis, reduce inflammation, increase astrocyte production, and significantly inhibit the process of depression. Ghrelin interacts with GHSR1a to increase the proliferation of hippocampal NSCs by increasing the expression of the Jak2/STAT3 signaling pathway, which also indirectly upregulates the PI3K/Akt/mTOR pathway to jointly exert neuronal protective effects. In addition, ghrelin binding to its receptor can also increase the expression of the PI3K/Akt signaling pathway, play a pro-neural cell proliferation role by promoting the phosphorylation of downstream molecules (mTOR and p70S6K), and inactivate GSK-3β by promoting GSK-3β phosphorylation, which can further increase the nuclear translocation of β-catenin, thus decreasing cellular apoptosis, playing a role in promoting cell survival. The activation of this pathway also promotes the proliferation of astrocytes and the communication of neurons in the brain. Moreover, ghrelin treatment can inhibit the p38-MAPK signaling pathway, increase GR expression, and inhibit glucocorticoid resistance in depressed patients. Additionally, ghrelin increases CREB phosphorylation by promoting the expression of the cAMP/CREB signaling pathway, thus regulating BDNF transcription and eventually exerting neuroprotective effects to inhibit depression. Besides this, ghrelin levels increase in the regulation of inflammatory response; further, this hormone downregulates neutrophil transport and the amount of pro-inflammatory cytokines, thus reducing inflammation, and it also increases the expression of BDNF and improves neurobehavioral function. In the figure, the subtraction symbol indicates inhibitory/suppressing action, while the addition symbol indicates stimulatory action. The P labeled in the circle represents substrate phosphorylation. ↑, increase; ↓, decrease.
Figure 1. The antidepressant potential of ghrelin, which exerts neuroprotective effects by promoting neuronal proliferation and neurotrophic factor production in the brain. Ghrelin can inhibit apoptosis, reduce inflammation, increase astrocyte production, and significantly inhibit the process of depression. Ghrelin interacts with GHSR1a to increase the proliferation of hippocampal NSCs by increasing the expression of the Jak2/STAT3 signaling pathway, which also indirectly upregulates the PI3K/Akt/mTOR pathway to jointly exert neuronal protective effects. In addition, ghrelin binding to its receptor can also increase the expression of the PI3K/Akt signaling pathway, play a pro-neural cell proliferation role by promoting the phosphorylation of downstream molecules (mTOR and p70S6K), and inactivate GSK-3β by promoting GSK-3β phosphorylation, which can further increase the nuclear translocation of β-catenin, thus decreasing cellular apoptosis, playing a role in promoting cell survival. The activation of this pathway also promotes the proliferation of astrocytes and the communication of neurons in the brain. Moreover, ghrelin treatment can inhibit the p38-MAPK signaling pathway, increase GR expression, and inhibit glucocorticoid resistance in depressed patients. Additionally, ghrelin increases CREB phosphorylation by promoting the expression of the cAMP/CREB signaling pathway, thus regulating BDNF transcription and eventually exerting neuroprotective effects to inhibit depression. Besides this, ghrelin levels increase in the regulation of inflammatory response; further, this hormone downregulates neutrophil transport and the amount of pro-inflammatory cytokines, thus reducing inflammation, and it also increases the expression of BDNF and improves neurobehavioral function. In the figure, the subtraction symbol indicates inhibitory/suppressing action, while the addition symbol indicates stimulatory action. The P labeled in the circle represents substrate phosphorylation. ↑, increase; ↓, decrease.
Cimb 46 00434 g001
Table 1. Effects of ghrelin/GHSR system on MDD in rodents.
Table 1. Effects of ghrelin/GHSR system on MDD in rodents.
Animal and Stress ParadigmBehavioral TestInterventionSignal MoleculesEffectsReferences
Mouse CSDSFST and TSTGHSR1a knock-outBDNF ↓, IL-6 ↑Pro-depression effect[26]
RatFST and TSTi.c.v. injection of ghrelinHPA ↑Immobility time ↑ in TST[126,127]
Male C57BL/J6 mice, RSTST,
OFT, and
FST
rAAV-Mediated overexpression of GHSR1ac-Fos ↑Antidepressant-like effect[128]
Male mice, restraint stressEPMGhrelin KOpERK ↓Decreases anxiety-like
behavior
[129]
C57BL/J6 mice, prenatal stressOFT, TST,
FST, and SPT
i.p. injection of crocinPI3K/Akt ↑
MTOR ↑
Antidepressant-like effect[122]
Male SD rats, HFD and DDRFST,
OFT, and
EPM
Intra-VTA administration of ghrelinTNF-α ↓
IL-1β↓ IL-6 ↓
Alleviates depression-like behavior[130]
Male SD rats, CAOSPT, OFT, and
EPM
Subcutaneous injection of ghrelinIba-1 ↓
GFAP ↓
Attenuates depression-like behavior[131]
Male C57BL/J6 mice, CSDSSIT, FST, OFT, and
EPM
Intrahippocampal ghrelin infusions,
AAV-siRNA of GHSR1a
p38-MAPK ↓Antidepressant effect[103]
Male C57BL/J6 miceTST and
FST
Lateral ventricle injection of ghrelinCREB ↑
BDNF ↑
Improves cognition and
antidepressant-like effects
[24,94]
Adult female mice, OBOFT and
TST
Ghrelin into the hippocampusMAPK ↑
CaMKIIa ↑
Antidepressant-like effects[25]
Male C57BL/J6 mice, CUMSOFT,
EPM, and
FST
Intraperitoneal (i.p.) injection of ghrelin, GHSR knock-downPI3K/Akt ↑
CDK2 ↑
CyclinD1 ↑
Spine density ↑,
proliferation of hippocampal NSCs, and
neurogenesis ↑
[132]
The upward arrow indicates increase; The downward arrow indicates decrease.
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

Pan, X.; Gao, Y.; Guan, K.; Chen, J.; Ji, B. Ghrelin/GHSR System in Depressive Disorder: Pathologic Roles and Therapeutic Implications. Curr. Issues Mol. Biol. 2024, 46, 7324-7338. https://doi.org/10.3390/cimb46070434

AMA Style

Pan X, Gao Y, Guan K, Chen J, Ji B. Ghrelin/GHSR System in Depressive Disorder: Pathologic Roles and Therapeutic Implications. Current Issues in Molecular Biology. 2024; 46(7):7324-7338. https://doi.org/10.3390/cimb46070434

Chicago/Turabian Style

Pan, Xingli, Yuxin Gao, Kaifu Guan, Jing Chen, and Bingyuan Ji. 2024. "Ghrelin/GHSR System in Depressive Disorder: Pathologic Roles and Therapeutic Implications" Current Issues in Molecular Biology 46, no. 7: 7324-7338. https://doi.org/10.3390/cimb46070434

Article Metrics

Back to TopTop