4.1.1. Inflammatory Bowel Disease
The interaction between SCFAs and IBD is multifaceted, involving the interplay among gut microbiota, immune responses, and the integrity of the gut epithelial barrier [
136,
137]. Butyrate, a primary energy source for colonocytes, exerts anti-inflammatory effects by inhibiting the activation of the nuclear factor kappa B and reducing proinflammatory gene expression [
138]. A decline in SCFA-producing bacteria characterizes IBD patients, notably butyrate producers like
Faecalibacterium prausnitizii and
Roseburia hominis [
139,
140,
141]. This results in reduced colonic SCFA levels linked to compromised gut barrier function in IBD [
142,
143].
SCFAs protect against IBD-associated intestinal inflammation through various mechanisms [
144]. They enhance the intestinal epithelial barrier by promoting mucus production and tightening tight junctions between epithelial cells [
144]. Additionally, SCFAs modulate immune responses by influencing the differentiation and function of Tregs, suppressing excessive immune reactions [
145]. Several pathways are involved in SCFA-mediated immune regulation, including GPCRs, HDACs, and the regulation of innate immune sensors like Toll-like receptors (TLRs) and Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. SCFAs inhibit the progression of IBD by regulating innate immune sensors, TLRs, and NLRP3 inflammasomes. SCFAs protect the intestinal barrier; acetate, propionate, and butyrate stimulate the intestinal NLRP3 inflammasome, increasing IL-18 secretion and enhancing intestinal barrier integrity [
146]. Moreover, SCFAs engage with GPR43 and GPR109A receptors essential for regulating intestinal immunity, stimulating the production of Treg. This has been demonstrated in preclinical studies, where controlling colonic Treg levels and function in a GPR43-dependent manner has been shown to mitigate inflammation, as seen in SCFA-mediated protection against colitis in GPR43-deficient (Gpr43(−/−)) mice [
147,
148]. Furthermore, SCFAs promote the differentiation of Tregs by inhibiting HDAC activity, and Tregs secrete protective cytokines, such as IL10, to suppress inflammation [
149]. SCFAs not only inhibit TLR signaling, but butyrate acts as an HDAC inhibitor to suppress TLR4 expression and the TLR2-mediated release of inflammatory factors [
150,
151,
152]. Finally, SCFAs participate in tissue repair processes within the gut, promoting the proliferation and differentiation of epithelial cells, thus facilitating the healing of damaged tissues caused by inflammation in IBD [
153].
A recent study investigated the utility of fecal SCFA concentrations as surrogate markers for gut microbiota diversity in patients with IBD and primary sclerosing cholangitis (PSC) [
154]. Results decreased fecal isobutyrate levels compared to healthy controls. Fecal acetate and butyrate positively correlated with fecal calprotectin and serum C-reactive protein in ulcerative colitis (UC) patients. Furthermore, UC patients with higher fecal calprotectin levels exhibited elevated fecal acetate, butyrate, and propionate levels. These findings suggest potential associations between SCFA levels and disease activity in UC patients.
Although SCFA concentrations are decreased in IBD patients, SCFA supplementation through diet or probiotics shows promise as an adjunct therapy, with minimal adverse effects reported [
139,
153,
155,
156,
157,
158]. However, the exact mechanisms underlying the therapeutic effects of SCFAs in IBD require further elucidation, highlighting the complexity of their relationship with the disease.
Figure 2 illustrates the mechanism of action of SCFAs.
4.1.2. Colorectal Cancer
Colorectal cancer (CRC) ranks among the top three causes of cancer-related mortality worldwide, with increasing recognition of the microbiota's contribution to its pathogenesis [
159]. Various factors contribute to CRC, including a high-fat diet, stress, antibiotics, synthetic food additives, a sedentary lifestyle, and environmental factors [
160]. High-fat diet, especially prevalent in Western diets featuring high red and processed meat consumption, high fructose corn syrup, and unhealthy cooking methods, significantly contributes to CRC [
161]. Current research has explored the protective role of dietary fibers in reducing the risk of CRC [
162,
163].
A systematic review and meta-analysis by Alvandi et al. explored the role of fecal SCFAs in CRC incidence and risk stratification [
164]. The study, encompassing seventeen case-control and six cross-sectional studies, revealed that individuals with lower concentrations of acetic, propionic, and butyric acid are at a higher risk of CRC. Although these findings suggest a potential association between decreased fecal SCFA concentrations and CRC susceptibility, emphasizing the importance of gut microbiota and bacterial metabolites in CRC prevention, their exact role in CRC prevention remains poorly understood. SCFAs, notably butyrate and propionate, are thought to influence CRC by regulating gene expression, expressing immunomodulatory effects, promoting immune cell differentiation, and mitigating inflammation. Moreover, compelling evidence underscores the role of SCFAs, including butyrate and propionate, in directly influencing intestinal epithelial cell transformation and inhibiting CRC by regulating tumor suppressor gene expression, promoting apoptosis, and modulating CRC cell proliferation and metabolism [
165,
166,
167]. Butyrate is an energy metabolite and supports normal colon cell proliferation. In CRC cells, butyrate reprograms cell metabolism by promoting pyruvate kinase isozyme 2 (PKM2) activity, inhibiting the Warburg effect and enhancing energy metabolism, therefore inhibiting cancerous colonocytes, which rely on glucose due to the Warburg effect [
133,
168]. SCFAs act as an HDAC inhibitor, fostering apoptosis in cancer cells [
166,
169,
170,
171,
172,
173]. Additionally, SCFAs play a pivotal anti-inflammatory role in regulating local and systemic immune cells, contributing to their antitumor efficacy [
174]. SCFAs mitigate inflammation by inhibiting NF-κB activation, decreasing pro-inflammatory cytokine expression like tumor necrosis factor-alpha (TNF-α), promoting anti-inflammatory cytokines such as IL-10, and transforming growth factor-beta, and facilitating the differentiation of naïve T cells into Tregs, thereby dampening immune responses [
175]. They promote antimicrobial compound production, neutrophil and macrophage inhibition, Treg activation, and dendritic cell induction of tolerogenic properties [
174]. In a recent
in vitro experiment by Mowat et al. CRC cells treated with SCFAs induced much greater activation of CD8+ T cells than untreated CRC cells [
175]. Surprisingly, the butyrate-producing bacterium
Fusobacterium nucleatum does not consistently inhibit colon cancer; instead, it may promote cancer progression via mechanisms such as TLR4/myeloid differentiation primary response 88 (MYD88)/NF-κB signaling [
176]. Furthermore, despite the anticipated decrease in DNA damage within cancer cells, numerous reports suggest that SCFAs might exacerbate DNA damage accumulation in CRC cells by disrupting DNA repair mechanisms [
173,
177,
178,
179,
180]. Hence, the antitumorigenic effects of SCFAs likely involve intricate mechanisms extending beyond the tumor cells themselves, particularly significant in CRC cells with underlying DNA repair defects, such as microsatellite instability-high (MSI) CRC subset known for its heightened immunogenicity. Given inflammation's potent role in tumor progression, these effects likely contribute to SCFAs' antitumor efficacy. However, as tumor-targeted T-cell responses are crucial for antitumor immunity and treatment efficacy, SCFAs like butyrate may suppress such responses, potentially fueling tumor progression and compromising treatment outcomes [
181,
182,
183,
184].
Tian et al. investigated the potential protective role of SCFAs in the development of colitis-associated CRC using a mouse model induced by azoxymethane (AOM) and dextran sodium sulfate (DSS) [
185]. The researchers administered a mix of SCFAs in the drinking water throughout the study. They found that the SCFA mix significantly reduced tumor incidence and size in the mice with colitis-associated colorectal cancer. Additionally, the SCFA mix improved colon inflammation and disease activity index score and suppressed the expression of proinflammatory cytokines such as IL-6, TNF-α, and IL-17. These findings suggest that SCFA mix administration could prevent tumor development and attenuate colonic inflammation, indicating its potential as an agent for the prevention and treatment of colitis-associated colorectal cancer. Further investigation is warranted to determine if supplementing with dietary butyrate or consuming foods rich in butyrate-producing bacteria, such as omega-3 polyunsaturated fatty acids, can effectively hinder colorectal cancer and lower its occurrence.
4.1.3. Disorders of the Gut-Brain Axis
The gut-brain axis facilitates bidirectional communication between the gastrointestinal and nervous systems through a complex signaling pathway network [
186,
187,
188]. This intricate system encompasses connections such as the enteric nervous system, vagus nerve, immune system, endocrine signals, microbiota, and metabolites. Disruption of communication along the gut-brain axis is increasingly recognized as a significant contributor to neuroinflammation, which is considered a common feature of several neurodegenerative diseases, including Alzheimer's and Parkinson's diseases, characterized by chronic and debilitating conditions marked by the progressive degeneration of neurons [
189,
190,
191,
192,
193,
194,
195]. Recent research suggests that neurodegenerative diseases may originate in the intestinal epithelium before affecting the brain via the gut-brain axis [
196,
197,
198,
199,
200,
201]. Various studies have documented the accumulation of protein aggregates, characteristic pathologies of neurodegenerative diseases such as Alzheimer's and Parkinson's, in enteric neurons or the gastrointestinal epithelium years before their detection in the central nervous system [
194,
202,
203,
204,
205]. Functional studies illuminate major microbiota components' roles in the gut-brain axis [
206,
207,
208,
209]. An important aspect is the observed close correlation between alteration in the microbiota, mucosal immunity, and intestinal vascular impairment, potentially leading to the gradual release of systemic inflammatory mediators and bacterial components such as LPS, thereby initiating or exacerbating the development of neurological disorders [
210,
211,
212]. Evidence suggests that microbial and systemic inflammatory molecules could contribute to cerebral vascular impairment, microglial activation, neuronal dysfunction, and pre- and post-synaptic activity imbalances. The microbiome of patients with Parkinson’s and Alzheimer's disease exhibits a reduction in SCFA-producing bacteria [
210,
213]. Recent research has highlighted their importance for learning and memory, with cuts in SCFAs associated with inflammation in Multiple Sclerosis patients and compromised neuronal function in various neurodegenerative diseases [
214,
215]. Furthermore, SCFAs appear to have neuroprotective roles, affecting the brain indirectly or directly by acting as ligands for GPCRs or as epigenetic modulators of HDAC to control transcriptional changes that affect neuronal functions [
216,
217,
218,
219,
220]. The diminished concentration of SCFAs is suggested to be a critical factor in disrupting gut-brain balance, but the role of SCFA in this context is under active investigation. These SCFAs can cross the blood-brain barrier, likely through the monocarboxylate transport system, influence brain function, and regulate blood flow, with dietary butyrate demonstrating an anti-inflammatory effect in the brain by influencing blood–brain barrier permeability [
221,
222]. SCFAs have also been implicated in maintaining gut and immune homeostasis in mammalian systems, highlighting their neuro-immunoendocrine regulatory role in the brain [
221,
223]. In Parkinson's disease, the decline in butyrate levels is thought to lead to intestinal barrier integrity impairment, release of LPS and other pro-inflammatory molecules into the bloodstream, and trigger microglial activation [
135,
224]. Furthermore, reduced SCFAs and microbiota alterations result in decreased circulating GLP-1 levels. The lowered SCFA-mediated secretion of GLP-1 may activate pro-inflammatory pathways and depressive symptoms in PD patients [
225,
226]. Additionally, butyrate can induce epigenetic modifications in the genome of neurodegenerative disorder patients. Methylation analysis on blood samples from Parkinson’s disease patients and controls revealed a correlation between alterations in butyrate-producing bacterial taxa and epigenetic changes in genes containing butyrate-associated methylation sites. Notably, these modified sites coincide with genes implicated in psychiatric and gastrointestinal disorders [
227].
In a study by Kong et al., 16S ribosomal RNA gene sequencing and gas chromatography-mass spectrometry analyses in a Drosophila model of Alzheimer's disease revealed a decrease in
Lactobacillus and
Acetobacter species correlating with a dramatic reduction in acetate [
228]. Similarly, in Drosophila models of Parkinson's disease, administration of sodium-butyrate reduced degeneration of dopaminergic neurons and improved locomotor defects in a pan-neuronal transgenic fly model expressing mutant-human-α-Synuclein [
222]. The SCFA composition derived from microbes also clinically correlates with neural activity and brain structure, as evidenced by functional and structural magnetic resonance imaging [
229]. Recently, Muller et al. examined the fecal SCFA profile of patients with a major depressive disorder/generalized anxiety disorder, comparing it with nuclear magnetic resonance spectroscopy and self-reported depressive and gut symptoms. The severity of depressive symptoms positively correlated with acetate levels and negatively correlated with butyrate levels [
230]. In preclinical studies focusing on Alzheimer’s disease, prebiotic and probiotic supplementation appear advantageous, although limited data is available specifically on SCFA. Bonfili et al. demonstrated the positive impacts of SLAB51 treatment on 8-week-old transgenic Alzheimer’s disease model mice over four months [
231,
232,
233]. SLAB51 administration enhanced performance in the novel object recognition test, reduced brain damage, decreased Aβ plaques, elevated SCFAs, and lowered plasma cytokine levels [
233]. Additionally, prebiotics have shown efficacy in Alzheimer’s disease amyloid models. Liu et al. treated 5XFAD transgenic Alzheimer’s disease model mice with prebiotic mannan oligosaccharide for eight weeks starting from birth. They observed improvements in cognitive deficits, reduction in Aβ plaques, decreased oxidative stress, diminished microglial activation, and alterations in the gut microbiome. Interestingly, gut microbiome-induced changes in the brain appeared to be mediated by SCFAs, as supplementation with SCFAs produced similar effects [
234]. Finally, a case report demonstrated that FMT improved cognitive function, microbiota diversity, and SCFA production in an Alzheimer's patient [
235].
Several studies have investigated the administration of probiotics in both murine models and human subjects with Parkinson’s disease, exploring their impact on gastrointestinal and neurological symptoms [
236,
237,
238,
239,
240,
241,
242]. A pilot study regarding FMT use in Parkinson’s patients has recently been published, with promising data [
243]. However, only a few studies have evaluated SCFA's role. Specifically,
Bifidobacterium has been demonstrated to be effective in modulating the host microbiota in a murine model induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [
244]. In mice overexpressing α-synuclein, a prebiotic diet altered the activation of microglia and motor deficits by changing the composition of the gut microbiome and levels of SCFAs [
245]. Combining polymannuronic acid with
Lacticaseibacillus rhamnosus GG demonstrated more potent neuroprotective effects against Parkinson’s disease than either treatment alone, suggesting the therapeutic promise of synbiotics in Parkinson’s disease [
246]. Oral administration of
B. breve CCFM1067 to MPTP-induced Parkinson’s disease mice led to a reduction in intestinal microbial alterations, marked by a decline in pathogenic bacteria (
Escherichia-Shigella) and an increase in
Bifidobacterium and
Akkermansia. This intervention also restored SCFA production (butyrate and acetate), which may account for the observed local and cerebral anti-inflammatory effects. Recently,
Bifidobacterium animalis subsp. Lactic Probio-M8 (Probio-M8) was examined to evaluate its additional beneficial effects and mechanisms when used as an adjunct treatment alongside conventional therapy (Benserazide and dopamine agonists) in patients with Parkinson’s. This investigation was conducted over three months in a randomized, double-blind, placebo-controlled trial [
247]. Clinical outcomes were assessed by analyzing changes in various clinical indices, gut microbiome composition, and serum metabolome profiles before, during, and after the intervention. The findings revealed that co-administration of Probio-M8 resulted in additional benefits, including improved sleep quality, reduced anxiety, and alleviated gastrointestinal symptoms. Metagenomic analysis demonstrated significant modifications in the participants' gut microbiome and serum metabolites following the intervention. The serum concentration of acetic acid was notably higher in the probiotic group.
IBS is a Disorder of Gut-Brain Interaction (DGBI) characterized by abdominal pain and changes in stool consistency or frequency. According to the Rome IV criteria, IBS can be divided into four subtypes based on the primary clinical features: IBS with diarrhea (IBS-D), IBS with constipation (IBS-C), IBS with mixed stool patterns (IBS-M), and unclassified IBS [
248,
249,
250]. SCFAs play a pivotal role in IBS, with reported findings indicating that patients with IBS exhibit significantly elevated levels of acetate, propionate, and total SCFAs in fecal samples, with the severity of symptoms correlating positively [
251]. Alterations in SCFA levels are subtype-specific, with reduced levels in IBS-C and increased levels in IBS-D compared to controls [
252,
253]. Treem et al. sought to investigate whether patients with IBS-D exhibit a distinct pattern and pace of carbohydrate and fiber fermentation in SCFA in vitro studies of fecal homogenates compared to controls. The fecal SCFA profile of IBS-D patients revealed diminished concentrations of total SCFA, acetate, and propionate alongside elevated levels and proportion of n-butyrate [
254]. Fredericks et al., in 2021, examined gut microbiota, concentrations of SCFA, and mRNA expression of monocarboxylate transporters in individuals with IBS-C, IBS-D, and healthy controls. They observed changes in fecal SCFA ratios in both IBS groups, with a decrease in all three measured SCFAs in IBS-C and a reduction specifically in acetic acid in IBS-D [
255]. Similarly, Undseth et al. aimed to compare colonic fermentation between individuals with IBS and healthy counterparts by examining serum SCFA concentrations before and 90 minutes after ingesting lactulose, an unabsorbable yet fermentable carbohydrate. They found that reduced serum SCFA levels post-lactulose ingestion may indicate compromised colonic fermentation in IBS patients [
256]. The dysregulated SCFA levels in feces are linked to shifts in intestinal bacterial composition in IBS patients, characterized by higher counts of acetate and propionate-producing bacteria like
Veillonella and
Lactobacillus and lower counts of butyrate-producing bacteria like
Roseburia-Eubacterium rectale group [
251,
257,
258]. Zhou et al. recently set out to investigate how linaclotide affects the gut microbiota and pinpoint essential bacterial genera that could influence linaclotide's effectiveness. Interestingly, they discovered a direct link between higher levels of
Blautia and SCFA concentrations and the amelioration of clinical symptoms in patients with IBS-C [
259].
SCFAs, particularly propionate and butyrate, show promise as non-invasive biomarkers for diagnosing IBS, with diagnostic properties consistent across all IBS subgroups. Farup et al. 2016 examined fecal SCFA as a potential diagnostic indicator for IBS in a study involving 25 IBS subjects and 25 controls. They assessed total SCFA levels and individual SCFA amounts to identify the most effective diagnostic approach. Their findings revealed that the discrepancy between propionic and butyric acid levels demonstrated superior diagnostic performance using a threshold of 0.015 mmol/l to indicate IBS, independent of the IBS subgroup [
260].
Several potential mechanisms exist through which SCFAs could influence the pathophysiology of IBS, many of which have been previously examined in the IBD section of this review. As already described, SCFAs interact with specific receptors, such as GPR41, GPR43, and GPR109A, expressed in various gastrointestinal cell types, modulating physiological responses. They play a multifaceted role in immunity and inflammation, influencing inflammatory mediator production, immune cell differentiation, and intestinal barrier integrity [
261,
262,
263,
264,
265]. Additionally, SCFAs influence the differentiation of immune cells, including T cells and Tregs, and suppress intestinal inflammation [
145,
266,
267]. They also contribute to the integrity of the intestinal barrier by promoting mucin secretion and enhancing tight junction assembly [
268,
269,
270,
271,
272,
273].
Furthermore, SCFAs impact gut motility through various mechanisms, including modulation of neural activity, neurotransmitter release, and regulation of calcium signaling and smooth muscle contractility [
274,
275,
276,
277,
278,
279,
280,
281,
282,
283,
284,
285]. The effects of SCFAs on colonic motility are nuanced and context-dependent, varying based on SCFA concentration and colonic segment [
274,
275,
276,
277,
278,
279,
280,
281,
282,
283,
284,
285]. Waseem et al., in their recent prospective observational study, investigated the associations between fecal SCFAs, colonic transit time, fecal bile acids, and dietary intake in individuals with IBS and healthy controls [
286]. They found that fecal SCFAs were inversely correlated with overall and segmental colonic transit time, with similar patterns observed in both IBS and healthy control groups. Additionally, the acetate-to-butyrate ratio was associated with slower transit times. Logistic regression analyses demonstrated that acetate could accurately predict delayed colonic transit time and BAD. These findings suggest that fecal SCFAs and dietary factors may play a role in the IBS pathophysiology and serve as diagnostic markers for bowel transit disorders [
286].