Metabolism and physiological effects of SCFAs
SCFAs are monocarboxylic acids with two to five carbon atoms derived from the fermentation of dietary carbohydrates in the host intestine.18 The vast majority of circulating SCFAs are derived from gut microbial fermentation and absorbed in the colon.63 Diet, microbial composition and gut transit time have an impact on the fermentation of dietary fibres. Humans generally produce approximately 500–600 mmol of SCFAs per day. SCFAs mainly consist of acetate (C2), propionate (C3) and butyrate (C4) in a ratio of 60:20:20.64 Multiple gut bacteria with diverse genetic potentials participate in the anaerobic fermentation of different SCFAs. In this regard, acetogenic commensal bacteria in the gut synthesise acetate from CO2 and as an electron source, where Bacteroidetes and Firmicutes preferentially generate propionate, and Firmicutes (Eubacterium, Anaerostipes, Roseburia and Faecalibacterium prausnitzii) predominantly synthesise butyrate and degrade indigestible polysaccharide molecules.65–68 Colonocytes take up the majority of SCFAs through H+-linked monocarboxylate transporters and sodium-linked monocarboxylate transporters and use them to maintain intestinal barrier function.69 Most SCFAs are transmitted to hepatocytes through portal circulation and are metabolised as an energy source.70 Only a fraction of acetate, propionate and butyrate reach the systemic circulation and exert physiological effects elsewhere.63
SCFAs in the systemic circulation can cross the blood-brain barrier (BBB) and have a pivotal impact on the microbiota-gut-brain crosstalk.71 72 The high expression of monocarboxylate transporters in endothelial cells facilitates the penetration of SCFA through the BBB.73 In human cerebrospinal fluid, the concentration of SCFAs is as follows at physiological levels: 0–171 µM acetate, 0–6 µM propionate and 0–2.8 µM butyrate.70 SCFAs can also regulate the transfer of nutrients and molecules involved in the maintenance of BBB integrity, directly influencing brain development and CNS homeostasis.74 Additionally, SCFAs mediate a plethora of basic behavioural and neurological processes through the modulation of the immune system, HPA axis and tryptophan metabolism, along with the synthesis of several metabolites such as neurotransmitters that have neuroactive properties.75–77 A disease-promoting imbalance in the human gastrointestinal tract can change the composition and abundance of the gut microbiota and alter the normal formation of metabolites, which elicits disease development or severity.
The role of SCFAs in the pathological mechanisms underlying gut microbiota-associated depression
SCFAs are the major mediators of the microbiota-gut-brain axis in the pathophysiology of depression. Chronic stress, along with gut dysbiosis, interferes with the metabolism of SCFAs and accelerates the dysfunction of the microbiota-gut-brain axis in depression. SCFAs have a neuroprotective effect and participate in the complex biological mechanisms and pathological processes involved in the onset and progression of depression, including chronic cerebral hypoperfusion (CCH), neuroinflammation, epigenetic modifications and neuroendocrine alterations (figure 1).
Figure 1Microbiota-gut-brain axis and central nervous system dysfunctions in depression. The interaction of the microbiota-gut-brain axis and depression are involved in the neural, endocrine and immune systems. Chronic stress induces a series of psychopathological processes including chronic cerebral hypoperfusion, neuroinflammation, epigenetic alteration, hypothalamic-pituitary-adrenal (HPA) axis and vagus activation through gut microbiota and its metabolites. SCFA, short-chain fatty acid.
SCFAs and chronic cerebral hypoperfusion
CCH has emerged as a major contributor to cognitive decline and degenerative processes that predispose the brain to mental disorders.78 CCH induces inadequate transportation of nutrients and oxygen in cerebral cells and aggravates brain injury, including BBB dysfunction,79 metabolic disturbance,80 activated neuroinflammation81 and neuroendocrine perturbation.82 In turn, the adverse consequences of repeated hypoperfusion/hypoxic events underline chronic cognitive and behavioural impairments (eg, memory deficits, anxiety and depression).83 Previous studies have indicated that CCH is closely associated with depression in animal models and humans.84 85
Bilateral common carotid artery occlusion (BCCAO) is adopted to model CCH-induced depressive behaviours in rats.85 Several recent studies have assessed the role of SCFAs in CCH-induced depression. Xiao et al
86 demonstrated that rats with BCCAO exhibit compromised gut barrier function and perturbed gut microbiota. The decrease in representative SCFA-producing flora results in hippocampal SCFA reduction in BCCAO rats that suffer from cognitive impairment and show depressive-like behaviours.86 Subsequently, rats were supplied with water containing a mixture of SCFAs (acetate, propionate and butyrate). SCFAs attenuated BCCAO-induced hippocampal neuroinflammation and neuronal apoptosis through the inhibition of nuclear factor kappa-B (NF-κB) and activation of the Erk1/2 pathway, accompanied by amelioration of cognitive decline and degenerative processes.86 NF-κB is a key transcription factor that regulates the expression of genes involved in immunity and inflammation. It should be noted that the intervention of oral SCFA intake could not be normalised in each animal, and it also elevates sodium intake. Hence, rebuilding the gut microbiota by faecal microbiota transplantation (FMT) appears to be a more reasonable method to replenish these beneficial metabolites.
Reestablishment of the gut microbiome has emerged as a potential approach for treating CCH-induced depression and cognitive impairment. Restoring balanced gut flora in BCCAO rats through FMT restores gastrointestinal motility and ameliorates hippocampal neuronal apoptosis and cognitive dysfunction.87 88 Nevertheless, it is still difficult to confirm whether FMT and SCFAs exert a protective or therapeutic effect on CCH or both. FMT may not eradicate a disease by itself, but it can replenish SCFAs and alleviate secondary impairment induced by the dysfunction of the microbiota-gut-brain axis.
SCFAs and neuroinflammation
The essential role of inflammation in depression has been widely investigated in recent decades, with the microbiota-gut-brain axis emerging as a key intermediate regulator.89–92 Gut dysbiosis affects the onset and maintenance of neuroinflammation in depression by altering SCFA metabolism.93 In chronic mild stress mouse models, physiological stress initiates alterations in the gut microbiota characterised by significantly decreased faecal microbial diversity.94 Rats with depressive-like behaviours exhibit a decreased relative abundance of SCFA-producing gut microbes and reduced serum levels of SCFAs, which are closely associated with cytokine expression and inflammatory pathway activation in neuroinflammation.95
The levels of cytokines in the blood, including interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α), are significantly higher in patients with depression than in healthy controls.96 These pro-inflammatory cytokines, particularly IL-6, IL-1β and TNF-α, promote the generation of T helper 17 (Th17) cells, which are strongly implicated in depression and other CNS diseases.97 Th17 cells may come in two ways: peripheral Th17 cells, which are probably released from the lamina propria of the small intestine, can directly infiltrate the brain parenchyma through the BBB leakage induced by Th17-derived cytokines,98 or they may originate from CD4+ T cells in the brain when stimulated by pro-inflammatory cytokines.99 In turn, the products of Th17 cells, interferon-γ (IFN-γ) and IL-17A, could promote neuroinflammation. IFN-γ has been proposed to confer a signal for Th17 infiltration in the CNS and enhance synapse elimination.100 IFN-γ and IL-17A contribute to the proliferation, polarisation and activation of microglia.101 102 Microglia are a major source of cytokines among all glial cells in the CNS.103 Microglia polarise to the M1 phenotype and release reactive oxygen species (ROS) and pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α.104 ROS, such as hydroxyl radicals (OH−), hydrogen peroxide (H2O2) and the superoxide anion (O2
−), impair neurogenesis, synaptic plasticity and neuronal transmission as a consequence of oxidative stress-induced mitochondrial injury and neuronal apoptosis.105 In summary, neuroinflammation mediated by pro-inflammatory cytokines evokes a detrimental response in the CNS that aggravates depression.
Apart from indirect mechanisms via chemical mediators, the onset of neuroinflammation is also activated through direct pathophysiological mechanisms via the vagus nerve pathway.106 The vagus nerve can stimulate infiltrated or resident immune cells (eg, microglia and dendritic cells) in the vicinity of the perineural sheath. Activated immune cells boost the immune response by relaying signals from pro-inflammatory cytokines to the CNS, which induces a series of physiological and behavioural responses.107 Pro-inflammatory cytokines in the gastrointestinal tract stimulate the vagus nerve and activate the HPA axis, which influences the central stress circuitry.108 In contrast, stimulation from cholinergic signalling enables the vagus nerve to decrease the inflammatory response and production of pro-inflammatory cytokines.109 Several studies revealed a key role of the subdiaphragmatic vagus nerve in the onset of depression by regulating the brain-gut microbiota axis.107 110 Additionally, the subdiaphragmatic vagotomy exhibited an antidepressant effect in lipopolysaccharide administration mice, which indicated an essential role of the vagus nerve in depression.111 Apart from subdiaphragmatic vagotomy, vagus nerve stimulation has been developed as an emerging treatment for depression.112 113 These results indicate the potential role of the vagus nerve in depression treatment.
Gut microbiota-derived SCFAs play a vital role in anti-inflammatory actions owing to their maintenance of gut-brain permeability and constant input to the CNS for the homeostasis of microglia.114 115 SCFAs decrease the permeability of both the intestinal barrier and the BBB. SCFAs, especially acetate, propionate and butyrate, have been reported to exhibit protective effects on the intestinal barrier as energy substrates.116 Butyrate enhances the expression of aryl hydrocarbon receptors and hypoxia-inducible factor 1α, which upregulates the levels of IL-12 through regulating the mammalian target of rapamycin and signal transducer and activator of transcription 3.117 IL-12 is a protective cytokine that contributes to resisting inflammatory stimuli and maintaining intestinal homeostasis. These findings suggested that SCFAs have an anti-inflammatory effect on maintaining gut homeostasis. In the CNS, propionate preserves the BBB through FFAR3 on the surface of endothelial cells.118 SCFAs protect hippocampal neurons from injury to the mitochondrial membrane potential and ROS accumulation.87 In addition, the regulatory function of SCFAs protects the CNS from peripheral inflammatory cytokines and toxic substances. SCFAs have a direct anti-inflammatory effect on microglial proliferation and activation by binding FFARs, as well as activating FFARs on neutrophils and dendritic cells to alleviate systemic inflammation.119 120 SCFAs bind to GPR41 on microglia, regulate microglial proliferation and inflammatory status and inhibit pro-inflammatory signalling pathways via NF-κB inhibition and Erk1/2 activation.86 121 Butyrate also activates GPR109A-mediated signalling pathways to downregulate NF-κB signalling in microglia. The inhibition of pro-inflammatory enzymes (inducible nitric oxide synthase and cyclooxygenase-2) and pro-inflammatory cytokine (TNF-α, IL-1β and IL-6) production in microglia prevents the onset and process of neuroinflammation.122 IL-10 and CD26, markers reflecting the anti-inflammatory status of microglia, are increased after butyrate treatment, indicating the neuroprotective effects of SCFAs in vivo.123 The intricate interactions between neuroinflammation and anti-inflammation through the SCFA-mediated microbiota-gut-brain axis are involved in the pathophysiology of depression (figure 2).
Figure 2The neuroinflammation and short-chain fatty acids in depression. Cytokines pass through the impaired BBB and promote the brain CD4+ T cells to the Th17 cells and peripheral Th17 cell infiltration. Th17 cells activate microglia and release pro-inflammatory cytokines and ROS, which induce the onset of neuroinflammation. SCFAs could inhibit microglia activation by inhibiting the NF-κB pathway and play an important role in anti-neuroinflammation processes. ACh, acetylcholine; BBB, blood-brain barrier; FFARs, free fatty acid receptors; IFN-γ, interferon-γ; IL-12, interleukin-12; IL-17A, interleukin-17A; IL-6, interleukin-6; NF-κB, nuclear factor kappa-B; ROS, reactive oxygen species; SCFAs, short-chain fatty acids; Th17, T helper 17 cells; TLRs, toll-like receptors; TNF, tumour necrosis factor.
SCFAs and host epigenome
The epigenome describes the heritable and reversible alterations of the genome that change DNA packaging or associated proteins without affecting nucleotide sequences.124 Epigenetic modifications include histone modifications, DNA methylation and non-coding RNAs (ncRNAs). These modifications integrate abundant environmental messages into phenotypes by regulating the three-dimensional architecture of chromatin. Therefore, epigenetic modifications have been implicated in the pathogenesis of depression, manipulation of the HPA axis, neuroinflammation, excitatory-inhibitory balance and monoamine pathways.125–129 Epigenetic pathways, especially DNA methylation and histone modifications, have been extensively investigated in depression and have been proven to be tightly correlated with the metabolism of the gut microbiota.130 SCFAs from the gastrointestinal tract also have a vital effect on epigenetic modifications as mediators in the CNS of patients with depression. In this section, we introduce the molecular mechanisms and interactions between the gut microbiota, SCFAs and host epigenome in depression. Figure 3 shows the neuroprotective effect of SCFAs through histone acetylation and DNA methylation in depression.
Figure 3SCFAs alleviate depressive symptoms by regulating epigenetic modifications in the CNS. SCFAs, especially propionate and butyrate, regulate the DNA methylation and histone acetylation in the neural cells and microglia. The decreased DNA methylation promotes BDNF synthesis and inhibits neuronal apoptosis. The increased histone acetylation decreases pro-inflammatory cytokines and inhibits microglia activation. These modifications elicited by SCFAs jointly decrease depressive symptoms. 5hmc, 5-hydroxymethylcytosine; 5mc, 5-methylcytosine; Ac, acetylation; BDNF, brain-derived neurotrophic factor; CD26, dipeptidyl peptidase 4; FFARs, free fatty acid receptors; H3K9, histone 3 lysine 9; HDACs, histone deacetylases; IL-10, interleukin-10; Me, methylation; RNA PII, RNA polymerase II; SCFAs, short-chain fatty acids; TETs, Ten-eleven translocation enzymes.
Histone modifications
Histones are the basic elements of nucleosomes. An octamer of two distinct copies of four types of histones (H2A, H2B, H3 and H4) wrapped with long DNA chains composes the nucleosome core particles. The -NH2 terminus of histone tails can be reversibly modified by several functional groups, such as acetyl and methyl groups, and the accessibility of genomic DNA is altered due to the disruption of electrostatic interactions between the modified residues and nucleic acids, thereby manipulating cell activities (eg, DNA replication, DNA repair, transcriptional activities and cell cycle progression).131 The addition and deletion of specific functional groups are regulated by antagonistic enzymes. For instance, histone acetyltransferases and histone deacetylases (HDACs) jointly control the addition or deletion of acetyl groups on the histone tails.132 HDACs could be divided into five major families, including class I (HDACs 1, 2, 3, 8), class IIA (HDACs 4, 5, 7, 9), class IIB (HDAC 6, 10), class III (SIRT1–7) and class IV (HDAC11), and the class I members play a key role in the initiation and progression of depression.
Epigenetic modifications in the CNS, particularly histone acetylation, are closely associated with the activation of glia and immune cells during the pro-inflammatory response.133 134 These modifications control microglial polarisation and inflammatory cytokine expression.123 Several studies have demonstrated the role of HDAC in the immune response, which epigenetically modulates gene expression and the inflammatory response.135 SCFAs are well-known HDAC inhibitors (HDACi), together with histone acetyltransferases, that upgrade histone and non-histone protein acetylation.136 Among the SCFAs, butyrate is the most potent and extensively applied in neuropsychiatric studies as a class I and class II HDACi. Butyrate induces an increased level of histone acetylation and upregulation of brain-derived neurotrophic factor (BDNF) levels in the corticolimbic portions of the mouse frontal cortex.137 BDNF is an important protein of growth factor, which plays a crucial role in neuronal growth and synaptic plasticity in brain development.138 139 Patients with depression show decreased levels of BDNF in the serum and cerebrospinal fluid.140 141 Bifidobacterium longum NCC3001, a SCFA-producing bacteria, has been demonstrated to upregulate BDNF, regulate neuronal plasticity in the enteric nerve and alleviate depression-like behaviours in mice.142 Moreover, fluoxetine (a selective serotonin reuptake inhibitor antidepressant) combined with butyrate substantially decreased behavioural despair compared with fluoxetine treatment alone, with an increasing transcript level of BDNF,137 suggesting that upregulation of BDNF expression might be important for alleviating depressive behaviours.
Propionate or butyrate treatment has been reported to promote histone 3 lysine 9 acetylation in the brain, with decreased HDAC1 expression in microglia.143 SCFA treatment reduces microglial activation and the levels of pro-inflammatory cytokines, including TNF‐α, IL‐1α and IL‐1β, and represses neuronal apoptosis and neurodegeneration to alleviate depressive-like behaviours.143 Lactate, an important intermediate of SCFAs, has reversible effects on stress-induced depression.144 The effects of lactate on HDACs have also been studied through distinct epigenetic mechanisms. In mouse models, lactate treatment increased the levels of hippocampal class I HDAC, especially HDAC2/3, enhanced resilience to stress and rescued social avoidance and anxiety.145 These results indicate that class I HDACs could be potential targets for reversing chronic unpredictable stress-induced depressive-like behaviours, and natural compound SCFAs may serve as antidepressants owing to their high security. However, further research has indicated the opposite roles of class I HDACs during different periods of depression. Treatment with the HDAC2/3 inhibitor CI-994 inhibited the activities of HDAC2/3 and alleviated depression-like behaviours in mice.145 HDAC2 and HDAC3 play different roles in the progression of depression. Thus, it remains difficult to elucidate the pathways by which lactate regulates HDAC2/3 during different periods of depression. These inconsistent results demonstrated that targeting HDAC2/3 may only serve as a therapeutic antidepressant, but not as a prophylactic.
DNA methylation
DNA methylation involves the covalent addition of a methyl group to the pyrimidine ring of cytosines in CpG regions, which appear in approximately 70% of the gene promoters.146 DNA methylation is also a dynamic and reversible process similar to histone modifications, whereas methylation in CpG regions transforms the genes into an inaccessible state and represses transcriptional activities. DNA methyltransferases (DNMTs) catalyse DNA methylation by transferring the methyl group from S-adenyl methionine to carbon-5 of cytosine to form 5-methyl cytosine.147 There are five major categories of DNMTs in mammals: DNMT1, DNMT2, DNMT3a, DNMT3b and DNMT3L. Ten-eleven translocation (TET) enzymes are involved in the demethylation of CpG regions by oxidising methylated bases, which activate gene expression and transcriptional activities.
Depression has been linked to alterations in BDNF, which influences GABA neurotransmission, synaptic plasticity and neuronal activity.147 148 Patients with depression have a higher level of methylation at specific CpG regions within BDNF promoters than healthy controls, which results in decreased expression of BDNF in the hippocampus.149 Further research has investigated the effects of SCFAs on the methylation of BDNF promoters. Butyrate treatment in depressed mice restored the decreased TET1 level, which promoted the hydroxylation of the cytosine residue to 5-hydroxymethylcytosine, reduced DNA methylation and facilitated BDNF transcription, exerting antidepressant-like effects.150 These results link neural activities to the metabolism of the gut microbiome through epigenetic alterations, which may be crucial to the biological mechanisms underlying depression and potential therapeutic strategies.
Non-coding RNA
ncRNAs, including long non-coding RNAs (lncRNAs), circular RNAs and microRNAs (miRNAs), are significant mediators of normal transcriptional and translational processes, and their abnormal metabolism has been extensively reported in the pathogenesis of depression.151–153 However, interactions between the gut microbiome and ncRNAs in the CNS remain largely unknown.
The competitive endogenous RNA (ceRNA) hypothesis indicates that miRNAs compete for shared miRNA-binding sites in post-transcriptional control, which is the key for neurodevelopment and neurodegeneration.154 Recently, aberrant expression of ncRNAs was observed in the hippocampus of mice with gut dysbiosis. An integrated analysis found two lncRNA-miRNA-mRNA ceRNA regulatory networks in depressed mice induced by gut dysbiosis.155 Regulatory networks are primarily associated with inflammatory responses and the neurodevelopmental regulation of microbial-related pathological changes. One consisted of two lncRNAs (4930417H01Rik and AI480526), one miRNA (mmu-miR-883b-3p) and two mRNAs (Adcy1 and Nr4a2). The other consists of six lncRNAs (5930412G12Rik, 6430628N08Rik, A530013C23Rik, A930007I19Rik, Gm15489 and Gm16251), one miRNA (mmu-miR-377-3p) and three mRNAs (Six4, Stx16 and Ube3a).155 These molecules have been proposed as pivotal mediators of ncRNA adjustment in the microbiota-gut-brain axis, and have the potential to be applied as epigenetic biomarkers in gut microbial-related depression.
Although emerging evidence has revealed the crosstalk between ncRNAs and the gut microbiota in depression, it is still challenging to make a conclusive narrative about how alterations of the microbiota-gut-brain axis mediate ncRNA activities in the CNS. There are several limitations in the current investigations of ncRNAs to form homogeneous groups compared with histone modifications and DNA methylation. For instance, ncRNA levels tend to be affected by multiple factors, including environment, diet, exercise, ageing, alcohol consumption and drug abuse. Despite the clear implication, convincing evidence is still lacking to demonstrate how certain ncRNAs mediate specific signalling pathways in the microbiota-gut-brain axis, thereby influencing depression. Hence, unlike histone modifications and DNA methylation, it is difficult to construct a generalised framework to depict ncRNA regulation by gut microbial metabolites.
In sum, depression can be considered a slowly developing but multifaceted maladaptation to long-term environmental stressors.156 Maladaptive neuronal plasticity in regions implicated in depression pathogenesis, such as the hippocampus, has been shown to be relevant to allostasis of the brain through epigenetic pathways. Delayed addition or deletion of epigenetic modifications may explain the slow development and insignificant initial effects of antidepressants in treating depression. Disrupted gut homeostasis induces altered epigenetic regulators synthesised by the gut microbiota, such as acetate, butyrate and propionate, following the activation of neuroinflammation and other pathways by epigenetic reprogramming. The underlying mechanisms in the microbiota-gut-brain axis may play an important role in the long-term abnormal synaptic plasticity and behavioural response to stress in depression.
SCFAs and neuroendocrine alterations
SCFAs participate in modulating the metabolism of multiple neuroactive substances in the gastrointestinal and nervous systems. SCFAs ligate to G protein-coupled receptors, for example, GPR43 (FFAR2), GPR41 (FFAR3), GPR109A, OLFR78 and OR51E2, located on the surface of intestinal epithelial cells and enteroendocrine cells.157–160 In addition to serving as energy supplements for epithelial cells, SCFAs strengthen intestinal barrier integrity by enhancing glucose metabolism, lipid homeostasis and the expression of tight junction protein to promote gut health.161 SCFAs, especially butyrate, increase the expression of claudin-1 and induce the redistribution of occludin and zonula occludens-1 to repair and enhance gut barrier function.162 Furthermore, they induce the release of hormones and neuropeptides, such as glucagon-like peptide 1 and peptide YY from intestinal enteroendocrine cells and regulate gastric motility and digestive absorption, which might activate a signalling cascade that affects brain circuits related to appetite and food intake.163 164 Taken together, these findings suggest that SCFAs play a crucial role in maintaining energy metabolism and intestinal homeostasis.
In the microbiota-gut-brain axis, SCFAs are involved in the synthesis and release of peripheral neurotransmitters, including acetylcholine and 5-HT.165 166 The enterochromaffin cells in the gut synthesise >90% of the circulating 5-HT, which participates in regulating diverse gastrointestinal functions including motility and secretory reflexes.167 The stimulatory activities of SCFAs, especially acetate and butyrate, promote the expression of colonic tryptophan hydroxylase 1, the rate-limiting enzyme in 5-HT synthesis,168 suggesting that SCFAs are actively involved in host 5-HT biosynthesis and crosstalk between the microbiome and gut homeostasis. Owing to the permeability of the BBB, peripheral neurotransmitters may be unable to access the brain and directly affect the functioning of the CNS. However, serotonin in the peripheral blood circulation can regulate gastrointestinal movement and excretion and indirectly affect emotional, cognitive and behavioural responses through neuroendocrine pathways or vagal afferents.169 170 Some evidence also shows that SCFAs that are taken up through the BBB regulate the levels of neurotransmitters in the CNS. Intraperitoneal acetate resulted in altered glutamate, glutamine and GABA in the hypothalamus and increased anorexigenic neuropeptide expression.114 The disordered gut microbiota, especially Firmicutes, could regulate the levels of acetate, propionate and butyrate, which could elicit the metabolism disturbance of peripheral and central glycerophospholipid. As a result, the levels of tryptophan, 5-HT, 5-hydroxyindoleacetic acid and indoleacetic acid were decreased in the hippocampus.171 As mentioned above, the metabolic disturbance of SCFAs elicits activation of a series of inflammatory pathways with increased levels of pro-inflammatory cytokines and microglial proliferation and activation in the CNS. Meanwhile, the disturbance throws the normal biosynthesis of neuroprotective factors into confusion, such as BDNF. Ultimately, the levels of 5-HT are decreased due to neuronal apoptosis and metabolic dysfunction. Decreased circulating levels of 5-HT represent a classical biomarker indicating mental disorders like depression.172 These findings suggest that decreased levels of SCFAs can partly explain the altered expression of 5-HT and other neurotransmitters through the gut-brain axis in patients with depression.
SCFAs also alleviate stress-induced CNS dysfunction by ameliorating HPA axis activity. Specific pathogen-free mice receiving faecal microbiome colonisation at early postnatal life (3 weeks) instead of at a later stage exhibited reversed stress-associated physiological alterations on the HPA axis.173 This suggests that the gut microbiota is involved in the normal development of the HPA axis and neuroendocrine response to stress. In addition, after solely receiving SCFA administration, the expression of hypothalamic genes involved in the stress response is decreased, including corticotrophin-releasing factor and related receptors (corticotrophin-releasing factor receptor (CRFR) 1 and 2, mineralocorticoid receptor).174 The reactivity of the HPA axis leads to increased levels of serum cortisol, a stress hormone that increases access to energy stores and promotes protein and fat mobilisation.175 Colon-delivered SCFAs at high doses (174.2 mmol acetate, 13.3 mmol propionate and 52.4 mmol butyrate daily) and low doses (87.1 mmol acetate, 6.6 mmol propionate and 26.2 mmol butyrate daily) both increase the levels of serum SCFA and attenuate the cortisol response to acute psychosocial stress in healthy humans.176 It is speculated that SCFAs penetrating through the BBB are involved in HPA axis integration by rich innervations in and projections to the medial parvocellular paraventricular nucleus.176 These findings demonstrate the mechanisms by which SCFAs participate in the regulation of HPA axis responsiveness in depression.
Nowadays, ketamine, an N-methyl-D-aspartate receptor antagonist, has been proven effective against treatment-resistant depression due to its anti-inflammatory effect and regulation of neurotransmitters.177 Previous research also revealed alterations in the gut microbiota during ketamine treatment.178 Ketamine could amplify the Lactobacillus and maintain the level of Bacteroidales and Clostridiales at the genus level in mice.179 180 However, while the effect of ketamine on the composition and abundance of gut microbiota has been well illustrated, how ketamine regulates the metabolism of gut microbiota and its metabolites, such as SCFAs, is unclear. Hence, further studies should concentrate on the mechanisms by which ketamine regulates the metabolism of gut microbiota and its metabolites as a promising antidepression agent.