Next Article in Journal
Prolonged On-Vine vs. Cold of Actinidia eriantha: Differences in Fruit Quality and Aroma Substances during Soft Ripening Stage
Next Article in Special Issue
Simultaneous Improvement of Final Product-Tolerance and Thermostability of GH39 Xylosidase for Prebiotic Production by Directed Evolution
Previous Article in Journal
Evaluating the Quality of Cheese Slices Packaged with Na-Alginate Edible Films Supplemented with Functional Lactic Acid Bacteria Cultures after High-Pressure Processing
Previous Article in Special Issue
Metabolomic Characteristics of Liver and Cecum Contents in High-Fat-Diet-Induced Obese Mice Intervened with Lactobacillus plantarum FRT10
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Health Benefits and Side Effects of Short-Chain Fatty Acids

1
Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
2
School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
3
Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
4
Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, National Agricultural Science & Technology Center, Chengdu 610213, China
*
Authors to whom correspondence should be addressed.
Foods 2022, 11(18), 2863; https://doi.org/10.3390/foods11182863
Submission received: 13 August 2022 / Revised: 8 September 2022 / Accepted: 13 September 2022 / Published: 15 September 2022

Abstract

:
The gut microbiota and their metabolites could play an important role in health and diseases of human beings. Short-chain fatty acids (SCFAs) are mainly produced by gut microbiome fermentation of dietary fiber and could also be produced by bacteria of the skin and vagina. Acetate, propionate, and butyrate are three major SCFAs, and their bioactivities have been widely studied. The SCFAs have many health benefits, such as anti-inflammatory, immunoregulatory, anti-obesity, anti-diabetes, anticancer, cardiovascular protective, hepatoprotective, and neuroprotective activities. This paper summarizes health benefits and side effects of SCFAs with a special attention paid to the mechanisms of action. This paper provides better support for people eating dietary fiber as well as ways for dietary fiber to be developed into functional food to prevent diseases.

1. Introduction

Short-chain fatty acids (SCFAs) are mainly metabolites of dietary fiber and protein in gut [1,2], and they could also be produced by bacteria of the skin and vagina [3,4]. Dietary fibers are main sources of SCFAs, and they can be classified as soluble (such as pectins and inulin) and insoluble dietary fibers (such as various forms of resistant starches) [5]. The SCFAs have less than six carbon atom numbers and mainly include acetate, propionate, butyrate, pentanoate, malonate, and so on [6]. Among them, acetate, propionate, and butyrate are three major SCFAs, which account for 90% of SCFAs produced by gut microbiota [6]. The chemical structures of three SCFAs are shown in Figure 1. The SCFAs are produced primarily in the cecum and proximal colon, and their concentrations decline from proximal to the distal colon as the substrates used for fermentation are exhausted gradually [7]. Several factors affected the production of SCFAs, such as substrate source, composition of gut microbes, colonic pH value, gut transit time, and site of substrate fermentation [8,9]. For example, a study showed that resistant starch 5 (amylose–lipid complexes) produced more butyric acid than resistant starch 2 (nongelatinized native starch granule) and resistant starch 3 (retrograded starch) [10]. Another study indicated that low colonic pH value promoted butyrate production and increased populations of butyrate-producing bacteria [11]. In addition, host physiology, such as intestinal environment, microbe–host interaction, and even social stress, could affect SCFA production [12]. For instance, a study found that stress exposure reduced the levels of colonic SCFAs in mice through modulating the gut microbiota, such as decreasing the SCFAs producing genera Anaerostipes, Butyricicoccus, Coprococcus, and Parabacteroides, as well as increasing the abundance of Odoribacter [13]. Furthermore, more than 90% of SCFAs are absorbed from the intestinal cavity and metabolized by colonocytes or liver [8]. Butyrate is an important energy source for colonocytes, and therefore only a small amount of butyrate reaches the hepatic system, while other absorbed SCFAs that have not been metabolized by colonocytes, particularly acetate and propionate, could reach the liver via the portal vein [7,14]. The liver is a major site for the metabolism of SCFAs in humans, since approximately 40% of acetate and 80% of propionate in portal vein are taken up and metabolize by the liver [15]. Moreover, a small portion of SCFAs in the rectum could bypass the liver and pass directly into the systemic circulation via the internal iliac vein [7,8].
In recent years, many studies have proven that intestinal microbiota and their metabolites play a vital role in human health [16,17]. The gut microbiota-derived SCFAs have shown a variety of biological effects on the host, such as anti-inflammatory, immunoregulatory, anti-obesity, anti-diabetes, anticancer, cardiovascular protective, hepatoprotective, and neuroprotective effects [6,18,19]. The role of SCFAs in human health and diseases has become a research hotspot. This narrative review collects the literature from the Web of Science Core Collection and PubMed databases in the past five years based on keywords in the title and abstract, including short-chain fatty acids, SCFAs, acetic acid, acetate, propionic acid, propionate, butyric acid, butyrate, isobutyric acid, isobutyrate, valeric acid, valerate, hexanoic acid, and hexanoate, and summarizes the bioactivities of SCFAs with special attention paid to their mechanisms of action.

2. The Bioactivities of SCFAs

The health benefits of SCFAs have been widely studied, and the mechanisms of action have been explored (Table 1, Table 2 and Table 3 and Figure 2), which are summarized and discussed in detail below. In each section, the results of epidemiological studies are first described, then the results of preclinical studies are discussed, and the results of clinical studies (if any) are finally elaborated.

2.1. Anti-Inflammatory Activity

Inflammation is related to the occurrence and development of many diseases. In recent years, many studies demonstrated that SCFAs could reduce the production of inflammatory factors through several signaling pathways. The SCFAs attenuated the inflammatory response by decreasing the production of pro-inflammatory mediators and enhancing the production of anti-inflammatory mediators. A study indicated that propionate and butyrate alleviated the inflammation in cells by inhibiting the expressions of interleukin (IL)-6, reactive oxygen species (ROS), as well as enhancing the expressions of IL-10 [20]. Besides, it was reported that butyrate attenuated the inflammation induced by lipopolysaccharide (LPS) via up-regulating IL-10 in septic shock [21]. Additionally, the evidence showed that acetate effectively resolved neutrophilic inflammation via inducing caspase-dependent apoptosis of neutrophils, decreasing the activity of nuclear factor-kappa B (NF-κB) and enhancing the production of anti-inflammatory mediators, such as IL-10, transforming growth factor-β (TGF-β), and annexin A1 [22]. In the LPS-treated neonatal mice model, pulmonary inflammation and oxidative stress were reduced by sodium propionate; in the LPS-treated human pulmonary microvascular endothelial cells (HPMECs) model, sodium propionate not only accelerated Nrf2 nuclear translocation, protected cells, and promoted angiogenesis, but also reduced inflammatory response via the NF-κB pathway [23]. Moreover, one in vivo study showed that propionate interfered with the production and migration of inflammatory mediators [24]. Furthermore, some SCFAs had complex bidirectional regulatory properties. It was found that the level of acetate negatively correlated with the pro-inflammatory biomarker interferon-γ (IFN-γ), while the levels of butyrate and valerate positively correlated with IFN-γ and tumor necrosis factor-α (TNF-α) [25].
In summary, SCFAs exhibit good anti-inflammatory activity, and the main mechanisms of action include inhibiting the production of pro-inflammatory mediators, such as IL-6 and TNF-α, as well as enhancing the production of anti-inflammatory mediators, such as IL-10, TGF-β, and annexin A1. In the future, more in vivo studies are needed to prove the bidirectional regulation of SCFAs on inflammatory factors and discover its mechanisms.

2.2. Immunoregulatory Activity

More and more studies have shown that gut microbiota play a vital role in the host’s immune system, and the effects are mainly carried out by producing metabolites, such as SCFAs [26]. The SCFAs could regulate the function of innate immune cells to participate in the immune system, such as dendritic cells. In an ovalbumin-induced allergic mice model, dietary supplement with SCFAs could prevent the exacerbation of lung inflammation induced by vancomycin, via attenuating dendritic cells migration and activation [27]. The SCFAs could also regulate the differentiation and function of T and B cells, and then mediate antigen-specific adaptive immunity. For example, SCFAs induced the production of IL-22 by CD4+ T cells through binding G-protein receptor 41 (GPR41) and inhibiting histone deacetylase (HDAC) [28]. Besides, butyrate promoted the production of IL-22 via increasing hypoxia-inducible factor (HIF) 1α binding to the Il22 promoter through histone modification [28]. For B cells, SCFAs promoted B cell differentiation by increasing acetyl-coenzyme A (acetyl-CoA), oxidative phosphorylation, glycolysis, and fatty acid synthesis [26]. Moreover, as efficient HDAC inhibitors, SCFAs could stimulate B cell differentiation via boosting the expression of B cell differentiation-related genes, such as Xbp-1, Aicda, and Prdm1 [26]. The SCFAs could also decrease circulating immunoglobulin (Ig) E level [27]. Furthermore, SCFAs could promote murine and human B10 cell generation via inhibiting HDAC [29].
In short, SCFAs participate in the function of immune system via reducing the migration and activation of dendritic cells to relieve allergy, as well as promoting T and B cells’ differentiation to regulate antigen-specific adaptive immunity. The immunoregulatory action of SCFAs are mainly achieved by directly binding SCFA-specific G-protein-coupled receptor (such as GPR41) on the cell surface and entering cells to regulate cell metabolism and inhibit HDAC.

2.3. Anti-Obesity Activity

Obesity is a metabolic disorder and mainly caused by an imbalance between energy intake and expenditure [19,30]. Recently, many studies indicated that SCFAs played a vital role in the management of obesity. For example, one epidemiological study suggested that human milk SCFAs exerted a protective effect against excess weight gain in infants [31].
SCFAs play an important part in obesity and energy metabolism by regulating the appetite. For example, a study showed that the mechanism of SCFAs suppressing food intake was related to vagal afferent, and the efficacy was butyrate > propionate > acetate. Moreover, butyrate exerted an anorexigenic effect through activating vagal afferent neurons and their projection sites, such as nucleus tractus solitaries (NTS) neurons, and directly increasing Ca2+ concentration in nodose ganglion neurons (NGNs) [32]. Besides, serum glucagon-like peptide 1 (GLP-1), peptide YY (PYY), and leptin participated in the short-term signal of satiety transferring to the appetite center of the brain. A 28-day experiment of pigs showed that acetate and propionate increased the concentrations of serum GLP-1, PYY, and leptin, and then reduced the appetite [33]. In addition, SCFAs reduced appetite and fat accumulation via modulating relevant genes and hormones, such as mitochondrial transcription factor A, tumor necrosis factor receptor superfamily member 9, cytochrome-C oxidase IV, and free fatty acid receptor 2 [34].
The SCFAs could also exert an anti-obesity effect via other different pathways. It was reported that SCFAs could restrain lipogenesis [35]. The SCFAs mediated the suppression of de novo lipogenesis in male rats by suppressing hepatic acetyl-CoA carboxylase-1(Acc1) expression [35]. Furthermore, propionate repressed the triglyceride (TG) accumulation via modulating the expression levels of PPARα-responsive genes, such as carnitine palmitoyl transferase II (CPTII) and trifunctional protein alpha (TFPα) [36]. Another study suggested that SCFAs could protect against high-fat diet-induced obesity and suppress lipid synthesis [37]. Besides, propionate reduced obesity-associated metabolic disturbances via decreasing the hepatic TG and increasing odd-chain fatty acids (OCFAs) formation [38]. Additionally, acetate could decrease lipid accumulation, inhibit white adipose tissue lipolysis and induce browning of white adipose tissue, which could reduce the body adiposity by increasing thermogenesis [39]. Furthermore, a randomized, placebo-controlled, single-blind crossover study showed that inulin-propionate ester (10 g/day) significantly decreased the striatal blood oxygen level-dependent signal, high energy food picture appeal, and energy intake at an ad libitum meal [40].
In other words, SCFAs have an obvious anti-obesity effect, and the mechanisms involve suppressing appetite, restraining the lipogenesis, and inducing browning of white adipose tissue (Figure 3). Furthermore, the effect of SCFAs on lipid accumulation needs to be confirmed by more studies.

2.4. Cardio-Protective Activity

Cardiovascular disease (CVD) is a chronic non-communicable disease with high morbidity and mortality on a global scale [41]. SCFAs have good protective effects on cardiovascular system [42], and the related mechanisms of action are discussed below.
SCFAs could protect cardiovascular system by decreasing atherosclerosis. For example, a study showed that butyrate inhibited the progression of diet-induced atherosclerosis by decreasing intestinal cholesterol absorption via regulating related transporters expression, such as Niemann-Pick C1-like 1 (Npc1l1, a major intestinal cholesterol transporter) and ATP-binding cassette (ABC) transporters G5 and G8 [43]. Moreover, propionate reduced intestinal cholesterol absorption and aortic atherosclerotic lesion area via increasing levels of regulatory T (Treg) cell and IL-10 and reduced the expression of Npc1l1 [44]. Moreover, the elevation of plasma total cholesterol (TC) is an important risk factor for atherosclerosis. The SCFAs significantly reduced plasma TC via enhancing fecal excretion of bile acids and up-regulating the gene expressions of sterol-regulatory element-binding protein 2 (SREBP2), low-density lipoprotein (LDL) receptor, and cholesterol 7 alpha-hydroxylase (CYP7A1) in the liver [45]. Furthermore, a randomized, double-blind clinical trial suggested that propionate intake (500 mg, twice daily) significantly reduced levels of LDL and non-high-density lipoprotein cholesterol, which were effective targets for atherosclerotic CVD prevention [44].
SCFAs could exert a cardiovascular protective effect by inhibiting hypertension. For instance, one study suggested that acetate, butyrate, and propionate could reduce blood pressure, and acetate showed the most powerful antihypertensive effect among these three SCFAs [46]. Moreover, acetate kept the balance of vasoconstriction and vasodilation shifts through up-regulation of SCFAs receptors, Olfr78, GPR41, and GPR43, in which way to prevent high-fructose diet-induced hypertension [47]. Besides, an in vivo study showed that propionate alleviated cardiac hypertrophy, fibrosis, vascular dysfunction, and hypertension in both wild-type NMRI and ApoE−/− mice models, and the mechanisms mainly depended on Treg cells [48].
The evidence indicated that SCFAs had protective effects against ischemia/reperfusion-related injuries (IRI). Butyrate significantly improved myocardial IRI via the gut–brain neural circuit and might be mediated by the paraventricular nucleus (PVN)-superior cervical ganglion (SCG) sympathetic pathway [49]. Besides, acetate, butyrate, and propionate repaired the myocardial infarction (MI) impairment by reducing the infiltration of CX3CR1+ monocytes to the peri-infarct zone after MI and played an important part in maintaining host immune composition [50].
In summary, SCFAs could be a potential therapeutic strategy to prevent and manage CVD through reducing blood lipid and blood pressure, alleviating IRI, and repairing MI injury.

2.5. Hepatoprotective Activity

Liver diseases are generally divided into non-viral liver diseases and viral liver diseases [51]. Recent studies have found that gut microbiota and its metabolites, such as SCFAs, could prevent and manage several liver diseases, particularly non-viral liver diseases [52,53]. An epidemiology study found that fecal SCFAs were negatively correlated with cirrhosis disease severity [54].
Non-viral liver diseases mainly involve alcoholic liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), and drug- or pollutant-induced liver injury. ALD could be caused by a long-term heavy alcohol intake, which involved alcoholic hepatitis, fibrosis, and cirrhosis [55]. A study found that propionate alleviated the ethanol-induced hepatic steatosis and enhanced hepatic function through maintaining the intestinal epithelial barrier function and inhibiting hepatic toll-like receptor 4 (TLR4)-NF-κB pathway [56]. The occurrence of NAFLD was closely related to intestinal flora disturbance, and gut microbiota-derived SCFAs could be a valuable strategy for preventing and treating NAFLD [57]. The SCFAs could regulate mucus secretion, microbial homeostasis, and intestinal epithelial tight junction to reduce the spread of intestinal endotoxin to the liver, thereby reducing the oxidative pressure and the level of inflammation in the liver and then delaying the development of NAFLD [58]. In addition, SCFAs changed the intestinal micro-ecology to protect the gut barrier, which could slow down the development of NAFLD-related diseases [58]. Another study suggested that SCFAs could inhibit hepatic steatosis by activating AMPK and PPAR signaling pathways and down-regulating the expression of genes related to lipid synthesis, such as sterol-regulatory element-binding protein 1 (SREBP-1), FAS, stearoyl-CoA desaturase 1 (SCD1), ACC1, and liver X receptor (LXR) [59]. Furthermore, pectin, Jaboticaba berry peel, and fu instant tea could alleviate fatty liver disease by regulating intestinal SCFAs [60,61,62]. As for drug- or pollutant-induced liver injury, SCFAs also exerted hepatoprotective effects. Cytochrome p450 (CYP) maturation in the liver is important for metabolic activity and xenobiotic detoxification. An in vitro study showed that the mixture of acetate, propionate, and butyrate increased the expression of CYP3A4 and ALB in human-induced pluripotent stem cell-derived liver organoids, which improved the hepatic maturation and enhanced metabolic activity and xenobiotic detoxification [63]. Furthermore, it was reported that acetate reduced serum levels of aspartate aminotransferase and alkaline phosphatase, which indicated that it improved hepatic function. Meanwhile, acetate increased mitochondrial efficiency and adenosine triphosphate production [39].
In brief, SCFAs play a positive role in non-viral liver diseases. The mechanisms of action involved maintaining the intestinal epithelial barrier, regulating the lipid metabolism and inflammatory response in liver, increasing mitochondrial efficiency, and promoting CYP maturation.

2.6. Anti-Diabetic Activity

Diabetes mellitus is characterized by hyperglycemia caused by decreasing insulin secretion or insulin resistance, and 592 million people will have diabetes mellitus by the year 2035 worldwide according to the International Diabetes Mellitus Federation prediction [64,65]. The effects of SCFAs on diabetes mellitus have been widely studied. A microbiome-wide association study on large population cohorts showed that butyrate and acetate had a causal relationship with type 2 diabetes using bidirectional Mendelian randomization analyses [66].
Propionate attenuated high-fat diet-induced insulin resistance and improved insulin sensitivity, and the mechanism of action might relate to stimulating OCFA production [38]. Besides, acetate and propionate improved insulin sensitivity and glucose tolerance [67]. In addition, the combination of acetate and propionate effectively improved insulin sensitivity in high-fat diet-fed mice via reducing inflammation through decrease of T helper 1 (Th1) and Th2 cells and increase of Treg cells in the spleen and mesenteric lymph node [67]. Moreover, butyrate could promote the growth of intestinal epithelial cells, strengthen the intestinal tight connection, and regulate the activities of gut microbiota and immune cells, in which way to maintain the gut barrier and ultimately prevent and treat type 1 diabetes mellitus [58]. Several studies also showed that oral and dietary supplementation of butyrate, as well as human acetate colonic infusions and vinegar administrations, could prevent high-fat diet-induced insulin resistance and improve glucose homeostasis [68,69]. Additionally, propionate could activate AMP-activated protein kinase (AMPK) by binding GPR43 and increasing intracellular Ca2+. Besides, propionate suppressed hepatic gluconeogenesis via down-regulating gluconeogenic enzymes, such as glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), through activation of AMPK [70]. Furthermore, acylated starch had a greater effect on the improvement of type 2 diabetes indexes compared to native resistant starch, such as fasting blood glucose, serum insulin level, and amino acid metabolism [71]. In addition, a randomized clinical trial based on 29 overweight/obese individuals suggested that after the Mediterranean diet intervention for 8 weeks, postprandial plasma butyric acid incremental area under the curve (IAUC) was significantly increased, which was negatively correlated with plasma insulin IAUC and oral glucose insulin sensitivity [72].
Overall, SCFAs could prevent and manage diabetes mellitus via increasing insulin sensitivity, improving glucose homeostasis, and suppressing hepatic gluconeogenesis. Furthermore, more high-quality clinical studies are also needed to determine the effect of SCFAs on glycemic control and diabetic mellitus.

2.7. Effects on Inflammatory Bowel Diseases

The inflammatory bowel diseases (IBDs) are complex immune-mediated diseases characterized by chronic inflammation in the gastrointestinal tract, and caused by the interaction among genetic, immunologic, microbial, and environmental factors [13]. SCFAs are believed to play a beneficial role in human gut health and prevent IBDs via different pathways [73], and the mechanisms of action are shown in Figure 4. The gut epithelium consists of a layer of constantly renewing epithelial cells and becomes the first line of defense against enteric infection [74]. A study showed that propionate promoted intestinal epithelial cell migration and increased cell speed and persistence in a HDAC inhibition, GPR43, and the signal transducer and activator of transcription 3 (STAT3) in a dependent manner [74]. Another study showed that butyrate promoted intestinal integrity and homeostasis via affecting metabolism, intestinal barrier function, and transporter expression [75]. Heat shock proteins (HSP) play a crucial role in maintaining and protecting the integrity of intestinal epithelial cells. Propionate contributed to intestinal homeostasis via increasing the level of Hspa1a (a major transcript of HSP70), up-regulating HSP70, and phosphorylating heat shock factor 1 [76]. Moreover, the transcriptional activation of HSP70 was proven to be related to the activation of mitogen-activated protein kinase (MEK)/ extracellular signal-regulated kinase (ERK) and mTOR pathways, as well as the inhibition of HDAC [76]. Besides, acetate increased the expression of MUC2 and CDX2, as well as the production of mucin proteins, in mucus-secreting colon epithelial cells (HT29-MTX), which indicated that it could improve the intestinal epithelium protective function [77]. SCFAs in an optimal dose could modulate the structure of gut microbiota, regulate the activities of immune cells and intestinal epithelial cells via regulating the gene expression of intestinal cells in an HDAC-dependent way, and subsequently improve the gut barrier function [58]. It was reported that acetate, propionate, and butyrate not only increased transepithelial electrical resistance (TER) and improved the formation of tight junction, but also inhibited the activation of NLRP3 inflammasome and autophagy induced by LPS to protect intestinal barrier function [78]. The SCFAs contributed to intestinal homeostasis via regulating the immune system and reducing inflammation. For example, butyric acid elicited α-defensin secretion by Paneth cells and improved enteric innate immunity through potent microbicidal activities, which contributed to intestinal homeostasis [79]. Besides, propionate repressed IL-17- and IL-22-producing γδ T cells to relieve intestinal inflammation [80]. Moreover, SCFAs transported by monocarboxylate transporter (MCT)-1 could suppress inflammatory responses in Caco-2 cells induced by TNF-α via decreasing IL-8 and IL-6 expression levels and inhibiting the activation of NF-κB, ERK, p38 mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK), and spleen tyrosine kinase (Syk) [81]. Additionally, SCFAs prevented the development of intestinal inflammation via inhibiting dual-specificity phosphatase 6 (DUSP6) through the up-regulation of microRNA-145 (miR-145) by decreasing the CCAAT enhancer-binding protein beta (CEBPB) expression, and they also improved the disease activity index of LPS-treated mice [82].
In short, SCFAs, especially butyrate, generally showed protective effects on IBDs. The mechanisms of action involved enhancing intestinal barrier function, regulating immune system, and reducing inflammation.

2.8. Effects on Constipation

Constipation is a multifactorial intestinal disorder with high incidence and difficult treatment, leading to a serious decline in quality of life [83]. Studies have identified SCFAs as important regulatory factors in constipation. For example, an epidemiological study found that acetate, propionate, and butyrate were negatively correlated with the severity of constipation [84]. Another study with analysis of human feces found that the concentrations of SCFAs, particularly butyrate, in the feces of constipated patients, were lower than those of healthy people [85]. Furthermore, studies showed that acetic acid and butyric acid, but not propionic acid, could relieve constipation [86,87]. A study indicated that acetic acid increased water content of feces and small intestinal transit rate, and butyric acid decreased the transit time through the gut [86]. Besides, butyrate attenuated constipation symptoms by promoting the secretion of colonic hormones and maintaining intestinal barrier integrity [85]. Moreover, butyrate promoted defecation, improved intestinal mobility, and promoted Cajal cells proliferation via activating the AKT-NF-κB signaling pathway [88].
In brief, SCFAs, especially butyrate, play a key role in regulating colonic motility and might be a novel therapy of constipation.

2.9. Neuroprotective Activity

The gut microbiota and brain communicate with each other in a variety of ways and involve microbial metabolites, such as SCFAs [89]. Recent studies have shown that SCFAs are important mediators of the microbiota–gut–brain axis and play a crucial role in regulating the physiology and behavior of the central nervous system [90,91]. A study based on 116 Polish women suggested that depressive women had significantly lower levels of acetate and propionate compared with healthy individuals [92]. A study showed that acetate improved cognitive impairment, decreased the cluster of differentiation 11b (CD11b, a tight junction protein) level, and suppressed neuroinflammation in the Alzheimer’s disease model mice. For BV2 cells, acetate exerted anti-neuroinflammatory effects via inhibiting the phosphorylation of NF-κB, p65, ERK, and JNK; decreasing cyclooxygenase-2 (COX-2) and IL-1β levels; and increasing the GPR41 level [93].
In summary, some SCFAs, such as acetate, may help to prevent and manage neurodegenerative and neuropsychiatric diseases.

2.10. Anticancer Activity

Cancer is a serious public health problem worldwide, and cancer cells exhibit specific characteristics compared with normal cells, such as infinite proliferation, weak adhesion, and strong agglutination [94,95]. SCFAs could exert anticancer activities through different pathways. For example, SCFAs inhibited proliferation of human cervical cancer HeLa cells via down-regulating free fatty acid receptor 2 (FFAR2) expression [96]. Another study showed that acetate and propionate inhibited the carcinogenesis process of colorectal cancer via inhibiting cancer cell proliferation and inducing cell cycle arrest [97]. The SCFAs decreased the incidence and size of colitis-associated colorectal tumor in azoxymethane/dextran sodium sulfate-treated mice by improving colon inflammation and decreasing cell proliferation [98]. Besides, propionate and butyrate reduced the lung metastasis of melanoma cells by increasing the expression of chemokine (C-C motif) ligand 20 (CCL20) in lung endothelial cells and reducing the recruitment of Th17 cells [99]. It was reported that valeric acid suppressed colony formation, migration, and invasion of liver cancer cells in vitro, and suppressed hepatocellular carcinoma development in vivo, as well as improving the survival rate of mice with liver cancer [100]. Additionally, pentanoate and butyrate enhanced the effect of cancer immunotherapy by enhancing cytotoxic T lymphocytes and chimeric antigen receptor T cells via epigenetic and metabolic reprogramming [101]. Furthermore, the anticancer mechanisms of SCFAs also involved inducing apoptosis, activating autophagy, and modulating hematopoiesis [18,102,103].
In short, SCFAs could be a potential agent against several cancers, such as cervical, colorectal, melanoma, and liver cancers. The mechanisms of action mainly included inhibiting cancer cell proliferation, arresting cell cycle, decreasing inflammation, reducing metastasis, and enhancing effect of immunotherapy.

2.11. Anti-Bacterial Activity

SCFAs have been shown to inhibit bacterial growth and viability. A study showed that butyrate could enhance the antimicrobial activity of macrophages without tissue damaging inflammation [104]. Moreover, bacterial infection is one of the main causes of diarrhea. Enterotoxin-producing Bacillus cereus, Clostridium difficile, Clostridium perfringens, Escherichia coli, Staphylococcus aureus, and Vibrio cholerae are the major pathogenic bacteria that secrete highly toxic proteins, which could induce diarrhea [105]. The SCFAs could inhibit these bacteria, decrease enterotoxin cytotoxicity via mimicking the structure of toxin receptors and inhibiting toxin adherence to host cells, and promote the production of beneficial microbiome [105]. In addition, SCFAs could inhibit other foodborne pathogenic bacteria, such as Campylobacter, Salmonella, and Shigella in a pH-, dose-, and complexity-dependent manner [106]. Furthermore, SCFAs could be used to prevent food spoilage due to its antimicrobial activity. The evidence showed that repeated treatments with acetic acid vapors could preserve table grapes’ fruit quality, which was safer compared with SO2 [107].
Overall, SCFAs could prevent and treat bacterial diarrhea via inhibiting the pathogenicity of enteropathogens and decreasing enterotoxin toxicity. Moreover, SCFAs could also play a vital role in prevention of food spoilage.

2.12. Effects of SCFAs on Other Diseases

SCFAs also have other health benefits besides the bioactivities mentioned above. For example, a study found that propionate could alleviate mitochondrial dysfunction, oxidative stress, and apoptosis induced by free fatty acids via up-regulating peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) [108]. Besides, SCFAs could contribute to infant sleep. A study indicated that the higher the proportion of propionate in total fecal SCFAs, the longer the infant at 7 and 8 months of age slept uninterrupted [109]. Moreover, SCFAs could mediate the effects of intestinal microbiota on the metabolism and function of skeletal muscle. It was reported that SCFAs could regulate lipid, carbohydrate, and protein metabolism in skeletal muscle tissues, and they could also increase skeletal muscle mass retention. The mechanisms of action might be related to the activation of AMPK, PPAR-δ, and PGC-1α, as well as the inhibition of HDACs [110]. Moreover, acetate and propionate might contribute to maintaining a positive balance of bone turnover by up-regulating differentiation in primary osteoblasts [111]. Another study suggested that propionate and butyrate increased systemic bone mass under steady-state conditions via inducing the reprogramming of osteoclasts metabolism, enhancing glycolysis, expensing oxidative phosphorylation, and down-regulating several osteoclast genes, such as TRAF6 and NFATc1 [112]. Besides, SCFAs could also prevent bone loss after menopause [112]. Additionally, acetate, propionate, and butyrate synergistically alleviated rheumatoid arthritis by regulating B cells differentiation via FFAR2 receptors [113]. Furthermore, high concentrations of acetate and butyrate could suppress periodontal disease by decreasing the accumulation of neutrophil granulocytes in the gingival pockets via binding FFAR2 and triggering neutrophil Ca2+ signaling and chemotaxis [114].
In brief, SCFAs also exerted beneficial effects on sleep, skeletal muscle, bone loss, arthritis, and periodontal disease.
Table 1. Health benefits of SCFAs from epidemiological studies.
Table 1. Health benefits of SCFAs from epidemiological studies.
Study TypeIndividualsOutcomesRef.
Anti-obesity
Prospective study1585 singleton late preterm or full-term bornHuman milk SCFAs could prevent excess weight gain in infants[31]
Hepatoprotective activity
Prospective study49 patients with cirrhosisSCFAs were negatively correlated with cirrhosis disease severity[54]
Effects on constipation
Cohort study30 patients with ascending colon cancer and 90 patients with mixed refractory constipationSCFAs were negatively correlated with the severity of constipation[84]
Neuroprotective activity
Cross-sectional study116 womenDepressive women had lower levels of acetate and propionate[92]
Abbreviation: SCFAs, short-chain fatty acids.
Table 2. Health benefits and molecular mechanisms of SCFAs from preclinical studies.
Table 2. Health benefits and molecular mechanisms of SCFAs from preclinical studies.
SCFAs SpeciesStudy TypeSubjectsDosesEffects and MechanismsRef.
Anti-inflammation
AcetateIn vivoC57BL/6 mice and C57BL6 GFP Het150 mM in drinking waterInduce caspase-dependent apoptosis of neutrophils;
Decrease the activity of NF-κB;
Enhance production of IL-10, TGF-β, and annexin A1.
[22]
PropionateIn vitroHPMECs0.6 mMAccelerate Nrf2 nuclear translocation;
Protect cells and promote angiogenesis;
Reduce inflammatory response via NF-κB pathway.
[23]
PropionateIn vivoC57BL/6J and Nrf2−/− mice1.2 mg/g i.p.Reduce pulmonary inflammation and oxidative stress.[23]
PropionateIn vivoBALB/c and C57BL/6 mice150 mM in drinking waterInterfere with the production and migration of inflammatory mediators.[24]
ButyrateIn vivoICR mice200 mg/kg i.p.Up-regulate the IL-10 in septic shock.[21]
ButyrateIn vitroRAW 264.7 cells100 μMDown-regulate the IL-6 and IL-1β;
Increase the IL-10.
[21]
Propionate; ButyrateIn vitroTHP-1 cells10 µMInhibit the expressions of IL-4, IL-6, and ROS;
Enhance the expressions of IL-10 and IFN-γ.
[20]
Immunoregulation
ButyrateIn vivoC57BL/6J mice200 mM in drinking waterPromote IL-22 production by CD4+ T cells and ILCs.[28]
Acetate; Propionate; ButyrateIn vitroCD4+ T cells10 mM acetate; 0.5 mM propionate; 0.5 mM butyrate Promote CD4+ T cell and ILC production of IL-22 through GPR41 and HDAC inhibition.[28]
Acetate; Propionate; ButyrateIn vitroSynovial fibroblasts250 µM propionate or the mixture (300 µM acetate, 100 µM propionate, 100 µM butyrate)Interfere with arthritogenic properties of synovial fibroblasts;
Induce cellular senescence.
[24]
Acetate; Propionate; ButyrateIn vivoC57BL/6J and C.129-IL4tm1Lky/J (4get) mice40 mM butyrate, 67.5 mM acetate and 25.9 mM propionate in drinking water Regulate T cells and DC activities;
Reduce the production of IL4-producing CD4+ T cells;
Decrease circulating IgE level.
[27]
Acetate; Propionate; Butyrate; PentanoateIn vitroSplenic B cells isolated from C57BL/6J mice0.5 mM of NaAc, NaPr, NaBu or NaPePromote B10 cell generation;
Enhance B10 cell function.
[29]
Acetate; Propionate; Butyrate; PentanoateIn vivoC57BL/6J mice150 mM acetate, propionate, butyrate or pentanoate in drinking waterPromote B10 cell generation via activation of GPCR.[29]
Anti-obesity
PropionateIn vitroYAMC cells5 mMRepress the TG accumulation via modulating the expression levels of PPARα-responsive genes.[36]
Acetate; Propionate; ButyrateIn vitro3T3-L1 cells6.4 mM acetic acid; 3.2 mM propionic acid or 0.4 mM butyric acid Accelerate the 3T3-L1 adipocyte differentiation;
Promote lipid accumulation via modulation of the expression of LPL, adipocyte FABP4, FATP4, and FAS.
[115]
Acetate; Propionate; ButyrateIn vivoC57BL/6J mice5% acetate, propionate, or butyrate in the dietProtect against high-fat diet-induced obesity;
Suppress hepatic weight and lipid synthesis.
[37]
Acetate; Propionate; ButyrateIn vivoC57BL/6 mice 6 mmol/kg acetate; 6 mmol/kg propionate; 1–6 mmol/kg butyrate, 10 mL/kg i.p.Activate vagal afferent neurons.[32]
Acetate; Propionate; ButyrateIn vivoC57BL/6J mice5% sodium acetate; 5% sodium propionate or 5% sodium butyrate in a high-fat dietReduce appetite and fat accumulation via modulating relevant genes and hormones;
Regulate the expressions of several mRNA.
[34]
Acetate; Propionate; ButyrateIn vivoBarrows (Duroc × Landrace × Yorkshire)0.1% acetate; 0.1% propionate; 0.1% butyrate; 0.1% mixed SCFAs (3:1:1) in diet supplementIncrease the concentrations of the serum GLP-1, PYY and leptin to regulate the appetite;
Down-regulate of the mRNA expressions of FAS, ACC, and SREBP-1C to participate the de novo synthesis of fatty acids;
Enhance the mRNA expressions of LIPE and CPT-1α to participate in fatty acids oxidation.
[33]
Acetate; Propionate; ButyrateIn vivoLong–Evans ratsMNaAc:MNaPr:MNaBu = 60:20:20, dissolve in 0.1 M PBS, 500 mg/kg i.p.Suppress the de novo lipogenesis by reducing mRNA expression of hepatic Acc1.[35]
Cardiovascular protection
AcetateIn vivoSD rats200 mmol/L magnesium acetate in drinking waterUp-regulate SCFAs receptors Olfr78, GPR41, and GPR43 to keep the balance of vasoconstriction and vasodilation shifts.[47]
PropionateIn vivoNMRI and ApoE−/− mice200 mmol/L in drinking waterReduce blood pressure;
Attenuate cardiac hypertrophy, fibrosis, and vascular dysfunction.
[48]
PropionateIn vivoApoE−/− mice200 mg/kg i.g.Reduce intestinal cholesterol absorption and aortic atherosclerotic lesion area;
Increase levels of regulatory T-cell numbers and IL-10;
Reduce the levels of NPC1l1.
[44]
ButyrateIn vitroCaco-2 cells0, 0.1, 1 and 10 mmol/LInhibit cholesterol absorption;
Reduce the levels of NPC1l1;
Increase the levels of ABCG5 and ABCG8.
[43]
ButyrateIn vivoSD rats200 mmol/L in drinking waterImproves myocardial I/R injury via gut-brain neural circuit.[49]
Acetate; Propionate; ButyrateIn vivoC57BL/6J mice100 mmol/L in drinking waterReduce blood pressure.[46]
Acetate; Propionate; Butyrate; valerateIn vivoGolden Syrian hamsters0.5 mol/kg sodium acetate, sodium propionate, sodium butyrate, and valeric acid in high-cholesterol dietEnhance fecal excretion of bile acids;
Up-regulate the gene expressions of SREBP2, LDLR, and CYP7A1 in the liver.
[45]
Hepatoprotection
AcetateIn vivoC57BL/6 mice200µL LITA-Rhd i.p.Decrease lipid accumulation;
Improve hepatic function;
Increase mitochondrial efficiency.
[39]
PropionateIn vitroHepG2 cells0.2, 0.4, 0.8 mMEnhance hepatic function;
Alleviate ethanol-induced hepatic steatosis.
[56]
PropionateIn vivoC57BL/6J mice100 or 200 mM in the dietPrevent ethanol-induced loss of hepatic function;
Alleviate ethanol-induced hepatic steatosis.
[56]
Acetate; Propionate; ButyrateIn vitroBRL 3A cell 10 mM NaAc, 5 mM NaPr, 2.5 Mm NaBu or 10 mM mixture (MNaAc:MNaPr:MNaBu = 3:1:1)Reduce the production of ROS and MDA;
Activate AMPK and PPAR signaling pathways;
Down-regulate the expression of genes related to lipid synthesis.
[59]
Acetate; Propionate; ButyrateIn vitrohuman-iPSC-derived liver organoids1 μM acetate; 1 μM propionate; 1 μM butyrateIncrease the expression of CYP3A4 and ALB.[63]
Anti-diabetes
PropionateIn vitroHepG2 cells0, 0.25, 0.5 mMSuppress gluconeogenesis by down-regulation of gluconeogenic enzymes;
Suppress hepatic gluconeogenesis by activating AMPK;
Activate AMPK via Ca2+/CaMKKβ pathway.
[70]
Acetate; PropionateIn vivoC3H/HeOuJ mice5% SCFA (Ac:Pr, 2.5:1 or Ac:Pr, 1:2.5) in the dietAttenuate high-fat diet-induced insulin resistance.[38]
Acetate; Propionate; ButyrateIn vivoC57BL/6 WT and IL22 KO mice67.5 mM acetate, 40 mM butyrate and 25.9 mM propionate in drinking waterPrevent type 1 diabetes;
Promote development of regulatory T cells.
[116]
Acetate; Propionate; ButyrateIn vivoC57BL/6 miceAcetate (5% w/w of diet), propionate (10% w/w of diet), butyrate (10% w/w of diet), acetate + propionate (5% + 10% w/w of diet)Improve insulin sensitivity.[67]
Prevention and management of inflammatory bowel disease
PropionateIn vivoWT C57BL / 6 mice200 mM in drinking waterPromote intestinal epithelial cell migration;
Increase cell speed and persistence.
[74]
ButyrateIn vivoWT C57BL/6J mice200 mM in drinking waterpromote IL-22;
Protect the intestines from Citrobacter rodentium infection.
[28]
ButyrateIn vitroICR mice crypt100μMElicit α-defensin secretion by Paneth cells;
Improve enteric innate immunity through potent microbicidal activities.
[79]
Acetate; Propionate; ButyrateIn vitroSheep ruminal tissue60 mM NaAc, 30 mM NaPr and 10 mM NaBuRelease protons;
Induce subacute ruminal acidosis.
[117]
Acetate; Propionate; ButyrateIn vitroCaco-2 cellsacetate (12.5, 25, and 50 mM), butyrate (5, 10, and 20 mM) and propionate (5, 10, and 20 mM)Increase the level of Hspa1a expression.
Up-regulate HSP70;
phosphorylate HSP1.
[76]
Acetate; Propionate; ButyrateIn vitroCaco-2 and T84 cellsAcetate: 0–20 mM, propionate: 0–10 mM, butyrate: 0–2.5 mMReduce IL-8 and IL-6 expression levels;
Reduce the activation of NF-κB, ERK, p38 MAPK, JNK, and Syk.
[81]
Acetate; Propionate; ButyrateIn vitroMODE-K and MC38 cell linesA mixture of 0.5 mM acetate, 0.01 mM propionate, and 0.01 mM butyrateInhibit DUSP6 by up-regulating miR-145 through decreasing the CEBPB expression.[82]
Acetate; Propionate; ButyrateIn vitroCaco-2 cells0.5 mM acetate, 0.01 mM butyrate, 0.01 mM propionateIncrease TER;
Improve the formation of tight junction;
Inhibit the activation of NLRP3 inflammasome and autophagy induced by LPS.
[78]
Acetate; Propionate; ButyrateIn vivoC57BL/6J mice200 mM propionate, 200 mM acetate or 100 mM butyrate in the drinking waterRepress IL-17- and IL-22-producing γδ T cells;
Reduce IL-17 production by γδ T cells by inhibiting HDAC.
[80]
Acetate; Propionate; ButyrateIn vivoC57BL/6J mice25 mM propionate, 40 mM butyrate and 67.5 mM acetate in drinking waterInhibit DUSP6 via up-regulating miR-145 by suppressing CEBPB;
Improve DAI.
[82]
Prevention and management of constipation
ButyrateIn vitroICCs0, 0.00005, 0.0005, 0.005, 0.05 and 0.5 mmol/LPromote mouse ICC proliferation by activating AKT/NF-κB signaling.[88]
ButyrateIn vivoKunming mice1.1% in the drinking waterPromote defecation;
Improve intestinal mobility;
Activate the AKT-NF-κB signaling pathway.
[88]
Acetate; Propionate; ButyrateIn vivoBALB/c miceDiets supplemented with 150 g/kg of either acetylated starch, propylated starch, butylated starchAcetylated starch and butylated starch relieve constipation;
Acetic acid increases WCF and SITR;
Butyric acid decreases the transit time through the gut.
[86]
Neuroprotection
AcetateIn vitroBV2 cells1200 μMImprove cognitive impairment;
Decrease the CD11b level;
Suppress neuroinflammation.
[93]
AcetateIn vivoAPP/PS1 transgenic and matched WT mice1.5 g/kg i.g.Inhibit the phosphorylation of NF-κB p65, ERK, and JNK;
Decrease COX-2 and IL-1β levels;
Increase GPR41 level.
[93]
PropionateIn vivoWestern albino rats75 mg/kg or 250 mg/kg i.g.Increase the levels of IFN-γ and caspase-3;
Decrease levels of nor-adrenaline, dopamine, and 5-HT.
[118]
PropionateIn vivoWestern albino rats75 mg/kg or 250 mg/kg i.g.Increase the levels of glutamate and the glutamate/glutamine ratio;
Decrease GABA, glutamine, and the GABA/glutamate ratio.
[119]
Anticancer
ValerateIn vitroHep3B, SNU-449, HepG2, THLE-3, MCF-7, MDA-MB-231, MCF-10A, A549, U-87 and A172, HeLa, DU145, and HL-60 cells0.5, 1, 2, 4, 8 mMSuppress colony formation, migration, and invasion of liver cancer cells;
Suppresses 3D spheroid formation of liver cancer cells.
[100]
ValerateIn vivoAthymic nude mice100 mg/kg tail injectionSuppress HCC development;
Improve the survival rate.
[100]
Butyrate; PropionateIn vivoC57BL/6 mice300 mg/kg acetate, 150 mg/kg propionate or 88 mg/kg butyrate i.p.Increase the expression of CCL20;
Reduce the recruitment of Th17 cells;
Inhibit the lung metastasis of melanoma cells.
[99]
Acetate; Butyrate; PropionateIn vivoBALB/c mice67.5 mM acetate, 40 mM butyrate and 25.9 mM propionate in drinking waterDecrease cell proliferation.[98]
Acetate; Propionate; Butyrate PentanoateIn vivoCD45.1 WT; CD45.2 WT; CD45.1 OT-I; CD45.2 Ffar2−/− Ffar3−/− mice;
CD45.2 FIR × tiger; Rag1−/− mice
0.5, 1.0, 2.5 mM Increase the anticancer activity of cytotoxic T lymphocytes and chimeric antigen receptor T cells via metabolic and epigenetic reprogramming.[101]
Prevention and management of arthritis
Acetate; Propionate; ButyrateIn vivoWT C57BL/6J mice and DBA/1J mice150 mM acetate, propionate or butyrate in drinking waterIncrease systemic bone mass via inducing the reprogramming of osteoclasts metabolism, enhancing glycolysis, and down-regulating TRAF6 and NFATc1;
Prevent bone loss after menopause;
Alleviate arthritis.
[112]
Acetate; Propionate; ButyrateIn vivoDBA/1JGpt, Ffar2fl/fl and Ffar2fl/fl/CD19-Cre mice 150 mM acetate, propionate or butyrate in drinking waterSynergistic treatment of CIA;
Regulate B cell differentiation via FFA2 receptors;
Suppress the inflammatory response.
[113]
Abbreviation: ABCG5, ATP-binding cassette transporters G5; ABCG8, ATP-binding cassette transporters G8; Ac, Acetate; Acc1, acetyl-CoA carboxylase-1; CaMKKβ, Ca2+-calmodulin-dependent protein kinase β; CCL20, chemokine (C-C motif) ligand 20; CEBPB, CCAAT enhancer-binding protein beta; CIA, collagen-induced arthritis; COX-2, cyclooxygenase-2; CPT-1α, carnitine palmitoyltransferase-1α; CYP7A1, cholesterol 7 alpha-hydroxylase; DAI, disease activity index; DC, dendritic cells; DUSP6, dual-specificity phosphatase 6; ERK, extracellular signal-regulated kinase; FABP4, fatty acid binding protein 4; FATP4, fatty acid transporter protein 4; FAS, fatty acid synthase; GABA, gamma amino butyric acid; GPR41, G-protein-coupled receptor 41; HAMS, High-amylose maize starch; HCC, hepatocellular carcinoma; HPMECs, human primary pulmonary microvascular endothelial cells; HSP, heat shock protein; ICCs, interstitial cells of Cajal; IFN-γ, Interferon-γ; Ig, immunoglobulin; i.g., intragastric administration; IL, interleukin; i.p., intraperitoneal injection; iPSC, induced pluripotent stem cell; I/R, ischemia/reperfusion; JNK, c-Jun N-terminal kinase; LDLR, low-density lipoprotein receptor; LIPE, lipase hormone sensitive; LPL, lipoprotein lipase; LPS, lipopolysaccharide; LITA, liposome-encapsulated acetate; MAPK, mitogen-activated protein kinase; miR, microRNA-145; NF-κB, nuclear factor-kappa B; NGNs, nodose ganglion neurons; NPC1l1, Niemann-Pick C1-like 1; NTS, nucleus tractus solitaries; OCFA, odd-chain fatty acids; Pr, Propionate; ROS, reactive oxygen species; SD, Sprague–Dawley; SITR, small intestinal transit rate; SREBP2, sterol-regulatory-element-binding protein 2; Syk, spleen tyrosine kinase; TER, transepithelial electrical resistance; TG, triglyceride; TGF-β, transforming growth factor-β; Th17, T helper 17; TNF, tumor necrosis factor; WCF, water content of feces; WT, wide type; YAMC, young adult mouse colon.
Table 3. Health benefits of SCFAs from clinical studies.
Table 3. Health benefits of SCFAs from clinical studies.
SCFAs SpeciesSTUDY TYPEIndividualsAdministration MethodsOutcomesRef.
Anti-obesity
PropionateSingle-blind crossover RCT20 healthy men Inulin-propionate ester (10 g/day) for 24 weeksReduce anticipatory reward responses in the human striatum to high-energy foods[40]
Cardio-protective activity
PropionateDouble-blind RCT62 participantsCalcium-Propionate (500 mg, twice daily) for 8 weeksReduce levels of LDL and non-high-density lipoprotein cholesterol[44]
Anti-diabetic activity
SCFAsRCT29 overweight/obese individualsMed-D intervention for 8 weeksIncrease plasma butyric acid
Improve postprandial glucose metabolism and insulin sensitivity
[72]
Abbreviations: LDL, low-density lipoprotein; Med-D, Mediterranean diet; RCT, Randomized controlled trial; SCFAs, short-chain fatty acids.

3. The Side Effects of SCFAs

Although most studies showed that SCFAs exerted health benefits, several studies found that some SCFAs could be useless or exhibit side effects in some conditions. Although SCFAs from inulin fermentation had potential to prevent or treat type 1 diabetes, direct SCFAs oral administration alone did not provide a significant impact on the diabetes, which might be attributed to oral administration did not allow for enough SCFAs to reach the site of action (cecum and colon) [116]. Moreover, a meta-analysis including 44 randomized controlled trial studies found that acetate, propionate, butyrate, and the mixed SCFAs had no effect on insulin and blood glucose in humans [120]. Another study showed that SCFAs accelerated the 3T3-L1 adipocyte differentiation and promoted lipid accumulation via modulating the expression of fatty acid metabolism-related enzymes in vitro, such as lipoprotein lipase, adipocyte fatty acid-binding protein 4, fatty acid transporter protein 4, and fatty acid synthase [115]. Additionally, a study suggested that, for those who had NAFLD and poor metabolic health, the gut-derived acetate could provide an extra precursor for de novo lipogenesis in the liver, which could increase hepatic lipid accumulation [15]. In a multi-organ model of ulcerative colitis (UC) ex vivo, SCFAs improved UC severity via increasing the production of ketone bodies, glycolysis, and lipogenesis, and significantly decreased innate immune activation of UC gut, but SCFAs also induced gut barrier disruption through metabolic reprograming during acute T cell-mediated inflammation [121]. Furthermore, some SCFAs could induce brain neurochemistry impairment. For example, an epidemiological study found that depressive women had higher concentrations of isocaproic acid than healthy subjects [92]. Another study showed that propionate caused brain neurochemistry impairment by increasing the levels of IFN-γ and caspase-3, as well as decreasing nor-adrenaline, dopamine, and 5-HT [118]. Moreover, propionate increased the level of glutamate and the glutamate/glutamine ratio, and decreased gamma amino butyric acid (GABA), glutamine and the GABA/glutamate ratio, which showed characteristics of autism [119].
In short, different SCFA types might exert different effects, and the bioactivities of SCFAs might depend on the health condition of hosts. When this section was compared with previous sections, the results from epidemiological studies could be inconsistent. In the future, more high-quality, large sample epidemiological studies are needed to verify the effects of SCFAs on humans.

4. Conclusions

SCFAs show various bioactivities, such as anti-inflammatory and immunoregulatory effects, as well as preventive and therapeutic effects on several diseases, including obesity, cardiovascular disease, liver disease, diabetes mellitus, IBDs, diarrhea, constipation, neurodegenerative diseases, neuropsychiatric diseases, cancers, arthritis and periodontal disease, and so on. Moreover, the underlying mechanisms are diverse, among which NF-κB, Nrf2, GPR41, and HDAC signaling pathways are highlighted. Furthermore, it is worth noting that, due to differences in SCFAs types and concentrations as well as health condition of hosts, some SCFAs could exhibit a double-sided effect. Therefore, in the application of SCFAs, attention should be paid to the selection of SCFA type, as well as the influence of their concentration and body condition. In the future, more bioactivities of SCFAs should be evaluated, and their mechanisms of action should be further studied. Furthermore, because most of the results comes from preclinical models, more clinical trials should be carried out to verify these potential effects of SCFAs on human beings. This paper is helpful for supporting people eating more dietary fiber, and for some SCFAs and dietary fiber to be developed into functional food to prevent and manage several diseases.

Author Contributions

Conceptualization, R.-G.X., R.-Y.G. and H.-B.L.; Writing—original draft preparation, R.-G.X., D.-D.Z., S.-X.W., S.-Y.H., A.S. (Adila Saimaiti), Z.-J.Y., A.S. (Ao Shang) and C.-N.Z.; Writing—review and editing, R.-Y.G. and H.-B.L.; Supervision, R.-Y.G. and H.-B.L.; Funding acquisition, R.-Y.G. and H.-B.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Local Financial Funds of National Agricultural Science and Technology Center, Chengdu (No. NASC2021KR01), and the Key Project of Guangdong Provincial Science and Technology Program (No. 2014B020205002).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We thank Hang-Yu Li in Department of Nutrition, School of Public Health, Sun Yat-Sen University, for collecting some materials for this paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
  2. Yamamura, R.; Nakamura, K.; Kitada, N.; Aizawa, T.; Shimizu, Y.; Nakamura, K.; Ayabe, T.; Kimura, T.; Tamakoshi, A. Associations of gut microbiota, dietary intake, and serum short-chain fatty acids with fecal short-chain fatty acids. Biosci Microbiota Food Health 2020, 39, 11–17. [Google Scholar] [CrossRef] [PubMed]
  3. Aldunate, M.; Srbinovski, D.; Hearps, A.C.; Latham, C.F.; Ramsland, P.A.; Gugasyan, R.; Cone, R.A.; Tachedjian, G. Antimicrobial and immune modulatory effects of lactic acid and short chain fatty acids produced by vaginal microbiota associated with eubiosis and bacterial vaginosis. Front. Physiol. 2015, 6, 164. [Google Scholar] [CrossRef] [PubMed]
  4. Keshari, S.; Balasubramaniam, A.; Myagmardoloonjin, B.; Herr, D.R.; Negari, I.P.; Huang, C.M. Butyric acid from probiotic staphylococcus epidermidis in the skin microbiome down-regulates the ultraviolet-induced pro-inflammatory IL-6 cytokine via short-chain fatty acid receptor. Int. J. Mol. Sci. 2019, 20, 4477. [Google Scholar] [CrossRef]
  5. Wang, M.; Wichienchot, S.; He, X.; Fu, X.; Huang, Q.; Zhang, B. In vitro colonic fermentation of dietary fibers: Fermentation rate, short-chain fatty acid production and changes in microbiota. Trends Food Sci. Technol. 2019, 88, 1–9. [Google Scholar] [CrossRef]
  6. Chen, X.F.; Chen, X.; Tang, X. Short-chain fatty acid, acylation and cardiovascular diseases. Clin. Sci. 2020, 134, 657–676. [Google Scholar] [CrossRef]
  7. van der Beek, C.M.; Dejong, C.H.C.; Troost, F.J.; Masclee, A.A.M.; Lenaerts, K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr. Rev. 2017, 75, 286–305. [Google Scholar] [CrossRef]
  8. McLoughlin, R.F.; Berthon, B.S.; Jensen, M.E.; Baines, K.J.; Wood, L.G. Short-chain fatty acids, prebiotics, synbiotics, and systemic inflammation: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2017, 106, 930–945. [Google Scholar] [CrossRef]
  9. Wong, J.M.; de Souza, R.; Kendall, C.W.; Emam, A.; Jenkins, D.J. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  10. Qin, R.; Wang, J.; Chao, C.; Yu, J.; Copeland, L.; Wang, S.; Wang, S. RS5 produced more butyric acid through regulating the microbial community of human gut microbiota. J. Agric. Food Chem. 2021, 69, 3209–3218. [Google Scholar] [CrossRef]
  11. Walker, A.W.; Duncan, S.H.; McWilliam Leitch, E.C.; Child, M.W.; Flint, H.J. pH and peptide supply can radically alter bacterial populations and short-chain fatty acid ratios within microbial communities from the human colon. Appl. Environ. Microbiol. 2005, 71, 3692–3700. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, Y.Q.; Zhu, Y.; Li, X.T.; Sun, B.G. Dynamic balancing of intestinal short-chain fatty acids: The crucial role of bacterial metabolism. Trends Food Sci. Technol. 2020, 100, 118–130. [Google Scholar] [CrossRef]
  13. Maltz, R.M.; Keirsey, J.; Kim, S.C.; Mackos, A.R.; Gharaibeh, R.Z.; Moore, C.C.; Xu, J.; Somogyi, A.; Bailey, M.T. Social stress affects colonic inflammation, the gut microbiome, and short-chain fatty acid levels and receptors. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 533–540. [Google Scholar] [CrossRef]
  14. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [PubMed]
  15. Chambers, E.S. Gut-derived short-chain fatty acids: A friend or foe for hepatic lipid metabolism? Nutr. Bull. 2019, 44, 154–159. [Google Scholar] [CrossRef]
  16. Li, H.Y.; Zhou, D.D.; Gan, R.Y.; Huang, S.Y.; Zhao, C.N.; Shang, A.; Xu, X.Y.; Li, H.B. Effects and mechanisms of probiotics, prebiotics, synbiotics, and postbiotics on metabolic diseases targeting gut microbiota: A narrative review. Nutrients 2021, 13, 3211. [Google Scholar] [CrossRef]
  17. Zhang, Y.J.; Li, S.; Gan, R.Y.; Zhou, T.; Xu, D.P.; Li, H.B. Impacts of gut bacteria on human health and diseases. Int. J. Mol. Sci. 2015, 16, 7493–7519. [Google Scholar] [CrossRef]
  18. Fernandez, J.; Redondo-Blanco, S.; Gutierrez-del-Rio, I.; Miguelez, E.M.; Villar, C.J.; Lombo, F. Colon microbiota fermentation of dietary prebiotics towards short-chain fatty acids and their roles as anti-inflammatory and antitumour agents: A review. J. Funct. Foods 2016, 25, 511–522. [Google Scholar] [CrossRef]
  19. Li, X.; Shimizu, Y.; Kimura, I. Gut microbial metabolite short-chain fatty acids and obesity. Biosci. Microbiota Food Health 2017, 36, 135–140. [Google Scholar] [CrossRef]
  20. Xia, W.; Dai, X.Y.; Ding, L.K.; Xi, Y.; Yan, M.; Zhang, M.; Wu, L.; Yi, C.X.; Xu, H.X. Three main short-chain fatty acids inhibit the activation of THP-1 cells by Mycoplasma pneumoniae. Biosci. Biotechnol. Biochem. 2021, 85, 923–930. [Google Scholar] [CrossRef]
  21. Wang, F.; Liu, J.; Weng, T.; Shen, K.; Chen, Z.; Yu, Y.; Huang, Q.; Wang, G.; Liu, Z.; Jin, S. The inflammation induced by lipopolysaccharide can be mitigated by short-chain fatty acid, butyrate, through upregulation of il-10 in septic shock. Scand. J. Immunol. 2017, 85, 258–263. [Google Scholar] [CrossRef] [PubMed]
  22. Vieira, A.T.; Galvao, I.; Macia, L.M.; Sernaglia, E.M.; Vinolo, M.A.R.; Garcia, C.C.; Tavares, L.P.; Amaral, F.A.; Sousa, L.P.; Martins, F.S.; et al. Dietary fiber and the short-chain fatty acid acetate promote resolution of neutrophilic inflammation in a model of gout in mice. J. Leukoc. Biol. 2017, 101, 275–284. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, D.; Gao, Z.Q.; Wang, Y.Y.; Wan, B.B.; Liu, G.; Chen, J.L.; Wu, Y.X.; Zhou, Q.; Jiang, S.Y.; Yu, R.Q.; et al. Sodium propionate enhances Nrf2-mediated protective defense against oxidative stress and inflammation in lipopolysaccharide-induced neonatal mice. J. Inflamm. Res. 2021, 14, 803–816. [Google Scholar] [CrossRef] [PubMed]
  24. Friscic, J.; Durholz, K.; Chen, X.; Engdahl, C.; Moller, L.; Schett, G.; Zaiss, M.M.; Hoffmann, M.H. Dietary derived propionate regulates pathogenic fibroblast function and ameliorates experimental arthritis and inflammatory tissue priming. Nutrients 2021, 13, 1643. [Google Scholar] [CrossRef]
  25. Olsson, A.; Gustavsen, S.; Nguyen, T.D.; Nyman, M.; Langkilde, A.R.; Hansen, T.H.; Sellebjerg, F.; Oturai, A.B.; Sondergaard, H.B. Serum short-chain fatty acids and associations with inflammation in newly diagnosed patients with multiple sclerosis and healthy controls. Front. Immunol. 2021, 12, 661493. [Google Scholar] [CrossRef]
  26. Kim, M.; Kim, C.H. Regulation of humoral immunity by gut microbial products. Gut Microbes 2017, 8, 392–399. [Google Scholar] [CrossRef]
  27. Cait, A.; Hughes, M.R.; Antignano, F.; Cait, J.; Dimitriu, P.A.; Maas, K.R.; Reynolds, L.A.; Hacker, L.; Mohr, J.; Finlay, B.B.; et al. Microbiome-driven allergic lung inflammation is ameliorated by short-chain fatty acids. Mucosal Immunol. 2018, 11, 785–795. [Google Scholar] [CrossRef]
  28. Yang, W.J.; Yu, T.M.; Huang, X.S.; Bilotta, A.J.; Xu, L.Q.; Lu, Y.; Sun, J.R.; Pan, F.; Zhou, J.; Zhang, W.B.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
  29. Zou, F.; Qiu, Y.; Huang, Y.; Zou, H.; Cheng, X.; Niu, Q.; Luo, A.; Sun, J. Effects of short-chain fatty acids in inhibiting HDAC and activating p38 MAPK are critical for promoting B10 cell generation and function. Cell Death Dis. 2021, 12, 582. [Google Scholar] [CrossRef]
  30. Cao, S.Y.; Zhao, C.N.; Xu, X.Y.; Tang, G.Y.; Corke, H.; Gan, R.Y.; Li, H.B. Dietary plants, gut microbiota, and obesity: Effects and mechanisms. Trends Food Sci. Technol. 2019, 92, 194–204. [Google Scholar] [CrossRef]
  31. Prentice, P.M.; Schoemaker, M.H.; Vervoort, J.; Hettinga, K.; Lambers, T.T.; van Tol, E.A.F.; Acerini, C.L.; Olga, L.; Petry, C.J.; Hughes, I.A.; et al. Human milk short-chain fatty acid composition is associated with adiposity outcomes in infants. J. Nutr. 2019, 149, 716–722. [Google Scholar] [CrossRef] [PubMed]
  32. Goswami, C.; Iwasaki, Y.; Yada, T. Short-chain fatty acids suppress food intake by activating vagal afferent. J. Nutr. Biochem. 2018, 57, 130–135. [Google Scholar] [CrossRef] [PubMed]
  33. Jiao, A.R.; Yu, B.; He, J.; Yu, J.; Zheng, P.; Luo, Y.H.; Luo, J.Q.; Mao, X.B.; Chen, D.W. Short chain fatty acids could prevent fat deposition in pigs via regulating related hormones and genes. Food Funct. 2020, 11, 1845–1855. [Google Scholar] [CrossRef] [PubMed]
  34. Jiao, A.R.; Yu, B.; He, J.; Yu, J.; Zheng, P.; Luo, Y.H.; Luo, J.Q.; Yan, H.; Wang, Q.Y.; Wang, H.F.; et al. Sodium acetate, propionate, and butyrate reduce fat accumulation in mice via modulating appetite and relevant genes. Nutrition 2021, 87, 111198. [Google Scholar] [CrossRef]
  35. Shah, S.; Fillier, T.; Pham, T.H.; Thomas, R.; Cheema, S.K. Intraperitoneal administration of short-chain fatty acids improves lipid metabolism of long-evans rats in a sex-specific manner. Nutrients 2021, 13, 892. [Google Scholar] [CrossRef]
  36. Higashimura, Y.; Naito, Y.; Takagi, T.; Uchiyama, K.; Mizushima, K.; Yoshikawa, T. Propionate promotes fatty acid oxidation through the up-regulation of peroxisome proliferator-activated receptor α in intestinal epithelial cells. J. Nutr. Sci. Vitaminol. 2015, 61, 511–515. [Google Scholar] [CrossRef]
  37. Shimizu, H.; Masujima, Y.; Ushiroda, C.; Mizushima, R.; Taira, S.; Ohue-Kitano, R.; Kimura, I. Dietary short-chain fatty acid intake improves the hepatic metabolic condition via FFAR3. Sci. Rep. 2019, 9, 16574. [Google Scholar] [CrossRef]
  38. Weitkunat, K.; Schumann, S.; Nickel, D.; Kappo, K.A.; Petzke, K.J.; Kipp, A.P.; Blaut, M.; Klaus, S. Importance of propionate for the repression of hepatic lipogenesis and improvement of insulin sensitivity in high-fat diet-induced obesity. Mol. Nutr. Food Res. 2016, 60, 2611–2621. [Google Scholar] [CrossRef]
  39. Sahuri-Arisoylu, M.; Brody, L.P.; Parkinson, J.R.; Parkes, H.; Navaratnam, N.; Miller, A.D.; Thomas, E.L.; Frost, G.; Bell, J.D. Reprogramming of hepatic fat accumulation and ’browning’ of adipose tissue by the short-chain fatty acid acetate. Int. J. Obes. 2016, 40, 955–963. [Google Scholar] [CrossRef]
  40. Byrne, C.S.; Chambers, E.S.; Alhabeeb, H.; Chhina, N.; Morrison, D.J.; Preston, T.; Tedford, C.; Fitzpatrick, J.; Irani, C.; Busza, A.; et al. Increased colonic propionate reduces anticipatory reward responses in the human striatum to high-energy foods. Am. J. Clin. Nutr. 2016, 104, 5–14. [Google Scholar] [CrossRef] [Green Version]
  41. Cao, S.Y.; Zhao, C.N.; Gan, R.Y.; Xu, X.Y.; Wei, X.L.; Corke, H.; Atanasov, A.G.; Li, H.B. Effects and Mechanisms of Tea and Its Bioactive Compounds for the Prevention and Treatment of Cardiovascular Diseases: An Updated Review. Antioxidants 2019, 8, 166. [Google Scholar] [CrossRef] [PubMed]
  42. Tindall, A.M.; Petersen, K.S.; Kris-Etherton, P.M. Dietary patterns affect the gut microbiome-the link to risk of cardiometabolic diseases. J. Nutr. 2018, 148, 1402–1407. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, Y.; Xu, C.; Huang, R.; Song, J.; Li, D.; Xia, M. Butyrate from pectin fermentation inhibits intestinal cholesterol absorption and attenuates atherosclerosis in apolipoprotein E-deficient mice. J. Nutr. Biochem. 2018, 56, 175–182. [Google Scholar] [CrossRef] [PubMed]
  44. Haghikia, A.; Zimmermann, F.; Schumann, P.; Jasina, A.; Roessler, J.; Schmidt, D.; Heinze, P.; Kaisler, J.; Nageswaran, V.; Aigner, A.; et al. Propionate attenuates atherosclerosis by immune-dependent regulation of intestinal cholesterol metabolism. Eur. Heart J. 2022, 43, 518–533. [Google Scholar] [CrossRef]
  45. Zhao, Y.M.; Liu, J.H.; Hao, W.J.; Zhu, H.Y.; Liang, N.; He, Z.Y.; Ma, K.Y.; Chen, Z.Y. Structure-specific effects of short-chain fatty acids on plasma cholesterol concentration in male syrian hamsters. J. Agric. Food Chem. 2017, 65, 10984–10992. [Google Scholar] [CrossRef]
  46. Kaye, D.M.; Shihata, W.A.; Jama, H.A.; Tsyganov, K.; Ziemann, M.; Kiriazis, H.; Horlock, D.; Vijay, A.; Giam, B.; Vinh, A.; et al. Deficiency of prebiotic fiber and insufficient signaling through gut metabolite-sensing receptors leads to cardiovascular disease. Circulation 2020, 141, 1393–1403. [Google Scholar] [CrossRef]
  47. Hsu, C.N.; Chang-Chien, G.P.; Lin, S.; Hou, C.Y.; Tain, Y.L. Targeting on gut microbial metabolite trimethylamine-N-oxide and short-chain fatty acid to prevent maternal high-fructose-diet-induced developmental programming of hypertension in adult male offspring. Mol. Nutr. Food Res. 2019, 63, e1900073. [Google Scholar] [CrossRef]
  48. Bartolomaeus, H.; Balogh, A.; Yakoub, M.; Homann, S.; Markó, L.; Höges, S.; Tsvetkov, D.; Krannich, A.; Wundersitz, S.; Avery, E.G.; et al. Short-chain fatty acid propionate protects from hypertensive cardiovascular damage. Circulation 2019, 139, 1407–1421. [Google Scholar] [CrossRef]
  49. Yu, Z.; Han, J.; Chen, H.; Wang, Y.; Zhou, L.; Wang, M.; Zhang, R.; Jin, X.; Zhang, G.; Wang, C.; et al. Oral supplementation with butyrate improves myocardial ischemia/reperfusion injury via a gut-brain neural circuit. Front. Cardiovasc. Med. 2021, 8, 718674. [Google Scholar] [CrossRef]
  50. Tang, T.W.H.; Chen, H.C.; Chen, C.Y.; Yen, C.Y.T.; Lin, C.J.; Prajnamitra, R.P.; Chen, L.L.; Ruan, S.C.; Lin, J.H.; Lin, P.J.; et al. Loss of gut microbiota alters immune system composition and cripples postinfarction cardiac repair. Circulation 2019, 139, 647–659. [Google Scholar] [CrossRef]
  51. Huang, W.F.; Kong, D.S. The intestinal microbiota as a therapeutic target in the treatment of NAFLD and ALD. Biomed. Pharmacother. 2021, 135, 111235. [Google Scholar] [CrossRef]
  52. Li, B.Y.; Mao, Q.Q.; Zhou, D.D.; Luo, M.; Gan, R.Y.; Li, H.Y.; Huang, S.Y.; Saimaiti, A.; Shang, A.; Li, H.B. Effects of tea against alcoholic fatty liver disease by modulating gut microbiota in chronic alcohol-exposed mice. Foods 2021, 10, 1232. [Google Scholar] [CrossRef]
  53. Meng, X.; Li, S.; Li, Y.; Gan, R.Y.; Li, H.B. Gut microbiota’s relationship with liver disease and role in hepatoprotection by dietary natural products and probiotics. Nutrients 2018, 10, 1457. [Google Scholar] [CrossRef]
  54. Bloom, P.P.; Luévano, J.M., Jr.; Miller, K.J.; Chung, R.T. Deep stool microbiome analysis in cirrhosis reveals an association between short-chain fatty acids and hepatic encephalopathy. Ann. Hepatol. 2021, 25, 100333. [Google Scholar] [CrossRef]
  55. Seitz, H.K.; Bataller, R.; Cortez-Pinto, H.; Gao, B.; Gual, A.; Lackner, C.; Mathurin, P.; Mueller, S.; Szabo, G.; Tsukamoto, H. Alcoholic liver disease. Nat. Rev. Dis. Primers 2018, 4, 16. [Google Scholar] [CrossRef]
  56. Xu, Q.; Zhang, R.; Mu, Y.; Song, Y.; Hao, N.; Wei, Y.; Wang, Q.; Mackay, C.R. Propionate ameliorates alcohol-induced liver injury in mice via the gut-liver axis: Focus on the improvement of intestinal permeability. J. Agric. Food Chem. 2022, 70, 6084–6096. [Google Scholar] [CrossRef]
  57. Zhang, S.; Zhao, J.; Xie, F.; He, H.; Johnston, L.J.; Dai, X.; Wu, C.; Ma, X. Dietary fiber-derived short-chain fatty acids: A potential therapeutic target to alleviate obesity-related nonalcoholic fatty liver disease. Obes. Rev. 2021, 22, e13316. [Google Scholar] [CrossRef]
  58. Liu, W.X.; Luo, X.L.; Tang, J.; Mo, Q.F.; Zhong, H.; Zhang, H.; Feng, F.Q. A bridge for short-chain fatty acids to affect inflammatory bowel disease, type 1 diabetes, and non-alcoholic fatty liver disease positively: By changing gut barrier. Eur. J. Nutr. 2021, 60, 2317–2330. [Google Scholar] [CrossRef]
  59. Li, M.; Wang, J.; Wang, F.F.; Strappe, P.; Liu, W.T.; Zheng, J.X.; Zhou, Z.K.; Zhang, Y. Microbiota fermentation characteristics of acylated starches and the regulation mechanism of short-chain fatty acids on hepatic steatosis. Food Funct. 2021, 12, 8659–8668. [Google Scholar] [CrossRef]
  60. Batista, A.G.; da Silva-Maia, J.K.; Mendonca, M.C.P.; Soares, E.S.; Lima, G.C.; Bogusz, S.; da Cruz-Hofling, M.A.; Marostica, M.R. Jaboticaba berry peel intake increases short chain fatty acids production and prevent hepatic steatosis in mice fed high-fat diet. J. Funct. Foods 2018, 48, 266–274. [Google Scholar] [CrossRef]
  61. Li, W.F.; Zhang, K.; Yang, H.Y. Pectin alleviates high fat (lard) diet-induced nonalcoholic fatty liver disease in mice: Possible role of short-chain fatty acids and gut microbiota regulated by pectin. J. Agric. Food Chem. 2018, 66, 8015–8025. [Google Scholar] [CrossRef]
  62. Yang, F.; Feng, B.; Niu, Y.J.; Hu, C.Y.; Meng, Y.H. Fu instant tea ameliorates fatty liver by improving microbiota dysbiosis and elevating short-chain fatty acids in the intestine of mice fed a high-fat diet. Food Biosci. 2021, 42, 101207. [Google Scholar] [CrossRef]
  63. Mun, S.J.; Lee, J.; Chung, K.S.; Son, M.Y.; Son, M.J. Effect of microbial short-chain fatty acids on CYP3A4-mediated metabolic activation of human pluripotent stem cell-derived liver organoids. Cells 2021, 10, 126. [Google Scholar] [CrossRef]
  64. Li, B.Y.; Xu, X.Y.; Gan, R.Y.; Sun, Q.C.; Meng, J.M.; Shang, A.; Mao, Q.Q.; Li, H.B. Targeting gut microbiota for the prevention and management of diabetes mellitus by dietary natural products. Foods 2019, 8, 440. [Google Scholar] [CrossRef]
  65. Meng, J.M.; Cao, S.Y.; Wei, X.L.; Gan, R.Y.; Wang, Y.F.; Cai, S.X.; Xu, X.Y.; Zhang, P.Z.; Li, H.B. Effects and mechanisms of tea for the prevention and management of diabetes mellitus and diabetic complications: An updated review. Antioxidants 2019, 8, 170. [Google Scholar] [CrossRef]
  66. Sanna, S.; van Zuydam, N.R.; Mahajan, A.; Kurilshikov, A.; Vich Vila, A.; Võsa, U.; Mujagic, Z.; Masclee, A.A.M.; Jonkers, D.; Oosting, M.; et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat. Genet. 2019, 51, 600–605. [Google Scholar] [CrossRef]
  67. Mandaliya, D.K.; Patel, S.; Seshadri, S. The combinatorial effect of acetate and propionate on high-fat diet induced diabetic inflammation or metaflammation and T cell polarization. Inflammation 2021, 44, 68–79. [Google Scholar] [CrossRef]
  68. Hernández, M.A.G.; Canfora, E.E.; Jocken, J.W.E.; Blaak, E.E. The short-chain fatty acid acetate in body weight control and insulin sensitivity. Nutrients 2019, 11, 1943. [Google Scholar] [CrossRef] [PubMed]
  69. McNabney, S.M.; Henagan, T.M. Short chain fatty acids in the colon and peripheral tissues: A focus on butyrate, colon cancer, obesity and insulin resistance. Nutrients 2017, 9, 1348. [Google Scholar] [CrossRef]
  70. Yoshida, H.; Ishii, M.; Akagawa, M. Propionate suppresses hepatic gluconeogenesis via GPR43/AMPK signaling pathway. Arch. Biochem. Biophys. 2019, 672, 108057. [Google Scholar] [CrossRef]
  71. Li, M.; Wang, F.F.; Wang, J.; Wang, A.Q.; Yao, X.; Strappe, P.; Zhou, Z.K.; Wu, Q.H.; Guo, T.L. Starch acylation of different short-chain fatty acids and its corresponding influence on gut microbiome and diabetic indexes. Food Chem. 2022, 389, 133089. [Google Scholar] [CrossRef]
  72. Vitale, M.; Giacco, R.; Laiola, M.; Della Pepa, G.; Luongo, D.; Mangione, A.; Salamone, D.; Vitaglione, P.; Ercolini, D.; Rivellese, A.A. Acute and chronic improvement in postprandial glucose metabolism by a diet resembling the traditional Mediterranean dietary pattern: Can SCFAs play a role? Clin. Nutr. 2021, 40, 428–437. [Google Scholar] [CrossRef]
  73. Blaak, E.E.; Canfora, E.E.; Theis, S.; Frost, G.; Groen, A.K.; Mithieux, G.; Nauta, A.; Scott, K.; Stahl, B.; van Harsselaar, J.; et al. Short chain fatty acids in human gut and metabolic health. Benef. Microbes 2020, 11, 411–455. [Google Scholar] [CrossRef]
  74. Bilotta, A.J.; Ma, C.; Yang, W.; Yu, Y.; Yu, Y.; Zhao, X.; Zhou, Z.; Yao, S.; Dann, S.M.; Cong, Y. Propionate enhances cell speed and persistence to promote intestinal epithelial turnover and repair. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 1023–1044. [Google Scholar] [CrossRef]
  75. Li, Q.; Chen, H.; Zhang, M.; Wu, T.; Liu, R. Altered short chain fatty acid profiles induced by dietary fiber intervention regulate AMPK levels and intestinal homeostasis. Food Funct. 2019, 10, 7174–7187. [Google Scholar] [CrossRef]
  76. Adesina, P.A.; Isayama, K.; Sitolo, G.C.; Yamamoto, Y.; Suzuki, T. Propionate and dietary fermentable fibers upregulate intestinal heat shock protein70 in intestinal Caco-2 cells and mouse colon. J. Agric. Food Chem. 2021, 69, 8460–8470. [Google Scholar] [CrossRef]
  77. Chang, Y.H.; Jeong, C.H.; Cheng, W.N.; Choi, Y.; Shin, D.M.; Lee, S.; Han, S.G. Quality characteristics of yogurts fermented with short-chain fatty acid-producing probiotics and their effects on mucin production and probiotic adhesion onto human colon epithelial cells. J. Dairy Sci. 2021, 104, 7415–7425. [Google Scholar] [CrossRef]
  78. Feng, Y.; Wang, Y.; Wang, P.; Huang, Y.; Wang, F. Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell. Physiol. Biochem. 2018, 49, 190–205. [Google Scholar] [CrossRef]
  79. Takakuwa, A.; Nakamura, K.; Kikuchi, M.; Sugimoto, R.; Ohira, S.; Yokoi, Y.; Ayabe, T. Butyric acid and leucine induce α-defensin secretion from small intestinal paneth cells. Nutrients 2019, 11, 2817. [Google Scholar] [CrossRef]
  80. Dupraz, L.; Magniez, A.; Rolhion, N.; Richard, M.L.; Da Costa, G.; Touch, S.; Mayeur, C.; Planchais, J.; Agus, A.; Danne, C.; et al. Gut microbiota-derived short-chain fatty acids regulate IL-17 production by mouse and human intestinal γδ T cells. Cell Rep. 2021, 36, 109332. [Google Scholar] [CrossRef]
  81. Hung, T.V.; Suzuki, T. Short-chain fatty acids suppress inflammatory reactions in Caco-2 cells and mouse colons. J. Agric. Food Chem. 2018, 66, 108–117. [Google Scholar] [CrossRef]
  82. Liu, Q.; Peng, Z.; Zhou, L.; Peng, R.; Li, X.; Zuo, W.; Gou, J.; Zhou, F.; Yu, S.; Huang, M.; et al. Short-chain fatty acid decreases the expression of CEBPB to inhibit mir-145-mediated DUSP6 and thus further suppresses intestinal inflammation. Inflammation 2022, 45, 372–386. [Google Scholar] [CrossRef]
  83. Ohkusa, T.; Koido, S.; Nishikawa, Y.; Sato, N. Gut microbiota and chronic constipation: A review and update. Front. Med. 2019, 6, 19. [Google Scholar] [CrossRef]
  84. Shi, Y.; Chen, Q.; Huang, Y.; Ni, L.; Liu, J.; Jiang, J.; Li, N. Function and clinical implications of short-chain fatty acids in patients with mixed refractory constipation. Colorectal Dis. 2016, 18, 803–810. [Google Scholar] [CrossRef]
  85. Zhuang, M.; Shang, W.T.; Ma, Q.C.; Strappe, P.; Zhou, Z.K. Abundance of probiotics and butyrate-production microbiome manages constipation via short-chain fatty acids production and hormones secretion. Mol. Nutr. Food Res. 2019, 63, e1801187. [Google Scholar] [CrossRef]
  86. Wang, L.L.; Cen, S.; Wang, G.; Lee, Y.K.; Zhao, J.X.; Zhan, H.; Chen, W. Acetic acid and butyric acid released in large intestine play different roles in the alleviation of constipation. J. Funct. Foods 2020, 69, 103953. [Google Scholar] [CrossRef]
  87. Wang, L.L.; Hu, L.J.; Yan, S.; Jiang, T.; Fang, S.G.; Wang, G.; Zhao, J.X.; Zhang, H.; Chen, W. Effects of different oligosaccharides at various dosages on the composition of gut microbiota and short-chain fatty acids in mice with constipation. Food Funct. 2017, 8, 1966–1978. [Google Scholar] [CrossRef]
  88. He, Q.L.; Han, C.P.; Huang, L.; Yang, H.J.; Hu, J.C.; Chen, H.X.; Dou, R.X.; Ren, D.L.; Lin, H.C. Astragaloside IV alleviates mouse slow transit constipation by modulating gut microbiota profile and promoting butyric acid generation. J. Cell. Mol. Med. 2020, 24, 9349–9361. [Google Scholar] [CrossRef]
  89. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  90. Cheng, Y.; Liu, J.; Ling, Z. Short-chain fatty acids-producing probiotics: A novel source of psychobiotics. Crit. Rev. Food Sci. Nutr. 2021; in press. [Google Scholar] [CrossRef]
  91. Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
  92. Skonieczna-Żydecka, K.; Grochans, E.; Maciejewska, D.; Szkup, M.; Schneider-Matyka, D.; Jurczak, A.; Łoniewski, I.; Kaczmarczyk, M.; Marlicz, W.; Czerwińska-Rogowska, M.; et al. Faecal short chain fatty acids profile is changed in polish depressive women. Nutrients 2018, 10, 1939. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, J.; Li, H.; Gong, T.; Chen, W.; Mao, S.; Kong, Y.; Yu, J.; Sun, J. Anti-neuroinflammatory effect of short-chain fatty acid acetate against Alzheimer’s disease via upregulating GPR41 and inhibiting ERK/JNK/NF-κB. J. Agric. Food Chem. 2020, 68, 7152–7161. [Google Scholar] [CrossRef] [PubMed]
  94. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  95. Tao, J.; Li, S.; Gan, R.Y.; Zhao, C.N.; Meng, X.; Li, H.B. Targeting gut microbiota with dietary components on cancer: Effects and potential mechanisms of action. Crit. Rev. Food Sci. Nutr. 2020, 60, 1025–1037. [Google Scholar] [CrossRef] [PubMed]
  96. Matsuya-Ogawa, M.; Shibata, T.; Itoh, H.; Murakami, H.; Yaguchi, C.; Sugihara, K.; Kanayama, N. Oncoprotective effects of short-chain fatty acids on uterine cervical neoplasia. Nutr. Cancer 2019, 71, 312–319. [Google Scholar] [CrossRef]
  97. Casanova, M.R.; Azevedo-Silva, J.; Rodrigues, L.R.; Preto, A. Colorectal cancer cells increase the production of short chain fatty acids by propionibacterium freudenreichii impacting on cancer cells survival. Front. Nutr. 2018, 5, 44. [Google Scholar] [CrossRef]
  98. Tian, Y.; Xu, Q.; Sun, L.; Ye, Y.; Ji, G. Short-chain fatty acids administration is protective in colitis-associated colorectal cancer development. J. Nutr. Biochem. 2018, 57, 103–109. [Google Scholar] [CrossRef]
  99. Chen, L.; Zhou, X.; Wang, Y.; Wang, D.; Ke, Y.; Zeng, X. Propionate and butyrate produced by gut microbiota after probiotic supplementation attenuate lung metastasis of melanoma cells in mice. Mol. Nutr. Food Res. 2021, 65, e2100096. [Google Scholar] [CrossRef]
  100. Han, R.; Nusbaum, O.; Chen, X.; Zhu, Y. Valeric acid suppresses liver cancer development by acting as a novel HDAC inhibitor. Mol. Ther.-Oncolytics 2020, 19, 8–18. [Google Scholar] [CrossRef]
  101. Luu, M.; Riester, Z.; Baldrich, A.; Reichardt, N.; Yuille, S.; Busetti, A.; Klein, M.; Wempe, A.; Leister, H.; Raifer, H.; et al. Microbial short-chain fatty acids modulate CD8(+) T cell responses and improve adoptive immunotherapy for cancer. Nat. Commun. 2021, 12, 4077. [Google Scholar] [CrossRef] [PubMed]
  102. Fattahi, Y.; Heidari, H.R.; Khosroushahi, A.Y. Review of short-chain fatty acids effects on the immune system and cancer. Food Biosci. 2020, 38, 100793. [Google Scholar] [CrossRef]
  103. Thiruvengadam, M.; Subramanian, U.; Venkidasamy, B.; Thirupathi, P.; Samynathan, R.; Shariati, M.A.; Rebezov, M.; Chung, I.M.; Rengasamy, K.R.R. Emerging role of nutritional short-chain fatty acids (SCFAs) against cancer via modulation of hematopoiesis. Crit. Rev. Food Sci. Nutr. 2021; in press. [Google Scholar] [CrossRef] [PubMed]
  104. Schulthess, J.; Pandey, S.; Capitani, M.; Rue-Albrecht, K.C.; Arnold, I.; Franchini, F.; Chomka, A.; Ilott, N.E.; Johnston, D.G.W.; Pires, E.; et al. The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity 2019, 50, 432–445.e7. [Google Scholar] [CrossRef] [PubMed]
  105. Asadpoor, M.; Ithakisiou, G.N.; Henricks, P.A.J.; Pieters, R.; Folkerts, G.; Braber, S. Non-digestible oligosaccharides and short chain fatty acids as therapeutic targets against enterotoxin-producing bacteria and their toxins. Toxins 2021, 13, 175. [Google Scholar] [CrossRef]
  106. Peng, M.; Biswas, D. Short chain and polyunsaturated fatty acids in host gut health and foodborne bacterial pathogen inhibition. Crit. Rev. Food Sci. Nutr. 2017, 57, 3987–4002. [Google Scholar] [CrossRef]
  107. Venditti, T.; Ladu, G.; Cubaiu, L.; Myronycheva, O.; D’Hallewin, G. Repeated treatments with acetic acid vapors during storage preserve table grapes fruit quality. Postharvest. Biol. Technol. 2017, 125, 91–98. [Google Scholar] [CrossRef]
  108. Wang, X.; Zhu, M.; Loor, J.J.; Jiang, Q.; Zhu, Y.; Li, W.; Du, X.; Song, Y.; Gao, W.; Lei, L.; et al. Propionate alleviates fatty acid-induced mitochondrial dysfunction, oxidative stress, and apoptosis by upregulating PPARG coactivator 1 alpha in hepatocytes. J. Dairy Sci. 2022, 105, 4581–4592. [Google Scholar] [CrossRef]
  109. Heath, A.M.; Haszard, J.J.; Galland, B.C.; Lawley, B.; Rehrer, N.J.; Drummond, L.N.; Sims, I.M.; Taylor, R.W.; Otal, A.; Taylor, B.; et al. Association between the faecal short-chain fatty acid propionate and infant sleep. Eur. J. Clin. Nutr. 2020, 74, 1362–1365. [Google Scholar] [CrossRef]
  110. Frampton, J.; Murphy, K.G.; Frost, G.; Chambers, E.S. Short-chain fatty acids as potential regulators of skeletal muscle metabolism and function. Nat. Metab. 2020, 2, 840–848. [Google Scholar] [CrossRef]
  111. Kondo, T.; Chiba, T.; Tousen, Y. Short-chain fatty acids, acetate and propionate, directly upregulate osteoblastic differentiation. Int. J. Food Sci. Nutr. 2022; in press. [Google Scholar] [CrossRef]
  112. Lucas, S.; Omata, Y.; Hofmann, J.; Böttcher, M.; Iljazovic, A.; Sarter, K.; Albrecht, O.; Schulz, O.; Krishnacoumar, B.; Krönke, G.; et al. Short-chain fatty acids regulate systemic bone mass and protect from pathological bone loss. Nat. Commun. 2018, 9, 55. [Google Scholar] [CrossRef]
  113. Yao, Y.; Cai, X.; Zheng, Y.; Zhang, M.; Fei, W.; Sun, D.; Zhao, M.; Ye, Y.; Zheng, C. Short-chain fatty acids regulate B cells differentiation via the FFA2 receptor to alleviate rheumatoid arthritis. Br. J. Pharmacol. 2022; in press. [Google Scholar] [CrossRef]
  114. Dahlstrand Rudin, A.; Khamzeh, A.; Venkatakrishnan, V.; Basic, A.; Christenson, K.; Bylund, J. Short chain fatty acids released by Fusobacterium nucleatum are neutrophil chemoattractants acting via free fatty acid receptor 2 (FFAR2). Cell. Microbiol. 2021, 23, e13348. [Google Scholar] [CrossRef]
  115. Yu, H.N.; Li, R.; Huang, H.Y.; Yao, R.; Shen, S.R. Short-chain fatty acids enhance the lipid accumulation of 3T3-L1 cells by modulating the expression of enzymes of fatty acid metabolism. Lipids 2018, 53, 77–84. [Google Scholar] [CrossRef]
  116. Zou, J.; Reddivari, L.; Shi, Z.; Li, S.; Wang, Y.; Bretin, A.; Ngo, V.L.; Flythe, M.; Pellizzon, M.; Chassaing, B.; et al. Inulin fermentable fiber ameliorates type I diabetes via il22 and short-chain fatty acids in experimental models. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 983–1000. [Google Scholar] [CrossRef]
  117. Meissner, S.; Hagen, F.; Deiner, C.; Günzel, D.; Greco, G.; Shen, Z.; Aschenbach, J.R. Key role of short-chain fatty acids in epithelial barrier failure during ruminal acidosis. J. Dairy Sci. 2017, 100, 6662–6675. [Google Scholar] [CrossRef]
  118. Al-Salem, H.S.; Bhat, R.S.; Al-Ayadhi, L.; El-Ansary, A. Therapeutic potency of bee pollen against biochemical autistic features induced through acute and sub-acute neurotoxicity of orally administered propionic acid. BMC Complement. Altern. Med. 2016, 16, 120. [Google Scholar] [CrossRef]
  119. El-Ansary, A.; Al-Salem, H.S.; Asma, A.; Al-Dbass, A. Glutamate excitotoxicity induced by orally administered propionic acid, a short chain fatty acid can be ameliorated by bee pollen. Lipids Health Dis. 2017, 16, 96. [Google Scholar] [CrossRef]
  120. Cherta-Murillo, A.; Pugh, J.E.; Alaraj-Alshehhi, S.; Hajjar, D.; Chambers, E.S.; Frost, G.S. The effect of short-chain fatty acids on glycemic control in humans: A systematic review and Meta-analysis. Am. J. Clin. Nutr. 2022; in press. [Google Scholar] [CrossRef]
  121. Trapecar, M.; Communal, C.; Velazquez, J.; Maass, C.A.; Huang, Y.J.; Schneider, K.; Wright, C.W.; Butty, V.; Eng, G.; Yilmaz, O.; et al. Gut-liver physiomimetics reveal paradoxical modulation of IBD-related inflammation by short-chain fatty acids. Cell Systems 2020, 10, 223–239.e9. [Google Scholar] [CrossRef]
Figure 1. (AC) The chemical structures of three major SCFAs.
Figure 1. (AC) The chemical structures of three major SCFAs.
Foods 11 02863 g001
Figure 2. Health benefits and the related mechanisms of SCFAs. ↓ represents decrease, and ↑ represents increase. Abbreviation: COX-2, cyclooxygenase-2; GPR41, G-protein-coupled receptor 41; IL, interleukin; NF-κB, nuclear factor-kappa B; SITR, small intestinal transit rate; WCF, water content of feces.
Figure 2. Health benefits and the related mechanisms of SCFAs. ↓ represents decrease, and ↑ represents increase. Abbreviation: COX-2, cyclooxygenase-2; GPR41, G-protein-coupled receptor 41; IL, interleukin; NF-κB, nuclear factor-kappa B; SITR, small intestinal transit rate; WCF, water content of feces.
Foods 11 02863 g002
Figure 3. The main effects and mechanisms of SCFAs on obesity. ↓ represents decrease, and ↑ represents increase. Abbreviation: Acc1, acetyl-CoA carboxylase-1; BAT, brown adipose tissue; CD137, tumor necrosis factor receptor superfamily member 9; COX IV, cytochrome-C oxidase IV; CPT, carnitine palmitoyl transferase; FAS, fatty acid synthase; FFAR2, free fatty acid receptor 2; GLP-1, glucagon-like peptide 1; LIPE, lipase hormone sensitive; NTS, nucleus tractus solitaries; PYY, peptide YY; SCFA, short-chain fatty acid; SREBP-1C, sterol-regulatory element-binding protein 1C; Tfam, mitochondrial transcription factor A; TFPα, trifunctional protein alpha; WAT, white adipose tissue.
Figure 3. The main effects and mechanisms of SCFAs on obesity. ↓ represents decrease, and ↑ represents increase. Abbreviation: Acc1, acetyl-CoA carboxylase-1; BAT, brown adipose tissue; CD137, tumor necrosis factor receptor superfamily member 9; COX IV, cytochrome-C oxidase IV; CPT, carnitine palmitoyl transferase; FAS, fatty acid synthase; FFAR2, free fatty acid receptor 2; GLP-1, glucagon-like peptide 1; LIPE, lipase hormone sensitive; NTS, nucleus tractus solitaries; PYY, peptide YY; SCFA, short-chain fatty acid; SREBP-1C, sterol-regulatory element-binding protein 1C; Tfam, mitochondrial transcription factor A; TFPα, trifunctional protein alpha; WAT, white adipose tissue.
Foods 11 02863 g003
Figure 4. The main effects and mechanisms of SCFAs on inflammatory bowel diseases. Abbreviation: CEBPB, CCAAT enhancer-binding protein beta; DUSP6, dual-specificity phosphatase 6; ERK, extracellular signal-regulated kinase; GRP, G-protein receptor; HDAC, histone deacetylase; HSF, heat shock factor; HSP, heat shock proteins; IL, interleukin; JNK, c-Jun N-terminal kinase; MAPK, p38 mitogen-activated protein kinase; MCT-1, monocarboxylate transporter 1; miR-145, microRNA-145; mTOR, mechanistic target of rapamycin; NF-κB, nuclear factor-kappa B; SCFAs, short-chain fatty acid; Syk, spleen tyrosine kinase; TGF-β, transforming growth factor-β.
Figure 4. The main effects and mechanisms of SCFAs on inflammatory bowel diseases. Abbreviation: CEBPB, CCAAT enhancer-binding protein beta; DUSP6, dual-specificity phosphatase 6; ERK, extracellular signal-regulated kinase; GRP, G-protein receptor; HDAC, histone deacetylase; HSF, heat shock factor; HSP, heat shock proteins; IL, interleukin; JNK, c-Jun N-terminal kinase; MAPK, p38 mitogen-activated protein kinase; MCT-1, monocarboxylate transporter 1; miR-145, microRNA-145; mTOR, mechanistic target of rapamycin; NF-κB, nuclear factor-kappa B; SCFAs, short-chain fatty acid; Syk, spleen tyrosine kinase; TGF-β, transforming growth factor-β.
Foods 11 02863 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xiong, R.-G.; Zhou, D.-D.; Wu, S.-X.; Huang, S.-Y.; Saimaiti, A.; Yang, Z.-J.; Shang, A.; Zhao, C.-N.; Gan, R.-Y.; Li, H.-B. Health Benefits and Side Effects of Short-Chain Fatty Acids. Foods 2022, 11, 2863. https://doi.org/10.3390/foods11182863

AMA Style

Xiong R-G, Zhou D-D, Wu S-X, Huang S-Y, Saimaiti A, Yang Z-J, Shang A, Zhao C-N, Gan R-Y, Li H-B. Health Benefits and Side Effects of Short-Chain Fatty Acids. Foods. 2022; 11(18):2863. https://doi.org/10.3390/foods11182863

Chicago/Turabian Style

Xiong, Ruo-Gu, Dan-Dan Zhou, Si-Xia Wu, Si-Yu Huang, Adila Saimaiti, Zhi-Jun Yang, Ao Shang, Cai-Ning Zhao, Ren-You Gan, and Hua-Bin Li. 2022. "Health Benefits and Side Effects of Short-Chain Fatty Acids" Foods 11, no. 18: 2863. https://doi.org/10.3390/foods11182863

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

Article Metrics

Back to TopTop