Next Article in Journal
Comprehensive Evaluation of the Quality of Tripterygium Glycosides Tablets Based on Multi-Component Quantification Combined with an In Vitro Biological Assay
Next Article in Special Issue
Curcumin-Based Nanoformulations: A Promising Adjuvant towards Cancer Treatment
Previous Article in Journal
Tetrahydrocurcumin-Related Vascular Protection: An Overview of the Findings from Animal Disease Models
Previous Article in Special Issue
Natural Polyphenols as SERCA Activators: Role in the Endoplasmic Reticulum Stress-Related Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiology of Depression: Stingless Bee Honey Promising as an Antidepressant

by
Fatin Haniza Zakaria
1,
Ismail Samhani
2,
Mohd Zulkifli Mustafa
1,* and
Nazlahshaniza Shafin
3,*
1
Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
2
Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Medical Campus, Jalan Sultan Mahmud, Kuala Terengganu 20400, Malaysia
3
Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kota Bharu 16150, Malaysia
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(16), 5091; https://doi.org/10.3390/molecules27165091
Submission received: 8 July 2022 / Revised: 30 July 2022 / Accepted: 6 August 2022 / Published: 10 August 2022

Abstract

:
Depression is a debilitating psychiatric disorder impacting an individual’s quality of life. It is the most prevalent mental illness across all age categories, incurring huge socio-economic impacts. Most depression treatments currently focus on the elevation of neurotransmitters according to the monoamine hypothesis. Conventional treatments include tricyclic antidepressants (TCAs), norepinephrine–dopamine reuptake inhibitors (NDRIs), monoamine oxidase inhibitors (MAOIs), and serotonin reuptake inhibitors (SSRIs). Despite numerous pharmacological strategies utilising conventional drugs, the discovery of alternative medicines from natural products is a must for safer and beneficial brain supplement. About 30% of patients have been reported to show resistance to drug treatments coupled with functional impairment, poor quality of life, and suicidal ideation with a high relapse rate. Hence, there is an urgency for novel discoveries of safer and highly effective depression treatments. Stingless bee honey (SBH) has been proven to contain a high level of antioxidants compared to other types of honey. This is a comprehensive review of the potential use of SBH as a new candidate for antidepressants from the perspective of the monoamine, inflammatory and neurotrophin hypotheses.

Graphical Abstract

1. Revisiting Depression

Depression is a psychiatric disorder characterized by psychological, behavioral and physiological symptoms that include a persistent low mood, marked loss of pleasure in most activities, poor concentration, disruptions in appetite and sleeping patterns, cognitive impairments, feelings of worthlessness, excessive guilt, and suicidal thoughts [1]. It is the leading cause of disability worldwide that poses a high emotional and financial burden [2,3]. The World Health Organization (WHO) estimated that depression will be declared a global burden by the year 2030 [4] affecting an estimation of 300 million people from all age categories [5].
Depression covers various subtypes and etiologies [6] from monoamines to inflammatory and neurotrophic propositions. In the 1960s, the “catecholamine hypothesis” appeared as a popular monoamine hypothesis for explaining depression development. It suggested that serotonin (5HT) deficiency and noradrenaline (NA) creates depression [7,8,9]. The inflammatory hypothesis proposes that depression is caused by the interaction of inflammatory cytokine with the hypothalamic–pituitary–adrenal (HPA) axis, consequently affecting the synthesis and reuptake of neurotransmitters [10,11], which subsequently triggers glucocorticoid resistance, glutamate excitotoxicity, and the reduction of brain-derived neurotrophic factor (BDNF) expression [12].
Since BDNF is reduced in the onset of depression, the neurotrophic hypothesis has become one of the critical etiologies of antidepressant progression. This hypothesis states that neurotrophic factors are essential to the development of neurons by promoting synaptic growth and maintaining neuronal survival. They play a crucial role in neuronal network formation and plasticity. On the contrary, the reduction of neurotrophic factors is implicated in the atrophy of stress-vulnerable hippocampal neurons, such as depression and cognitive disorder [13]. This deficiency is believed to be reversed by antidepressant treatments that contribute to the resolution of depressive symptoms [14].
Since the number of depression cases is increasing day by day, the discovery of new treatments is imperative. At present, there is a vibrant demand for new treatment strategies since the flaws of conventional treatments are striking. For instance, many sources purport that antidepressants have a therapeutic delay onset, taking weeks rather than days to become effective [15,16]. Prolonged exposure to antidepressant drugs imposes susceptibility to adverse side effects, such as interferences in sexual functioning, gastrointestinal disturbances, altered sleep pattern, and weight gain [17,18,19,20,21,22]. Moreover, 30% of patients have been reported to be non-compliant with currently available treatments [23,24,25]. Thus, there is a dire need for the development of new antidepressant treatments with better efficacy and that are safer for patients [26].
This has caused an urgent call for complementary and alternative medicines in treating depression [27]. Honey, which contains a variety of active compounds beneficial to brain regulation and treats emotional and psychological disorders including depression [28,29,30] is one of the natural products serving as an alternative medicine [31]. Among the various types of honey, here we focus on stingless bee honey (SBH). In Malaysia, SBH is well known as “madu kelulut” [32]. In addition to SBH, there are other honeys capable of treating several health problems named Tualang and Manuka [33,34]. However, in terms of nutritional composition, SBH contains a higher level of polyphenol [35,36,37], an important active compound that participates in modulating signaling pathways, thus influencing neuronal survival and cell regeneration and development, which suffer detrimental effects after injury [38,39]. To date, there are limited studies highlighting the potential of SBH as an alternative supplement to treat depression. Therefore, this review discusses the different hypotheses associated with depression and how SBH’s mechanism of action could act as a potential antidepressant as a brain supplement. We highlight the different types of etiology hypotheses in the pathophysiology of depression followed by its mechanism of action.

2. Pathophysiology of Depression

2.1. Monoamine Hypothesis

Depression is a well-known psychiatric disorder that involves the dysregulation of the monoamine system that leads to an imbalance of neurotransmitters, such as 5HT, dopamine, and NA [40,41]. Monoamines are molecules involved in information transmission processes by connecting presynaptic to postsynaptic neurons [42]. They are classified according to their chemical structure and mechanism of action [43]. Since they have different chemical structures, every monoamine is specific to its respective receptors [42] and has a different function in the brain [40,44,45]. For example, 5HT is a central nervous system monoamine that has a crucial role in regulating appetite, circadian cycle, anxiety, memory, and learning. In addition to 5HT, dopamine is another important monoamine that fuels motivation and modulates pleasure, reward, and emotion. In addition, NA is another essential monoamine responsible for attentiveness, emotions, cognition, and social interactions.
The monoamine hypothesis was formulated in the mid-1960s due to the underactivity of brain monoamines such as serotonin, dopamine, and NA in patients’ brains [46]. This hypothesis is based on antidepressant drug efficacy, such as selective serotonin reuptake inhibitors (SSRIs), norepinephrine–dopamine reuptake inhibitors (NDRIs), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs) [47,48]. The mechanisms of action for this hypothesis with antidepressants are: (1) inhibition of the reuptake of 5HT and/or NA; (2) antagonistic presynaptic inhibition of 5HT and/or NA; and (3) inhibition of monoamine oxidase (MAO) [45]. Findings on these mechanisms of action showed that chronic treatment with antidepressants ultimately causes increased levels of monoamines.
Apart from 5HT, dopamine, and NA, γ-aminobutyric acid (GABA) is also reported to affect depression [49,50,51,52]. GABA plays a role in depression and anxiety through its interaction with inflammatory cytokines, NF-kB, and p38 MAPK signaling pathways [53].

2.2. Inflammation Hypothesis in Depression

The inflammation theory has also been linked to depression, which surprises many people. It acts as a key point regarding treatment direction for depression cases. Believed to be fueled by lifestyle, the inflammatory process is related to the nuclear factor-κB (NF-κB) pathway [54], a transcriptional factor that regulates various gene expressions. It is activated by extracellular stimuli, such as lipopolysaccharide (LPS), administration, or chronic stress [55,56,57,58], giving it a propensity to go haywire. Once it is activated, an inflammatory response takes place [59,60].
An inflammatory response includes the secretion of cytokines, which have a specific effect on the interactions and communications between cells [61]. Cytokines are signaling proteins secreted in response to the immune system’s activation by stressors, such as injury, infection, or psychosocial factors [62]. Moreover, the cytokines induce anti- or pro-inflammatory responses, whereby the anti-inflammatory cytokines are secreted to counteract the pro-inflammatory cytokines [63,64]. Cytokines comprise lymphokine (cytokine made by lymphocytes), monokine (cytokine made by monocytes), chemokine (cytokines with chemotactic activities), and interleukin (cytokines made by one leukocyte and acting on other leukocytes). Part of them is recognized as IL-2, IFN-γ, IL-1β, TNF-α, IL-6, IL-12, IL-15 for pro-inflammatory functions [65,66,67] and as IL-4, IL-5, IL-13, IL-1Ra, IL-10 for anti-inflammatory action [68].
Inflammatory responses play a primary role in eliminating or inactivating harmful entities or damaged tissues in the body. However, the over-activation of this system can cause detrimental effects, such as depressive-like behavior [69,70]. Previous studies have shown that depressed people have increased levels of inflammatory mediators, such as C-reactive protein (CRP) and pro-inflammatory cytokines [71,72]. In response to inflammation, the translocation of inflammatory mediators interferes with neuronal and glial well-being, resulting in cognitive and behavioral manifestations, and synaptic plasticity that leads to neurodegeneration [73].
There are two major pathways for inflammatory cytokines that disrupt the synthesis of monoamine neurotransmitters, particularly 5HT, glutamate, and dopamine, as shown in Figure 1.
They are important for neurotransmitter regulation and ultimately affect mood regulation in depression, namely kynurenine and tetrahydrobiopterin (BH4) [66,74,75]. Activation of the kynurenine pathway (KP) within areas of the brain, such as the hippocampus, has been shown to cause alterations in emotional behaviors [76,77,78]. This is because KP affects the most important neurotransmitter for the regulation of emotion, which is 5HT [79]. When inflammation occurs, levels of indoleamine 2, 3-dioxygenase (IDO) and tryptophan 2, 3-dioxygenase (TDO) are elevated and the tryptophan is used by the IDO and TDO in kynurenine production [80]. This eventually causes the depletion of the tryptophan level for 5HT production. This has been proven in animal models and drug therapy patients with interferon-α [81,82]. IDO and TDO are induced by pro-inflammatory cytokines, such as IL-1, IL-2, IL-6 and IFN-γ [80]. KP causes the increased production of several harmful metabolites, such as 3-hydroxykynurenine (3HK) and quinolinic acid (QA), causing the over-activation of the N-methyl-D-aspartate (NMDA) receptor and inducing oxidative stress and kynurenic acid [83]. The link between inflammation and KP is evident through the increased number of astrocytes that are synthesized by kynurenic acid and the increased production of quinolinic acid by microglia [79]. Alongside kynurenine, the tetrahydrobiopterin (BH4) pathway is also significant due to the monoamine neurotransmitter synthesis that is disrupted in depression [67]. Analyzed SBH sample identified compounds such as phenylalanine, alanine, tyrosine, valine, acetate, lactate, trigonelline, ethanol metabolites, glucose, fructose, sucrose, and maltose [84]. Phenylalanine, which is consistently found in SBH, converted to tyrosine, which simultaneously converts BH4 to 4a-Hydroxytetrahydrobiopterin and is catalyzed by phenylalanine hydroxylase [85]. BH4 is a cofactor for precursors of neurotransmitters, namely 5HT, dopamine, and NA [75]. For example, the serotonergic pathway biosynthesis of 5HT comes from tryptophan, whereas dopaminergic, noradrenergic, and adrenergic pathways are intermediated by the precursor L-3,4-dihydroxyphenylalanine (L-DOPA) for the synthesis of dopamine, adrenaline, and NA [86,87]. Inflammatory cytokines can disrupt BH4 production, which is crucial for neurotransmitter synthesis [67]. There are two mechanisms that are involved in the disruption of BH4. Firstly, inflammatory cytokines stimulate NOS to produce NO. The elevated activity of NOS causes the increased utilization of BH4 that will be converted to 7, 8-dihydrobiopterin (BH2).
The conversion of arginine to nitric oxide (NO) by nitric oxide synthase (NOS) is enhanced by BH4, which acts as an enzyme co-factor [88]. Furthermore, BH4 is very sensitive to oxidative stress. Inflammatory cytokines are known to increase oxidative stress through the production of both nitrogen and oxygen-free radicals. This causes the irreversible degradation of BH4 to dihydroxyanthopterin [89].

2.3. Neurotrophin Hypothesis

In addition to the monoamine and inflammatory hypothesis, the neurotrophin hypothesis also has a vital role in the pathophysiology of depression [90]. Neurotrophin is a type of protein that is essential for the growth, survival, and differentiation of neurons [91,92]. Four types of neurotrophins are important in mammals, namely brain-derived neurotrophin factor (BDNF), nerve growth factor (NGF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4) [93]. BDNF is critical in the central nervous system (CNS) for neurogenesis, synaptic plasticity, development, survival, and neuron maintenance [94,95,96,97]. BDNF is an example of a neurotrophin that has an impact on the pathophysiology of depression [13,97,98]. BDNF and its receptor tropomyosin receptor kinase B (TrKB) are involved in different intracellular signaling pathways, such as mitogen-activated protein kinase/extracellular signal-regulated protein kinase (MAPK/ERK), phospholipase Cγ (PLCγ), and phosphoinositide 3-kinase (PI3K) [99]. These pathways have a biological impact on the central nervous system, such as on memory and mood regulation [95,100,101]. ERK is one of the downstream BDNF pathways that is implicated in the regulation of mood and behavior in the depression model that mediates the effects of antidepressants [102,103,104,105]. Meanwhile, PI3K signaling is an important component of long-term potentiation (LTP) [106]. This signaling pathway acts as a biochemical cascade for α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) for synaptic plasticity, thus causing behavioral alteration [107]. Moreover, all the intracellular signaling pathways that were mentioned earlier have been discussed in previous studies that are related to depression. Changes in BDNF levels in the central nervous system disrupt the entire signaling pathway, which can lead to various psychological disorders, including depression [108,109,110,111,112].
BDNF promotes neurogenesis, which is part of neuroplasticity. Neuroplasticity involves changes or alterations in the structure, functions, and connections of the central nervous system (CNS) in response to intrinsic or extrinsic stimuli [113]. These changes include the morphology of mature neurons, such as axonal and dendritic arborization and pruning, increased spine density, and synaptogenesis [114]. Neurogenesis is defined as the formation of newborn neurons in proliferative areas that include the subventricular zone (SVZ) and the subgranular zone (SGZ) of the dentate gyrus region in the hippocampus area [114,115]. This region is crucial for memory, learning, and other cognitive functions [116]. The alteration of BDNF levels is known to be detrimental to neurogenesis in the hippocampus, specifically in the dentate gyrus region [117]. The dentate gyrus is a region in the hippocampus that is widely discussed in depression [118,119,120]. Based on previous studies, BDNF levels in the hippocampus and prefrontal cortex are reduced in cases of depression [13,121,122]. This consequently resulted in the decreased size of the hippocampal area in the brain in both clinical and preclinical studies of depression [118,119,120]. Similarly, the condition is observed in the prefrontal cortex [118,123,124] causing neuronal loss and synaptic dysfunction in cortical limbic regions that ultimately disrupt mood and emotions [73].
In addition to neurogenesis as a part of neuroplasticity related to BDNF in the brain, synaptic plasticity is also associated with depression [41,125,126]. Synaptic plasticity is essential for the physiological morphology of neurons, and BDNF is one of the crucial regulators in this process making it a therapeutic target in depression [127]. Therefore, the BDNF level is vulnerable to synaptic plasticity in the brain. Long-term potentiation (LTP) is the main mechanism that mediates neuroplasticity at a functional level; synaptic strength is crucial for the connection between neurons in the brain [113]. BDNF facilitated LTP in the Schaffer collaterals of the hippocampus in a young animal model by inducing the release of presynaptic neurotransmitters [128]. Furthermore, the postsynaptic release of BDNF induces LTP in the dentate gyrus [129]. Increased hippocampal dendritic spine by LTP is contributed by BDNF signaling together with local protein translation [130]. Based on previous studies, patients showed decreased hippocampal volume and BDNF expression during depressive episodes compared to patients in remission, which altered synaptic plasticity by elevating hippocampal dendritic atrophy and cell death contributing to the decline of LTP [122,131,132]. These features have also been observed in rodents [133,134,135]. Moreover, the reduction of LTP caused by depression has been observed especially in the hippocampus and the prefrontal brain area [73,136,137]. The same effect has been observed in long-term depression (LTD) [138].

3. Stingless Bee Honey (SBH) as an Antidepressant

Earlier in this review, the etiology of pathological depression was discussed briefly. In this section, the role of SBH as a prophylactic against depression is reviewed. The focus will be on certain properties of SBH that are related to depression, which include amino acid (phenylalanine), antioxidant properties, and anti-inflammatory effects.

3.1. Neurotrophic Factors

The complex biological properties of SBH consist of amino acids (phenylalanine, alanine, tyrosine, and valine), phenolic compounds, carbohydrates, organic acids, vitamins, minerals, lipids, and enzymes [139,140,141,142,143]. They have potential roles in the regulation of signaling pathways in depression development. The amino acid that is highlighted in this review is phenylalanine. Phenylalanine is an essential amino acid that needs to be ingested through diet since it is not naturally synthesized by the body [144]. It is an important amino acid for the synthesis of neurotransmitters and a precursor for dopamine and NA [87]. This can be related to the role of SBH as a prophylactic against depression, which is in line with the monoamine hypothesis. The monoamine hypothesis, which was explained earlier, stated low levels of neurotransmitters in depressed patients as well as in animal studies. Furthermore, the role of NA has been emphasized in attenuating microglial activation in the brain, thus inhibiting pro-inflammatory cytokines (inflammatory hypothesis) as well as enhancing the production of neurotrophic factors (neurotrophin hypothesis) for neurogenesis in the brain [145]. Moreover, neurotransmitters have been reported to enhance BDNF release in the brain [146]. Therefore, neurotransmitters and BDNF have a bilateral effect that can ameliorate depressive behavior. This indicates that the administration of SBH during depressive episodes could regulate the deficiency of neurotransmitters as well as BDNF, which is important to the neurological process. The regulation of the neurological process during depressive episodes would impede depressive behavior symptoms, such as sickness behavior, loss of motivation, and anhedonia [147,148,149].

3.2. Antioxidant

Stingless bee honey (SBH) has high levels of phenolic compounds compared to other honey [36,37,150]. Examples of phenolic acids in SBH are p-coumaric acid, gallic acid, caffeic acid, chrysin, and apigenin [28,150,151,152]. Antioxidant properties in honey have been reported to strongly correlate with phenolic compounds [153,154,155]. The color intensity of honey is also an indicator of antioxidant activity due to the presence of pigments such as carotenoids and flavonoids [156]. According to Kek and colleagues (2014), SBH has higher color intensity compared to Tualang, Gelam, Pineapple, Borneo, and commercial honey [36]. Antioxidants are important as scavengers in preventing oxidative stress that leads to DNA damage [157,158]. The brain is a highly susceptible organ to the elevation of oxidative stress due to its high oxygen demand [48,159,160]. Several researchers have reported the neuroprotective effect that resulted from the polyphenol content in honey [29]. These studies support SBH as a potential antidepressant since depression is also related to oxidative stress within the brain [41,161,162]. According to a study by Czarny and colleagues (2018), depressed patients had elevated reactive oxygen species (ROS) and nitrogen species (RNS) from oxidative DNA damage after depressive episodes [163].
Moreover, the relationship between oxidative stress and depression has been reviewed and discussed relating to its usage as a natural compound with antioxidant properties as a constituent of their polyphenols that can alleviate depression [41,164]. For example, antioxidant activity by p-coumaric acid has been identified in animal models of depression [48]. It has also been reported to show a neuroprotective response through increased levels of glutathione and superoxide dismutase that subsequently reduce oxidative stress capacity and neurotoxicity [165,166,167]. In addition to p-coumaric acid, chrysin also exhibited oxidative stress in the preclinical model of depression [168,169]. The properties of phenolic compounds proven to show efficacy based on both animal and human studies are displayed in Table 1.
The antioxidant activity in SBH can act as ROS scavengers that can regulate the mechanism of action to improve depression in patients. Antioxidants can cause prophylactic effects in depression since elevated ROS have been reported to affect BH4. In the inflammatory hypothesis, BH4 is briefly discussed as a cofactor for the biosynthesis of neurotransmitters 5HT, dopamine, and NA. The regulation of BH4 after the administration of SBH regulates neurotransmitters that consequently enhance mood, emotion, and behaviors after depressive episodes. Neurotransmitters are known for their role as modulators in brain activity. In addition, SBH possesses high intensity in terms of color pigment indicating the presence of flavonoids. Flavonoids found in honey activate the ERK and protein kinase B (PKB/Akt), leading to the activation of cAMP response element-binding protein (CREB), a transcription factor responsible for increasing BDNF expression [161]. Specifically, a gene expression analysis showed that BDNF and Itpr1 were affected following SBH treatments, and the results indicate that supplementation with SBH leads to specific upregulations of the gene expression tested and leads to an improvement in the depressive state [84]. There are also reports about the use of flavonoids in neurodegenerative disorders to regulate BDNF [189,190,191,192]. This evidence strongly suggests that SBH, which contains a high number of flavonoids, has the ability to enhance BDNF and regulate its levels as a determinant for antidepressant efficacy [91,93,193,194].

3.3. Anti-Inflammatory

Stingless bee honey (SBH) has been reported to possess anti-inflammatory properties [150,195]. According to a study by Ranneh and colleagues (2019), SBH has an anti-inflammatory effect in the LPS animal model. The etiology of depression has been discussed in depth through the inflammatory hypothesis perspective, especially with regard to how inflammation affects depression. Inflammation is known to activate the kynurenine pathway in depression [196]. This pathway causes cascade effects through the elevation of ROS and glutamate toxicity as well as a reduction of 5HT and BDNF [197,198,199]. Due to its anti-inflammatory properties, SBH can also regulate BDNF, hence alleviating depression symptoms. This is also essential due to its bilateral effect on the regulation of monoamine systems, which is important in ameliorating symptoms of depression [14,200].
The immune system is very sensitive to oxidative stress and with moderate exercise, immune functionality can be enhanced [48]. Exercise also has been recognized as a useful non-pharmacological strategy to improve the treatment of depression [201,202]. Relaxation responses significantly reduced the neuropsychological scores tested, decreased cortisol, decreased the trend of NGF, and increased BDNF levels [201]. BDNF binds to the tyrosine kinase β receptor (TrKβ) and activates the phosphoinositide 3-kinase and Akt pathway, which inhibits the activity of glycogen synthase kinase-3 beta (GSK-3β) [202,203]. GSK-3β activity cleaves cadherin–β-catenin binding; therefore, GSK-3β inhibition stabilizes β-catenin, which regulates gene expression, synaptic plasticity, and neurogenesis, which in turn has antidepressant effects. A study using flavonoids found they seem to exert additional positive effects with exercise, where a combination of quercetin and exercise training exerted potent anti-tumor and anti-depressive effects through the suppression of inflammation and the upregulation of the BDNF/TrKβ/β-catenin axis in the prefrontal cortex of 1,2-dimethylhydrazine (DMH)-induced colorectal cancer-induced rats [202]. Thus, it is speculated that flavonoids in SBH express anti-inflammatory properties and are involved in the BDNF/ TrKβ pathway; however, the specification of active possible chemotherapeutic modality needs further investigation. This statement supports the potential of SBH as an antidepressant due to its anti-inflammatory properties.
Recent studies on treatment resistance in depressed patients have related the condition to inflammation. Patients with treatment-resistant depression (TRD) have been reported to show dysregulated inflammatory activity compared to non-TRD patients [204,205]. They also exhibited increased levels of inflammatory cytokines (IL-6, IL8, TNF-α, CRP, and macrophage inflammatory protein-1 (MIP)-1 alpha) that resulted in poorer treatment outcomes [206]. Inflammatory cytokines are known as critical mediators in the inflammatory response that disrupt the signaling pathways or mechanisms of action of conventional antidepressants [204]. Therefore, anti-inflammatory treatments might be effective in preventing TRD in patients [207].

4. Conclusions

In conclusion, stingless bee honey could regulate the detrimental effects during or after depressive episodes that can lead to prophylactic effects. This review summarized how the amino acid (phenylalanine), antioxidant, and anti-inflammatory properties of SBH have the potential to control depression symptoms according to the etiology of depression—the monoamine, neurotrophin, and inflammatory hypotheses—through neurotrophic factors and its antioxidant and anti-inflammatory properties as shown in Figure 2.
Our review reports the possible mechanisms of stingless bee honey pertaining to its antioxidant and anti-inflammatory properties, which contribute to its antidepressant properties. Follow-up studies are still required to comprehensively analyze the bioactive compound of SBH responsible for the underlying mechanisms as well as to investigate other possible pathways contributing to its anti-depressive effects.

Author Contributions

F.H.Z. and I.S. contributed to preparing the manuscript. M.Z.M. and N.S. contributed to reviewing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by the Research Creativity and Management Office (RCMO), Universiti Sains Malaysia (USM), School of Medical Sciences, USM and Research University Incentive Grant with the Project Code: USM/1001/PPSP/8012252.

Institutional Review Board Statement

Ethical review and approval were waived for this study due to the study being a review of currently available literature.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Knol, M.J.; Twisk, J.W.; Beekman, A.T.; Heine, R.J.; Snoek, F.J.; Pouwer, F. Depression as a risk factor for the onset of type 2 diabetes mellitus. A meta-analysis. Diabetologia 2006, 49, 837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mauskopf, J.A.; Simon, G.E.; Kalsekar, A.; Nimsch, C.; Dunayevich, E.; Cameron, A. Nonresponse, partial response, and failure to achieve remission: Humanistic and cost burden in major depressive disorder. Depress. Anxiety 2009, 26, 83–97. [Google Scholar] [CrossRef] [PubMed]
  3. Sousa, R.D.D.; Rodrigues, A.M.; Gregório, M.J.; Branco, J.D.C.; Gouveia, M.J.; Canhão, H.; Dias, S.S. Anxiety and depression in the Portuguese older adults: Prevalence and associated factors. Front. Med. 2017, 4, 196. [Google Scholar] [CrossRef] [Green Version]
  4. Cohn, D.W.H.; Kinoshita, D.; Palermo-Neto, J. Antidepressants prevent hierarchy destabilization induced by lipopolysaccharide administration in mice: A neurobiological approach to depression. Ann. N. Y. Acad. Sci. 2012, 1262, 67–73. [Google Scholar] [CrossRef] [PubMed]
  5. Zhu, X.L.; Chen, J.J.; Han, F.; Pan, C.; Zhuang, T.T.; Cai, Y.F.; Lu, Y.P. Novel antidepressant effects of Paeonol alleviate neuronal injury with concomitant alterations in BDNF, Rac1 and RhoA levels in chronic unpredictable mild stress rats. Psychopharmacology 2018, 235, 2177–2191. [Google Scholar] [CrossRef] [PubMed]
  6. Cai, S.; Huang, S.; Hao, W. New hypothesis and treatment targets of depression: An integrated view of key findings. Neurosci. Bull. 2015, 31, 61–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Coppen, A. The biochemistry of affective disorders. Br. J. Psychiatry 1967, 113, 1237–1264. [Google Scholar] [CrossRef]
  8. Schildkraut, J.J. The catecholamine hypothesis of affective disorders: A review of supporting evidence. Am. J. Psychiatry 1965, 122, 509–522. [Google Scholar] [CrossRef]
  9. Belmaker, R.H.; Agam, G. Major depressive disorder. N. Engl. J. Med. 2008, 358, 55–68. [Google Scholar] [CrossRef] [Green Version]
  10. Hayley, S.; Poulter, M.O.; Merali, Z.; Anisman, H. The pathogenesis of clinical depression: Stressor-and cytokine-induced alterations of neuroplasticity. Neuroscience 2005, 135, 659–678. [Google Scholar] [CrossRef]
  11. Hiles, S.A.; Baker, A.L.; de Malmanche, T.; Attia, J. A meta-analysis of differences in IL-6 and IL-10 between people with and without depression: Exploring the causes of heterogeneity. Brain Behav. Immun. 2012, 26, 1180–1188. [Google Scholar] [CrossRef] [PubMed]
  12. Duman, R.S.; Deyama, S.; Fogaça, M.V. Role of BDNF in the pathophysiology and treatment of depression: Activity-dependent effects distinguish rapid-acting antidepressants. Eur. J. Neurosci. 2021, 53, 126–139. [Google Scholar] [CrossRef] [PubMed]
  13. Duman, R.S.; Heninger, G.R.; Nestler, E.J. A molecular and cellular theory of depression. Arch. Gen. Psychiatry 1997, 54, 597–606. [Google Scholar] [CrossRef]
  14. Kharade, S.M.; Gumate, D.S.; Naikwade, D. A Review: Hypothesis of Depression and Role of Antidepressant Drugs. Depression 2010, 15, 17. [Google Scholar]
  15. Delgado, P.L. How antidepressants help depression: Mechanisms of action and clinical response. J. Clin. Psychiatry 2004, 65, 25–30. [Google Scholar]
  16. Taylor, C.; Fricker, A.D.; Devi, L.A.; Gomes, I. Mechanisms of action of antidepressants: From neurotransmitter systems to signaling pathways. Cell. Signal. 2005, 17, 549–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Masand, P.S.; Gupta, S. Long-term side effects of newer-generation antidepressants: SSRIS, venlafaxine, nefazodone, bupropion, and mirtazapine. Ann. Clin. Psychiatry 2002, 14, 175–182. [Google Scholar] [CrossRef]
  18. Schweitzer, I.; Maguire, K.; Ng, C. Sexual side-effects of contemporary antidepressants. Aust. N. Z. J. Psychiatry 2009, 43, 795–808. [Google Scholar] [CrossRef]
  19. Mihaljević-Peleš, A.; Šagud, M.; Bajs Janović, M.; Kudlek Mikulić, S.; Jevtović, S. Do we need new therapeutic strategies for depression? Psychiatr. Danub. 2011, 23, 300–301. [Google Scholar] [PubMed]
  20. Bet, P.M.; Hugtenburg, J.G.; Penninx, B.W.; Hoogendijk, W.J. Side effects of antidepressants during long-term use in a naturalistic setting. Eur. Neuropsychopharmacol. 2013, 23, 1443–1451. [Google Scholar] [CrossRef] [Green Version]
  21. Richelson, E. Multi-modality: A new approach for the treatment of major depressive disorder. Int. J. Neuropsychopharmacol. 2013, 16, 1433–1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Sharma, T.; Guski, L.S.; Freund, N.; Gøtzsche, P.C. Suicidality and aggression during antidepressant treatment: Systematic review and meta-analyses based on clinical study reports. BMJ 2016, 352, i65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Crisafulli, C.; Fabbri, C.; Porcelli, S.; Drago, A.; Spina, E.; De Ronchi, D.; Serretti, A. Pharmacogenetics of antidepressants. Front. Pharmacol. 2011, 2, 6. [Google Scholar] [CrossRef] [Green Version]
  24. Ruhé, H.G.; van Rooijen, G.; Spijker, J.; Peeters, F.P.; Schene, A.H. Staging methods for treatment resistant depression. A systematic review. J. Affect. Disord. 2012, 137, 35–45. [Google Scholar] [CrossRef]
  25. Chiu, C.H.; Chyau, C.C.; Chen, C.C.; Lee, L.Y.; Chen, W.P.; Liu, J.L.; Lin, W.H.; Mong, M.C. Erinacine A-enriched Hericium erinaceus mycelium produces antidepressant-like effects through modulating BDNF/PI3K/Akt/GSK-3β signaling in mice. Int. J. Mol. Sci. 2018, 19, 341. [Google Scholar] [CrossRef] [Green Version]
  26. Manosso, L.M.; Moretti, M.; Ribeiro, C.M.; Gonçalves, F.M.; Leal, R.B.; Rodrigues, A.L.S. Antidepressant-like effect of zinc is dependent on signaling pathways implicated in BDNF modulation. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2015, 59, 59–67. [Google Scholar]
  27. Ali, S.S.; Abd El Wahab, M.G.; Ayuob, N.N.; Suliaman, M. The antidepressant-like effect of Ocimum basilicum in an animal model of depression. Biotech. Histochem. 2017, 92, 390–401. [Google Scholar] [CrossRef]
  28. Erejuwa, O.O.; Sulaiman, S.A.; Ab Wahab, M.S. Honey: A novel antioxidant. Molecules 2012, 17, 4400–4423. [Google Scholar] [CrossRef]
  29. Mijanur Rahman, M.; Gan, S.H.; Khalil, M. Neurological effects of honey: Current and future prospects. Evid.-Based Complement. Altern. Med. 2014, 2014, 958721. [Google Scholar] [CrossRef]
  30. Münstedt, K.; Voss, B.; Kullmer, U.; Schneider, U.; Hübner, J. Bee pollen and honey for the alleviation of hot flushes and other menopausal symptoms in breast cancer patients. Mol. Clin. Oncol. 2015, 3, 869–874. [Google Scholar] [CrossRef] [Green Version]
  31. Abd Wahab, M.S.; Othman, N.; Othman, N.H.I.; Jamari, A.A.; Ali, A.A. Exploring the use of and perceptions about honey as complementary and alternative medicine among the general public in the state of Selangor, Malaysia. J. Appl. Pharm. Sci. 2017, 7, 144–150. [Google Scholar]
  32. Fatima, I.J.; Ab, M.H.; Salwani, I.; Lavaniya, M. Physicochemical characteristics of Malaysian stingless bee honey from trigona species. IIUM Med. J. Malays. 2018, 17, 187–191. [Google Scholar]
  33. Roowi, S.; Muhamad, S.A.; Sipon, H.; Jaafar, M.; Daud, M.; Hisham, N.; Othman, R. Asid fenolik bebas dalam madu kelulut. Bul. Teknol. MARDI 2012, 2, 145–147. [Google Scholar]
  34. Biswa, R.; Sarkar, A.; Khewa, S.S. Ethnomedicinal uses of honey of stingless bee by Nepali community of Darjeeling foothills of West Bengal, India. Indian J. Tradit. Knowl. 2017, 16, 648–653. [Google Scholar]
  35. Özbalci, B.; Boyaci, İ.H.; Topcu, A.; Kadılar, C.; Tamer, U. Rapid analysis of sugars in honey by processing Raman spectrum using chemometric methods and artificial neural networks. Food Chem. 2013, 136, 1444–1452. [Google Scholar] [CrossRef]
  36. Kek, S.P.; Chin, N.L.; Yusof, Y.A.; Tan, S.W.; Chua, L.S. Total phenolic contents and colour intensity of Malaysian honeys from the Apis spp. and Trigona spp. bees. Agric. Agric. Sci. Procedia 2014, 2, 150–155. [Google Scholar]
  37. Biluca, F.C.; Della Betta, F.; de Oliveira, G.P.; Pereira, L.M.; Gonzaga, L.V.; Costa, A.C.O.; Fett, R. 5-HMF and carbohydrates content in stingless bee honey by CE before and after thermal treatment. Food Chem. 2014, 159, 244–249. [Google Scholar] [CrossRef] [Green Version]
  38. Sun, J.; Wang, H.; Liu, B.; Shi, W.; Shi, J.; Zhang, Z.; Xing, J. Rutin attenuates H2O2-induced oxidation damage and apoptosis in Leydig cells by activating PI3K/Akt signal pathways. Biomed. Pharmacother. 2017, 88, 500–506. [Google Scholar] [CrossRef]
  39. Kanimozhi, S.; Bhavani, P.; Subramanian, P. Influence of the flavonoid, quercetin on antioxidant status, lipid peroxidation and histopathological changes in hyperammonemic rats. Indian J. Clin. Biochem. 2017, 32, 275–284. [Google Scholar] [CrossRef]
  40. Nutt, D.J. Relationship of neurotransmitters to the symptoms of major depressive disorder. J. Clin. Psychiatry 2008, 69, 4–7. [Google Scholar]
  41. Zhang, Z.; Deng, T.; Wu, M.; Zhu, A.; Zhu, G. Botanicals as modulators of depression and mechanisms involved. Chin. Med. 2019, 14, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Südhof, T.C. The cell biology of synapse formation. J. Cell Biol. 2021, 220, e202103052. [Google Scholar] [CrossRef] [PubMed]
  43. Edmondson, D.E.; Mattevi, A.; Binda, C.; Li, M.; Hubalek, F. Structure and mechanism of monoamine oxidase. Curr. Med. Chem. 2004, 11, 1983–1993. [Google Scholar] [CrossRef] [PubMed]
  44. Kulkarni, S.K.; Dhir, A.; Akula, K.K. Potentials of curcumin as an antidepressant. Sci. World J. 2009, 9, 1233–1241. [Google Scholar] [CrossRef]
  45. Moret, C.; Briley, M. The importance of norepinephrine in depression. Neuropsychiatr. Dis. Treat. 2011, 7 (Suppl 1), 9. [Google Scholar]
  46. Freis, E.D. Mental depression in hypertensive patients treated for long periods with large doses of reserpine. N. Engl. J. Med. 1954, 251, 1006–1008. [Google Scholar] [CrossRef] [Green Version]
  47. Pletscher, A. The discovery of antidepressants: A winding path. Experientia 1991, 47, 4–8. [Google Scholar] [CrossRef]
  48. Radak, Z.; Marton, O.; Nagy, E.; Koltai, E.; Goto, S. The complex role of physical exercise and reactive oxygen species on brain. J. Sport Health Sci. 2013, 2, 87–93. [Google Scholar] [CrossRef] [Green Version]
  49. Lee, S.; Kim, H.B.; Hwang, E.S.; Kim, E.S.; Kim, S.S.; Jeon, T.D.; Song, M.C.; Lee, J.S.; Chung, M.C.; Maeng, S.; et al. Antidepressant-like effects of p-coumaric acid on LPS-induced depressive and inflammatory changes in rats. Exp. Neurobiol. 2018, 27, 189. [Google Scholar] [CrossRef]
  50. Tunnicliff, G.; Malatynska, E. Central GABAergic systems and depressive illness. Neurochem. Res. 2003, 28, 965–976. [Google Scholar] [CrossRef]
  51. Rupprecht, R.; Eser, D.; Zwanzger, P.; Möller, H.J. GABAA receptors as targets for novel anxiolytic drugs. World J. Biol. Psychiatry 2006, 7, 231–237. [Google Scholar] [CrossRef] [PubMed]
  52. Sequeira, A.; Turecki, G. Genome wide gene expression studies in mood disorders. Omics A J. Integr. Biol. 2006, 10, 444–454. [Google Scholar] [CrossRef] [PubMed]
  53. Sanacora, G.; Saricicek, A. GABAergic contributions to the pathophysiology of depression and the mechanism of antidepressant action. CNS Neurol. Disord.-Drug Targets (Former. Curr. Drug Targets-CNS Neurol. Disord.) 2007, 6, 127–140. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, M.; Schwab, C.; Mcgeer, P.L. Astrocytes are GABAergic cells that modulate microglial activity. Glia 2011, 59, 152–165. [Google Scholar] [CrossRef]
  55. Oeckinghaus, A.; Ghosh, S. The NF-κB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034. [Google Scholar] [CrossRef]
  56. Wichers, M.; Maes, M. The psychoneuroimmuno-pathophysiology of cytokine-induced depression in humans. Int. J. Neuropsychopharmacol. 2002, 5, 375–388. [Google Scholar] [CrossRef]
  57. Karin, M.; Greten, F.R. NF-κB: Linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol. 2005, 5, 749. [Google Scholar] [CrossRef]
  58. Napetschnig, J.; Wu, H. Molecular basis of NF-κB signaling. Annu. Rev. Biophys. 2013, 42, 443–468. [Google Scholar] [CrossRef] [Green Version]
  59. Yu, B.; Chang, J.; Liu, Y.; Li, J.; Kevork, K.; Al-Hezaimi, K.; Graves, D.T.; Park, N.H.; Wang, C.Y. Wnt4 signaling prevents skeletal aging and inflammation by inhibiting nuclear factor-κB. Nat. Med. 2014, 20, 1009. [Google Scholar] [CrossRef] [Green Version]
  60. FitzGerald, G.A. COX-2 and beyond: Approaches to prostaglandin inhibition in human disease. Nat. Rev. Drug Discov. 2003, 2, 879. [Google Scholar] [CrossRef]
  61. Munhoz, C.D.; Lepsch, L.B.; Kawamoto, E.M.; Malta, M.B.; de Sá Lima, L.; Avellar, M.C.W.; Sapolsky, R.M.; Scavone, C. Chronic unpredictable stress exacerbates lipopolysaccharide-induced activation of nuclear factor-κB in the frontal cortex and hippocampus via glucocorticoid secretion. J. Neurosci. 2006, 26, 3813–3820. [Google Scholar] [CrossRef] [PubMed]
  62. Miller, A.H.; Maletic, V.; Raison, C.L. Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression. Biol. Psychiatry 2009, 65, 732–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Han, Q.Q.; Yu, J. Inflammation: A mechanism of depression? Neurosci. Bull. 2014, 30, 515–523. [Google Scholar] [CrossRef] [Green Version]
  64. Llorens-Martin, M.; Jurado-Arjona, J.; Fuster-Matanzo, A.; Hernandez, F.; Rabano, A.; Avila, J. Peripherally triggered and GSK-3β-driven brain inflammation differentially skew adult hippocampal neurogenesis, behavioral pattern separation and microglial activation in response to ibuprofen. Transl. Psychiatry 2014, 4, e463. [Google Scholar] [CrossRef]
  65. Norden, D.M.; McCarthy, D.O.; Bicer, S.; Devine, R.D.; Reiser, P.J.; Godbout, J.P.; Wold, L.E. Ibuprofen ameliorates fatigue-and depressive-like behavior in tumor-bearing mice. Life Sci. 2015, 143, 65–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Koo, J.W.; Duman, R.S. IL-1β is an essential mediator of the antineurogenic and anhedonic effects of stress. Proc. Natl. Acad. Sci. USA 2008, 105, 751–756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Pérez-Sánchez, G.; Becerril-Villanueva, E.; Arreola, R.; Martínez-Levy, G.; Hernández-Gutiérrez, M.E.; Velasco-Velásquez, M.A.; Alvarez-Herrera, S.; Cruz-Fuentes, C.; Palacios, L.; De La Peña, F.; et al. Inflammatory profiles in depressed adolescents treated with fluoxetine: An 8-week follow-up open study. Mediat. Inflamm. 2018, 2018, 4074051. [Google Scholar] [CrossRef] [Green Version]
  69. Kasai, T.; Inada, K.; Takakuwa, T.; Yamada, Y.; Inoue, Y.; Shimamura, T.; Taniguchi, S.; Sato, S.; Wakabayashi, G.; Endo, S. Anti-inflammatory cytokine levels in patients with septic shock. Res. Commun. Mol. Pathol. Pharmacol. 1997, 98, 34–42. [Google Scholar]
  70. Rubio-Perez, J.M.; Morillas-Ruiz, J.M. A Review: Inflammatory Process in Alzheimer’s Disease, Role of Cytokines. Sci. World J. 2012, 2012, 756357. [Google Scholar] [CrossRef]
  71. Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.; Reim, E.K.; Lanctôt, K.L. A Meta-analysis of cytokines in major depression. Biol. Psychiatry 2010, 67, 446–457. [Google Scholar] [CrossRef] [PubMed]
  72. Leighton, S.P.; Nerurkar, L.; Krishnadas, R.; Johnman, C.; Graham, G.J.; Cavanagh, J. Chemokines in depression in health and in inflammatory illness: A systematic review and meta-analysis. Mol. Psychiatry 2018, 23, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Duman, R.S.; Aghajanian, G.K. Synaptic dysfunction in depression: Potential therapeutic targets. Science 2012, 338, 68–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Capuron, L.; Miller, A.H. Immune system to brain signaling: Neuropsychopharmacological implications. Pharmacol. Ther. 2011, 130, 226–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Haroon, E.; Miller, A.H.; Sanacora, G. Inflammation, glutamate, and glia: A trio of trouble in mood disorders. Neuropsychopharmacology 2017, 42, 193. [Google Scholar] [CrossRef] [PubMed]
  76. Park, S.E.; Dantzer, R.; Kelley, K.W.; McCusker, R.H. Central administration of insulin-like growth factor-I decreases depressive-like behavior and brain cytokine expression in mice. J. Neuroinflamm. 2011, 8, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Dobos, N.; de Vries, E.F.; Kema, I.P.; Patas, K.; Prins, M.; Nijholt, I.M.; Dierckx, R.A.; Korf, J.; den Boer, J.A.; Luiten, P.G.; et al. The role of indoleamine 2, 3-dioxygenase in a mouse model of neuroinflammation-induced depression. J. Alzheimer’s Dis. 2012, 28, 905–915. [Google Scholar] [CrossRef] [Green Version]
  78. Lawson, M.A.; Parrott, J.M.; McCusker, R.H.; Dantzer, R.; Kelley, K.W.; O’Connor, J.C. Intracerebroventricular administration of lipopolysaccharide induces indoleamine-2, 3-dioxygenase-dependent depression-like behaviors. J. Neuroinflamm. 2013, 10, 875. [Google Scholar] [CrossRef] [Green Version]
  79. Vancassel, S.; Capuron, L.; Castanon, N. Brain kynurenine and BH4 pathways: Relevance to the pathophysiology and treatment of inflammation-driven depressive symptoms. Front. Neurosci. 2018, 12, 499. [Google Scholar] [CrossRef]
  80. Hestad, K.A.; Engedal, K.; Whist, J.E.; Farup, P.G. The relationships among tryptophan, kynurenine, indoleamine 2, 3-dioxygenase, depression, and neuropsychological performance. Front. Psychol. 2017, 8, 1561. [Google Scholar] [CrossRef] [Green Version]
  81. O’connor, J.C.; Lawson, M.A.; Andre, C.; Moreau, M.; Lestage, J.; Castanon, N.; Kelley, K.W.; Dantzer, R. Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2, 3-dioxygenase activation in mice. Mol. Psychiatry 2009, 14, 511. [Google Scholar] [CrossRef] [Green Version]
  82. Raison, C.L.; Dantzer, R.; Kelley, K.W.; Lawson, M.A.; Woolwine, B.J.; Vogt, G.; Spivey, J.R.; Saito, K.; Miller, A.H. CSF concentrations of brain tryptophan and kynurenines during immune stimulation with IFN-α: Relationship to CNS immune responses and depression. Mol. Psychiatry 2010, 15, 393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Schwarcz, R.; Stone, T.W. The kynurenine pathway and the brain: Challenges, controversies and promises. Neuropharmacology 2017, 112, 237–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Mustafa, M.Z.; Zulkifli, F.N.; Fernandez, I.; Mariatulqabtiah, A.R.; Sangu, M.; Nor Azfa, J.; Mohamed, M.; Roslan, N. Stingless bee honey improves spatial memory in mice, probably associated with brain-derived neurotrophic factor (BDNF) and inositol 1, 4, 5-triphosphate receptor type 1 (Itpr1) genes. Evid.-Based Complement. Altern. Med. 2019, 2019, 8258307. [Google Scholar] [CrossRef] [Green Version]
  85. Lu, L.; Ben, X.; Xiao, L.; Peng, M.; Zhang, Y. AMP-activated protein kinase activation in mediating phenylalanine-induced neurotoxicity in experimental models of phenylketonuria. J. Inherit. Metab. Dis. 2018, 41, 679–687. [Google Scholar] [CrossRef] [PubMed]
  86. Strasser, B.; Sperner-Unterweger, B.; Fuchs, D.; Gostner, J.M. Mechanisms of inflammation-associated depression: Immune influences on tryptophan and phenylalanine metabolisms. Inflamm.-Assoc. Depress. Evid. Mech. Implic. 2016, 31, 95–115. [Google Scholar]
  87. Froböse, M.I.; Cools, R. Chemical neuromodulation of cognitive control avoidance. Curr. Opin. Behav. Sci. 2018, 22, 121–127. [Google Scholar] [CrossRef]
  88. Haroon, E.; Raison, C.L.; Miller, A.H. Psychoneuroimmunology meets neuropsychopharmacology: Translational implications of the impact of inflammation on behavior. Neuropsychopharmacology 2012, 37, 137. [Google Scholar] [CrossRef] [PubMed]
  89. Neurauter, G.; Schrocksnadel, K.; Scholl-Burgi, S.; Sperner-Unterweger, B.; Schubert, C.; Ledochowski, M.; Fuchs, D. Chronic immune stimulation correlates with reduced phenylalanine turnover. Curr. Drug Metab. 2008, 9, 622–627. [Google Scholar] [CrossRef] [PubMed]
  90. Duman, R.S.; Li, N. A neurotrophic hypothesis of depression: Role of synaptogenesis in the actions of NMDA receptor antagonists. Philos. Trans. R. Soc. B Biol. Sci. 2012, 367, 2475–2484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Begni, V.; Riva, M.A.; Cattaneo, A. Cellular and molecular mechanisms of the brain-derived neurotrophic factor in physiological and pathological conditions. Clin. Sci. 2017, 131, 123–138. [Google Scholar] [CrossRef]
  92. Galea, R.; Cassar, D. Brain-Derived Neurotrophic Factor—Major Depressive Disorder and Suicide. Open Access Libr. J. 2019, 6, e5106. [Google Scholar] [CrossRef]
  93. Björkholm, C.; Monteggia, L.M. BDNF–a key transducer of antidepressant effects. Neuropharmacology 2016, 102, 72–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lee, B.H.; Kim, Y.K. The roles of BDNF in the pathophysiology of major depression and in antidepressant treatment. Psychiatry Investig. 2010, 7, 231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Park, H.; Poo, M.M. Neurotrophin regulation of neural circuit development and function. Nat. Rev. Neurosci. 2013, 14, 7. [Google Scholar] [CrossRef] [PubMed]
  96. Lu, B.; Nagappan, G.; Lu, Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. Neurotrophic Factors 2014, 220, 223–250. [Google Scholar]
  97. Zheleznyakova, G.Y.; Cao, H.; Schiöth, H.B. BDNF DNA methylation changes as a biomarker of psychiatric disorders: Literature review and open access database analysis. Behav. Brain Funct. 2016, 12, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Altar, C.A. Neurotrophins and depression. Trends Pharmacol. Sci. 1999, 20, 59–62. [Google Scholar] [CrossRef]
  99. Numakawa, T.; Odaka, H.; Adachi, N. Actions of brain-derived neurotrophin factor in the neurogenesis and neuronal function, and its involvement in the pathophysiology of brain diseases. Int. J. Mol. Sci. 2018, 19, 3650. [Google Scholar] [CrossRef] [Green Version]
  100. Zhu, G.; Li, J.; He, L.; Wang, X.; Hong, X. MPTP-induced changes in hippocampal synaptic plasticity and memory are prevented by memantine through the BDNF-TrkB pathway. Br. J. Pharmacol. 2015, 172, 2354–2368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Yang, S.J.; Song, Z.J.; Wang, X.C.; Zhang, Z.R.; Wu, S.B.; Zhu, G.Q. Curculigoside facilitates fear extinction and prevents depression-like behaviors in a mouse learned helplessness model through increasing hippocampal BDNF. Acta Pharmacol. Sin. 2019, 40, 1269–1278. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, B.B.; Luo, L.; Liu, X.L.; Geng, D.; Liu, Q.; Yi, L.T. 7-Chlorokynurenic acid (7-CTKA) produces rapid antidepressant-like effects: Through regulating hippocampal microRNA expressions involved in TrkB-ERK/Akt signaling pathways in mice exposed to chronic unpredictable mild stress. Psychopharmacology 2015, 232, 541–550. [Google Scholar] [CrossRef] [PubMed]
  103. Yan, T.; He, B.; Wan, S.; Xu, M.; Yang, H.; Xiao, F.; Bi, K.; Jia, Y. Antidepressant-like effects and cognitive enhancement of Schisandra chinensis in chronic unpredictable mild stress mice and its related mechanism. Sci. Rep. 2017, 7, 6903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Zhang, X.; Song, Y.; Bao, T.; Yu, M.; Xu, M.; Guo, Y.; Wang, Y.; Zhang, C.; Zhao, B. Antidepressant-like effects of acupuncture involved the ERK signaling pathway in rats. BMC Complement. Altern. Med. 2016, 16, 380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Chen, X.Q.; Li, C.F.; Chen, S.J.; Liang, W.N.; Wang, M.; Wang, S.S.; Dong, S.Q.; Yi, L.T.; Li, C.D. The antidepressant-like effects of Chaihu Shugan San: Dependent on the hippocampal BDNF-TrkB-ERK/Akt signaling activation in perimenopausal depression-like rats. Biomed. Pharmacother. 2018, 105, 45–52. [Google Scholar] [CrossRef]
  106. Man, H.Y.; Wang, Q.; Lu, W.Y.; Ju, W.; Ahmadian, G.; Liu, L.; D’Souza, S.; Wong, T.P.; Taghibiglou, C.; Lu, J.; et al. Activation of PI3-kinase is required for AMPA receptor insertion during LTP of mEPSCs in cultured hippocampal neurons. Neuron 2003, 38, 611–624. [Google Scholar] [CrossRef] [Green Version]
  107. Stornetta, R.L.; Zhu, J.J. Ras and Rap signaling in synaptic plasticity and mental disorders. Neuroscientist 2011, 17, 54–78. [Google Scholar] [CrossRef]
  108. Duman, R.S.; Monteggia, L.M. A neurotrophic model for stress-related mood disorders. Biol. Psychiatry 2006, 59, 1116–1127. [Google Scholar] [CrossRef]
  109. 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] [Green Version]
  110. Tomita, H.; Ziegler, M.E.; Kim, H.B.; Evans, S.J.; Choudary, P.V.; Li, J.Z.; Meng, F.; Dai, M.; Neal, C.R.; Myers, R.M.; et al. G protein-linked signaling pathways in bipolar and major depressive disorders. Front. Genet. 2013, 4, 297. [Google Scholar] [CrossRef] [Green Version]
  111. Plattner, F.; Hayashi, K.; Hernández, A.; Benavides, D.R.; Tassin, T.C.; Tan, C.; Day, J.; Fina, M.W.; Yuen, E.Y.; Yan, Z.; et al. The role of ventral striatal cAMP signaling in stress-induced behaviors. Nat. Neurosci. 2015, 18, 1094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Duman, R.S.; Aghajanian, G.K.; Sanacora, G.; Krystal, J.H. Synaptic plasticity and depression: New insights from stress and rapid-acting antidepressants. Nat. Med. 2016, 22, 238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Cramer, S.C.; Sur, M.; Dobkin, B.H.; O’Brien, C.; Sanger, T.D.; Trojanowski, J.Q.; Rumsey, J.M.; Hicks, R.; Cameron, J.; Chen, D.; et al. Harnessing neuroplasticity for clinical applications. Brain 2011, 134, 1591–1609. [Google Scholar] [CrossRef] [PubMed]
  114. Levy, M.J.; Boulle, F.; Steinbusch, H.W.; van den Hove, D.L.; Kenis, G.; Lanfumey, L. Neurotrophic factors and neuroplasticity pathways in the pathophysiology and treatment of depression. Psychopharmacology 2018, 235, 2195–2220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Vilar, M.; Mira, H. Regulation of neurogenesis by neurotrophins during adulthood: Expected and unexpected roles. Front. Neurosci. 2016, 10, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Seib, D.R.; Martin-Villalba, A. Neurogenesis in the normal ageing hippocampus: A mini-review. Gerontology 2015, 61, 327–335. [Google Scholar] [CrossRef]
  117. Eisch, A.J.; Petrik, D. Depression and hippocampal neurogenesis: A road to remission? Science 2012, 338, 72–75. [Google Scholar] [CrossRef] [Green Version]
  118. Bremner, J.D.; Narayan, M.; Anderson, E.R.; Staib, L.H.; Miller, H.L.; Charney, D.S. Hippocampal volume reduction in major depression. Am. J. Psychiatry 2000, 157, 115–118. [Google Scholar] [CrossRef]
  119. Cole, J.; Costafreda, S.G.; McGuffin, P.; Fu, C.H. Hippocampal atrophy in first episode depression: A meta-analysis of magnetic resonance imaging studies. J. Affect. Disord. 2011, 134, 483–487. [Google Scholar] [CrossRef]
  120. Frodl, T.; Schüle, C.; Schmitt, G.; Born, C.; Baghai, T.; Zill, P.; Bottlender, R.; Rupprecht, R.; Bondy, B.; Reiser, M.; et al. Association of the brain-derived neurotrophic factor Val66Met polymorphism with reduced hippocampal volumes in major depression. Arch. Gen. Psychiatry 2007, 64, 410–416. [Google Scholar] [CrossRef] [Green Version]
  121. Krishnan, V.; Nestler, E.J. The molecular neurobiology of depression. Nature 2008, 455, 894. [Google Scholar] [CrossRef] [PubMed]
  122. Castrén, E.; Rantamäki, T. The role of BDNF and its receptors in depression and antidepressant drug action: Reactivation of developmental plasticity. Dev. Neurobiol. 2010, 70, 289–297. [Google Scholar] [CrossRef] [PubMed]
  123. Drevets, W.C. Functional anatomical abnormalities in limbic and prefrontal cortical structures in major depression. Prog. Brain Res. 2000, 126, 413–431. [Google Scholar] [PubMed]
  124. Cook, S.C.; Wellman, C.L. Chronic stress alters dendritic morphology in rat medial prefrontal cortex. J. Neurobiol. 2004, 60, 236–248. [Google Scholar] [CrossRef] [PubMed]
  125. Pittenger, C. Disorders of memory and plasticity in psychiatric disease. Dialogues Clin. Neurosci. 2013, 15, 455. [Google Scholar] [CrossRef] [PubMed]
  126. Gerhard, D.M.; Wohleb, E.S.; Duman, R.S. Emerging treatment mechanisms for depression: Focus on glutamate and synaptic plasticity. Drug Discov. Today 2016, 21, 454–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Castrén, E.; Hen, R. Neuronal plasticity and antidepressant actions. Trends Neurosci. 2013, 36, 259–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Figurov, A.; Pozzo-Miller, L.D.; Olafsson, P.; Wang, T.; Lu, B. Regulation of synaptic responses to high-frequency stimulation and LTP by neurotrophins in the hippocampus. Nature 1996, 381, 706. [Google Scholar] [CrossRef] [PubMed]
  129. Kovalchuk, Y.; Hanse, E.; Kafitz, K.W.; Konnerth, A. Postsynaptic induction of BDNF-mediated long-term potentiation. Science 2002, 295, 1729–1734. [Google Scholar] [CrossRef] [PubMed]
  130. Tanaka, J.I.; Horiike, Y.; Matsuzaki, M.; Miyazaki, T.; Ellis-Davies, G.C.; Kasai, H. Protein synthesis and neurotrophin-dependent structural plasticity of single dendritic spines. Science 2008, 319, 1683–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Kempton, M.J.; Salvador, Z.; Munafò, M.R.; Geddes, J.R.; Simmons, A.; Frangou, S.; Williams, S.C. Structural neuroimaging studies in major depressive disorder: Meta-analysis and comparison with bipolar disorder. Arch. Gen. Psychiatry 2011, 68, 675–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Miller, B.R.; Hen, R. The current state of the neurogenic theory of depression and anxiety. Curr. Opin. Neurobiol. 2015, 30, 51–58. [Google Scholar] [CrossRef] [Green Version]
  133. Arnone, D.; McIntosh, A.M.; Ebmeier, K.P.; Munafò, M.R.; Anderson, I.M. Magnetic resonance imaging studies in unipolar depression: Systematic review and meta-regression analyses. Eur. Neuropsychopharmacol. 2012, 22, 1–16. [Google Scholar] [CrossRef] [PubMed]
  134. Videbech, P.; Ravnkilde, B. Hippocampal volume and depression: A meta-analysis of MRI studies. Am. J. Psychiatry 2004, 161, 1957–1966. [Google Scholar] [CrossRef] [PubMed]
  135. Pittenger, C.; Duman, R.S. Stress, depression, and neuroplasticity: A convergence of mechanisms. Neuropsychopharmacology 2008, 33, 88. [Google Scholar] [CrossRef] [PubMed]
  136. Kim, J.J.; Diamond, D.M. The stressed hippocampus, synaptic plasticity and lost memories. Nat. Rev. Neurosci. 2002, 3, 453. [Google Scholar] [CrossRef] [PubMed]
  137. Goldwater, D.S.; Pavlides, C.; Hunter, R.G.; Bloss, E.B.; Hof, P.R.; McEwen, B.S.; Morrison, J.H. Structural and functional alterations to rat medial prefrontal cortex following chronic restraint stress and recovery. Neuroscience 2009, 164, 798–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Xu, L.; Anwyl, R.; Rowan, M.J. Behavioural stress facilitates the induction of long-term depression in the hippocampus. Nature 1997, 387, 497. [Google Scholar] [CrossRef] [PubMed]
  139. Fuenmayor, C.A.; Zuluaga-Domínguez, C.M.; Díaz-Moreno, A.C.; Quicazán, M.C. ‘Miel de Angelita’: Nutritional composition and physicochemical properties of Tetragonisca angustula honey. Interciencia 2012, 37, 142–147. [Google Scholar]
  140. Manzanares, A.B.; García, Z.H.; Galdón, B.R.; Rodríguez, E.R.; Romero, C.D. Physicochemical characteristics of minor monofloral honeys from Tenerife, Spain. LWT-Food Sci. Technol. 2014, 55, 572–578. [Google Scholar] [CrossRef]
  141. Ismail, W.W. A review on beekeeping in Malaysia: History, importance and future directions. J. Sustain. Sci. Manag. 2016, 11, 70–80. [Google Scholar]
  142. Bakar, M.A.; Sanusi, S.B.; Bakar, F.A.; Cong, O.J.; Mian, Z. Physicochemical and antioxidant potential of raw unprocessed honey from Malaysian stingless bees. Pak. J. Nutr. 2017, 16, 888–894. [Google Scholar] [CrossRef] [Green Version]
  143. Mustafa, M.Z.; Yaacob, N.S.; Sulaiman, S.A. Reinventing the honey industry: Opportunities of the stingless bee. Malays. J. Med. Sci. 2018, 25, 1–5. [Google Scholar] [CrossRef] [PubMed]
  144. Litwack, G. Chapter 13—Metabolism of Amino Acids. In Human Biochemistry; Academic Press: Cambridge, MA, USA, 2018. [Google Scholar]
  145. O’Neill, E.; Harkin, A. Targeting the noradrenergic system for anti-inflammatory and neuroprotective effects: Implications for Parkinson’s disease. Neural Regen. Res. 2018, 13, 1332. [Google Scholar] [PubMed]
  146. Mahar, I.; Bambico, F.R.; Mechawar, N.; Nobrega, J.N. Stress, serotonin, and hippocampal neurogenesis in relation to depression and antidepressant effects. Neurosci. Biobehav. Rev. 2014, 38, 173–192. [Google Scholar] [CrossRef] [PubMed]
  147. Gotlib, I.H.; Joormann, J. Cognition and depression: Current status and future directions. Annu. Rev. Clin. Psychol. 2010, 6, 285–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Belujon, P.; Grace, A.A. Dopamine system dysregulation in major depressive disorders. Int. J. Neuropsychopharmacol. 2017, 20, 1036–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Mallik, S.B.; Mudgal, J.; Nampoothiri, M.; Hall, S.; Anoopkumar-Dukie, S.; Grant, G.; Rao, C.M.; Arora, D. Caffeic acid attenuates lipopolysaccharide-induced sickness behaviour and neuroinflammation in mice. Neurosci. Lett. 2016, 632, 218–223. [Google Scholar] [CrossRef]
  150. Ranneh, Y.; Akim, A.M.; Hamid, H.A.; Khazaai, H.; Fadel, A.; Mahmoud, A.M. Stingless bee honey protects against lipopolysaccharide induced-chronic subclinical systemic inflammation and oxidative stress by modulating Nrf2, NF-κB and p38 MAPK. Nutr. Metab. 2019, 16, 1–17. [Google Scholar] [CrossRef] [PubMed]
  151. Uthurry, C.A.; Hevia, D.; Gomez-Cordoves, C. Role of honey polyphenols in health. J. ApiProduct ApiMedical Sci. 2011, 3, 141–159. [Google Scholar] [CrossRef]
  152. Khalil, M.I.; Alam, N.; Moniruzzaman, M.; Sulaiman, S.A.; Gan, S.H. Phenolic acid composition and antioxidant properties of Malaysian honeys. J. Food Sci. 2011, 76, C921–C928. [Google Scholar] [CrossRef] [PubMed]
  153. Beretta, G.; Granata, P.; Ferrero, M.; Orioli, M.; Facino, R.M. Standardization of antioxidant properties of honey by a combination of spectrophotometric/fluorimetric assays and chemometrics. Anal. Chim. Acta 2005, 533, 185–191. [Google Scholar] [CrossRef]
  154. Bertoncelj, J.; Doberšek, U.; Jamnik, M.; Golob, T. Evaluation of the phenolic content, antioxidant activity and colour of Slovenian honey. Food Chem. 2007, 105, 822–828. [Google Scholar] [CrossRef]
  155. Meda, A.; Lamien, C.E.; Romito, M.; Millogo, J.; Nacoulma, O.G. Determination of the total phenolic, flavonoid and proline contents in Burkina Fasan honey, as well as their radical scavenging activity. Food Chem. 2005, 91, 571–577. [Google Scholar] [CrossRef]
  156. Moniruzzaman, M.; Khalil, M.I.; Sulaiman, S.A.; Gan, S.H. Physicochemical and antioxidant properties of Malaysian honeys produced by Apis cerana, Apis dorsata and Apis mellifera. BMC Complement. Altern. Med. 2013, 13, 43. [Google Scholar] [CrossRef] [Green Version]
  157. Esposito, E.; Rotilio, D.; Di Matteo, V.; Di Giulio, C.; Cacchio, M.; Algeri, S. A review of specific dietary antioxidants and the effects on biochemical mechanisms related to neurodegenerative processes. Neurobiol. Aging 2002, 23, 719–735. [Google Scholar] [CrossRef]
  158. Lau, F.C.; Shukitt-Hale, B.; Joseph, J.A. The beneficial effects of fruit polyphenols on brain aging. Neurobiol. Aging 2005, 26, 128–132. [Google Scholar] [CrossRef]
  159. Schmitt-Schillig, S.; Schaffer, S.; Weber, C.C.; Eckert, G.P.; Muller, W.E. Flavonoids and the aging brain. J. Physiol. Pharmacol. Suppl. 2005, 56, 23–36. [Google Scholar]
  160. Azman, K.F.; Zakaria, R.; Abdul Aziz, C.B.; Othman, Z. Tualang honey attenuates noise stress-induced memory deficits in aged rats. Oxidative Med. Cell. Longev. 2016, 2016, 1549158. [Google Scholar] [CrossRef] [Green Version]
  161. Kubera, M.; Obuchowicz, E.; Goehler, L.; Brzeszcz, J.; Maes, M. In animal models, psychosocial stress-induced (neuro) inflammation, apoptosis and reduced neurogenesis are associated to the onset of depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2011, 35, 744–759. [Google Scholar] [CrossRef]
  162. Lindqvist, D.; Dhabhar, F.S.; James, S.J.; Hough, C.M.; Jain, F.A.; Bersani, F.S.; Reus, V.I.; Verhoeven, J.E.; Epel, E.S.; Mahan, L.; et al. Oxidative stress, inflammation and treatment response in major depression. Psychoneuroendocrinology 2017, 76, 197–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Czarny, P.; Wigner, P.; Galecki, P.; Sliwinski, T. The interplay between inflammation, oxidative stress, DNA damage, DNA repair and mitochondrial dysfunction in depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 80, 309–321. [Google Scholar] [CrossRef] [PubMed]
  164. Hritcu, L.; Ionita, R.; Postu, P.A.; Gupta, G.K.; Turkez, H.; Lima, T.C.; Carvalho, C.U.S.; de Sousa, D.P. Antidepressant flavonoids and their relationship with oxidative stress. Oxidative Med. Cell. Longev. 2017, 2017, 5762172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Abdel-Wahab, M.H.; El-Mahdy, M.A.; Abd-Ellah, M.F.; Helal, G.K.; Khalifa, F.; Hamada, F.M.A. Influence of p-coumaric acid on doxorubicin-induced oxidative stress in rat’s heart. Pharmacol. Res. 2003, 48, 461–465. [Google Scholar] [CrossRef]
  166. Ferguson, L.R.; Zhu, S.T.; Harris, P.J. Antioxidant and antigenotoxic effects of plant cell wall hydroxycinnamic acids in cultured HT-29 cells. Mol. Nutr. Food Res. 2005, 49, 585–593. [Google Scholar] [CrossRef] [PubMed]
  167. Vauzour, D.; Corona, G.; Spencer, J.P. Caffeic acid, tyrosol and p-coumaric acid are potent inhibitors of 5-S-cysteinyl-dopamine induced neurotoxicity. Arch. Biochem. Biophys. 2010, 501, 106–111. [Google Scholar] [CrossRef]
  168. Filho, C.B.; Jesse, C.R.; Donato, F.; Del Fabbro, L.; de Gomes, M.G.; Goes, A.T.R.; Souza, L.C.; Giacomeli, R.; Antunes, M.; Luchese, C.; et al. Neurochemical factors associated with the antidepressant-like effect of flavonoid chrysin in chronically stressed mice. Eur. J. Pharmacol. 2016, 791, 284–296. [Google Scholar] [CrossRef]
  169. Filho, C.B.; Jesse, C.R.; Donato, F.; Del Fabbro, L.; de Gomes, M.G.; Goes, A.T.R.; Souza, L.C.; Boeira, S.P. Chrysin promotes attenuation of depressive-like behavior and hippocampal dysfunction resulting from olfactory bulbectomy in mice. Chem. Biol. Interact. 2016, 260, 154–162. [Google Scholar] [CrossRef]
  170. Yoon, J.H.; Youn, K.; Ho, C.T.; Karwe, M.V.; Jeong, W.S.; Jun, M. p-Coumaric acid and ursolic acid from corni fructus attenuated β-Amyloid25–35-induced toxicity through regulation of the NF-κB signaling pathway in PC12 cells. J. Agric. Food Chem. 2014, 62, 4911–4916. [Google Scholar] [CrossRef] [PubMed]
  171. Abdel-Moneim, A.; Yousef, A.I.; El-Twab, A.; Sanaa, M.; Abdel Reheim, E.S.; Ashour, M.B. Gallic acid and p-coumaric acid attenuate type 2 diabetes-induced neurodegeneration in rats. Metab. Brain Dis. 2017, 32, 1279–1286. [Google Scholar] [CrossRef]
  172. Ekinci-Akdemir, F.N.; Gülçin, I.; Gürsul, C.; Alwasel, S.H.; Bayir, Y. Effect of p-coumaric acid against oxidative stress induced by cisplatin in brain tissue of rats. J. Anim. Plant Sci. 2017, 27, 1560–1564. [Google Scholar]
  173. Sakamula, R.; Thong-Asa, W. Neuroprotective effect of p-coumaric acid in mice with cerebral ischemia reperfusion injuries. Metab. Brain Dis. 2018, 33, 765–773. [Google Scholar] [CrossRef]
  174. Chhillar, R.; Dhingra, D. Antidepressant-like activity of gallic acid in mice subjected to unpredictable chronic mild stress. Fundam. Clin. Pharmacol. 2013, 27, 409–418. [Google Scholar] [CrossRef] [PubMed]
  175. Pemminati, S.; Shetty, B.S.; Gopalakrishna, H.N.; Bethi, Y.; Rao, D.; Udaykumar, J.; Rai, A.; Shenoy, A.K. Evaluation of antidepressant activity of gallic acid in mice. Res. J. Pharm. Biol. Chem. Sci. 2014, 5, 575–580. [Google Scholar]
  176. Can, Ö.D.; Turan, N.; Özkay, Ü.D.; Öztürk, Y. Antidepressant-like effect of gallic acid in mice: Dual involvement of serotonergic and catecholaminergic systems. Life Sci. 2017, 190, 110–117. [Google Scholar] [CrossRef] [PubMed]
  177. Bortolotto, V.C.; Pinheiro, F.C.; Araujo, S.M.; Poetini, M.R.; Bertolazi, B.S.; Mariane Trindade de Paula, M.; Meichtry, L.B.; Polet de Almeida, F.; Shanda de Freitas, C.; Jesse, C.R.; et al. Chrysin reverses the depressive-like behavior induced by hypothyroidism in female mice by regulating hippocampal serotonin and dopamine. Eur. J. Pharmacol. 2018, 822, 78–84. [Google Scholar] [CrossRef] [PubMed]
  178. Rezai-Zadeh, K.; Ehrhart, J.; Bai, Y.; Sanberg, P.R.; Bickford, P.; Tan, J.; Shytle, R.D. Apigenin and luteolin modulate microglial activation via inhibition of STAT1-induced CD40 expression. J. Neuroinflamm. 2008, 5, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Shahamat, Z.; Abbasi-Maleki, S.; Motamed, S.M. Evaluation of antidepressant-like effects of aqueous and ethanolic extracts of Pimpinella anisum fruit in mice. Avicenna J. Phytomed. 2016, 6, 322. [Google Scholar] [PubMed]
  180. Li, R.; Wang, X.; Qin, T.; Qu, R.; Ma, S. Apigenin ameliorates chronic mild stress-induced depressive behavior by inhibiting interleukin-1β production and NLRP3 inflammasome activation in the rat brain. Behav. Brain Res. 2016, 296, 318–325. [Google Scholar] [CrossRef] [PubMed]
  181. Zhang, X.; Bu, H.; Jiang, Y.; Sun, G.; Jiang, R.; Huang, X.; Duan, H.; Huang, Z.; Wu, Q. The antidepressant effects of apigenin are associated with the promotion of autophagy via the mTOR/AMPK/ULK1 pathway. Mol. Med. Rep. 2019, 20, 2867–2874. [Google Scholar] [CrossRef]
  182. Balez, R.; Steiner, N.; Engel, M.; Muñoz, S.S.; Lum, J.S.; Wu, Y.; Wang, D.; Vallotton, P.; Sachdev, P.; O’Connor, M.; et al. Neuroprotective effects of apigenin against inflammation, neuronal excitability and apoptosis in an induced pluripotent stem cell model of Alzheimer’s disease. Sci. Rep. 2016, 6, 31450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Kashyap, D.; Sharma, A.; Tuli, H.S.; Sak, K.; Garg, V.K.; Buttar, H.S.; Setzer, W.N.; Sethi, G. Apigenin: A natural bioactive flavone-type molecule with promising therapeutic function. J. Funct. Foods 2018, 48, 457–471. [Google Scholar] [CrossRef]
  184. Nakazawa, T.; Yasuda, T.; Ueda, J.; Ohsawa, K. Antidepressant-like effects of apigenin and 2,4,5-trimethoxycinnamic acid from Perilla frutescens in the forced swimming test. Biol. Pharm. Bull. 2003, 26, 474–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Yi, L.T.; Li, J.M.; Li, Y.C.; Pan, Y.; Xu, Q.; Kong, L.D. Antidepressant-like behavioral and neurochemical effects of the citrus-associated chemical apigenin. Life Sci. 2008, 82, 741–751. [Google Scholar] [CrossRef] [PubMed]
  186. Li, R.; Zhao, D.; Qu, R.; Fu, Q.; Ma, S. The effects of apigenin on lipopolysaccharide-induced depressive-like behavior in mice. Neurosci. Lett. 2015, 594, 17–22. [Google Scholar] [CrossRef] [PubMed]
  187. Weng, L.; Guo, X.; Li, Y.; Yang, X.; Han, Y. Apigenin reverses depression-like behavior induced by chronic corticosterone treatment in mice. Eur. J. Pharmacol. 2016, 774, 50–54. [Google Scholar] [CrossRef] [PubMed]
  188. Anusha, C.; Sumathi, T.; Joseph, L.D. Protective role of apigenin on rotenone induced rat model of Parkinson’s disease: Suppression of neuroinflammation and oxidative stress mediated apoptosis. Chem. Biol. Interact. 2017, 269, 67–79. [Google Scholar] [CrossRef] [PubMed]
  189. Jang, S.W.; Liu, X.; Yepes, M.; Shepherd, K.R.; Miller, G.W.; Liu, Y.; Wilson, W.D.; Xiao, G.; Blanchi, B.; Sun, Y.E.; et al. A selective TrkB agonist with potent neurotrophic activities by 7, 8-dihydroxyflavone. Proc. Natl. Acad. Sci. USA 2010, 107, 2687–2692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Oboh, G.; Ogunsuyi, O.B.; Ogunbadejo, M.D.; Adefegha, S.A. Influence of gallic acid on α-amylase and α-glucosidase inhibitory properties of acarbose. J. Food Drug Anal. 2016, 24, 627–634. [Google Scholar] [CrossRef]
  191. Wurzelmann, M.; Romeika, J.; Sun, D. Therapeutic potential of brain-derived neurotrophic factor (BDNF) and a small molecular mimics of BDNF for traumatic brain injury. Neural Regen. Res. 2017, 12, 7. [Google Scholar]
  192. Rahvar, M.; Owji, A.A.; Mashayekhi, F.J. Effect of quercetin on the brain-derived neurotrophic factor gene expression in the rat brain. Bratisl. Lek. Listy 2018, 119, 28–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Caviedes, A.; Lafourcade, C.; Soto, C.; Wyneken, U. BDNF/NF-κB Signaling in the Neurobiology of Depression. Curr. Pharm. Des. 2017, 23, 3154–3163. [Google Scholar] [CrossRef] [PubMed]
  194. Kowiański, P.; Lietzau, G.; Czuba, E.; Waśkow, M.; Steliga, A.; Moryś, J. BDNF: A key factor with multipotent impact on brain signaling and synaptic plasticity. Cell. Mol. Neurobiol. 2018, 38, 579–593. [Google Scholar] [CrossRef] [PubMed]
  195. Borsato, D.M.; Prudente, A.S.; Doell-Boscardin, P.M.; Borsato, A.V.; Luz, C.F.; Maia, B.H.; Cabrini, D.A.; Otuki, M.F.; Miguel, M.D.; Farago, P.V.; et al. Topical anti-inflammatory activity of a monofloral honey of Mimosa scabrella provided by Melipona marginata during winter in Southern Brazil. J. Med. Food 2014, 17, 817–825. [Google Scholar] [CrossRef]
  196. Kaster, M.P.; Gadotti, V.M.; Calixto, J.B.; Santos, A.R.; Rodrigues, A.L.S. Depressive-like behavior induced by tumor necrosis factor-α in mice. Neuropharmacology 2012, 62, 419–426. [Google Scholar] [CrossRef]
  197. Tavares, R.G.; Schmidt, A.P.; Abud, J.; Tasca, C.I.; Souza, D.O. In vivo quinolinic acid increases synaptosomal glutamate release in rats: Reversal by guanosine. Neurochem. Res. 2005, 30, 439–444. [Google Scholar] [CrossRef]
  198. McNally, L.; Bhagwagar, Z.; Hannestad, J. Inflammation, glutamate, and glia in depression: A literature review. CNS Spectr. 2008, 13, 501–510. [Google Scholar] [CrossRef]
  199. Dantzer, R.; O’Connor, J.C.; Lawson, M.A.; Kelley, K.W. Inflammation-associated depression: From serotonin to kynurenine. Psychoneuroendocrinology 2011, 36, 426–436. [Google Scholar] [CrossRef] [Green Version]
  200. Yu, S.; Holsboer, F.; Almeida, O.F. Neuronal actions of glucocorticoids: Focus on depression. J. Steroid Biochem. Mol. Biol. 2008, 108, 300–309. [Google Scholar] [CrossRef]
  201. Zappella, M.; Biamonte, F.; Balzamino, B.O.; Manieri, R.; Cortes, M.; Santucci, D.; Di Stasio, E.; Rizzuto, M.; Micera, A. Relaxation Response in Stressed Volunteers: Psychometric Tests and Neurotrophin Changes in Biological Fluids. Front. Psychiatry 2021, 12, 790. [Google Scholar] [CrossRef]
  202. Sadighparvar, S.; Darband, S.G.; Yousefi, B.; Kaviani, M.; Ghaderi-Pakdel, F.; Mihanfar, A.; Babaei, G.; Mobaraki, K.; Majidinia, M. Combination of quercetin and exercise training attenuates depression in rats with 1,2-dimethylhydrazine-induced colorectal cancer: Possible involvement of inflammation and BDNF signalling. Exp. Physiol. 2020, 105, 1598–1609. [Google Scholar] [CrossRef] [PubMed]
  203. Zhang, J.C.; Yao, W.; Hashimoto, K. Brain-derived Neurotrophic Factor (BDNF)-TrkB Signaling in Inflammation-related Depression and Potential Therapeutic Targets. Curr. Neuropharmacol. 2016, 14, 721–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Raison, C.L.; Felger, J.C.; Miller, A.H. Inflammation and treatment resistance in major depression: A perfect storm. Psychiatr. Times 2013, 30, 17. [Google Scholar]
  205. Strawbridge, R.; Young, A.H.; Cleare, A.J. Inflammation as a marker of clinical response to treatment: A focus on treatment-resistant depression. In Inflammation and Immunity in Depression; Academic Press: Cambridge, MA, USA, 2018; pp. 473–487. [Google Scholar]
  206. Strawbridge, R.; Hodsoll, J.; Powell, T.R.; Hotopf, M.; Hatch, S.L.; Breen, G.; Cleare, A.J. Inflammatory profiles of severe treatment-resistant depression. J. Affect. Disord. 2019, 246, 42–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Husain, M.I.; Strawbridge, R.; Stokes, P.R.; Young, A.H. Anti-inflammatory treatments for mood disorders: Systematic review and meta-analysis. J. Psychopharmacol. 2017, 31, 1137–1148. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Summary of the mechanism of action for inflammation leading to depression.
Figure 1. Summary of the mechanism of action for inflammation leading to depression.
Molecules 27 05091 g001
Figure 2. Summary of the anti-depressive effects of SBH.
Figure 2. Summary of the anti-depressive effects of SBH.
Molecules 27 05091 g002
Table 1. Phenolic compounds that showed therapeutic effects on psychiatric and neurological disorders.
Table 1. Phenolic compounds that showed therapeutic effects on psychiatric and neurological disorders.
TypeCompoundsTherapeutic Effects
Phenolic acidsp-Coumaric acid
Molecules 27 05091 i001
Anti-inflammatoryAntidepressantAntioxidant
[49,170,171][49][171,172,173]
Gallic acid
Molecules 27 05091 i002
[171][174,175,176][171,174]
Caffeic acid
Molecules 27 05091 i003
[149]
FlavonoidsChrysin
Molecules 27 05091 i004
[168,169][177][168,169,177]
Apigenin
Molecules 27 05091 i005
[178,179,180,181,182,183][180,181,184,185,186,187][180,183,188]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zakaria, F.H.; Samhani, I.; Mustafa, M.Z.; Shafin, N. Pathophysiology of Depression: Stingless Bee Honey Promising as an Antidepressant. Molecules 2022, 27, 5091. https://doi.org/10.3390/molecules27165091

AMA Style

Zakaria FH, Samhani I, Mustafa MZ, Shafin N. Pathophysiology of Depression: Stingless Bee Honey Promising as an Antidepressant. Molecules. 2022; 27(16):5091. https://doi.org/10.3390/molecules27165091

Chicago/Turabian Style

Zakaria, Fatin Haniza, Ismail Samhani, Mohd Zulkifli Mustafa, and Nazlahshaniza Shafin. 2022. "Pathophysiology of Depression: Stingless Bee Honey Promising as an Antidepressant" Molecules 27, no. 16: 5091. https://doi.org/10.3390/molecules27165091

Article Metrics

Back to TopTop