Next Article in Journal
Effect of Potassium Deficiency on Physiological Responses and Anatomical Structure of Basil, Ocimum basilicum L.
Previous Article in Journal
Temporary Survival Increasing the Diversity of Culturable Heterotrophic Bacteria in the Newly Exposed Moraine at a Glacier Snout
Previous Article in Special Issue
Adipokines in Non-Alcoholic Fatty Liver Disease: Are We on the Road toward New Biomarkers and Therapeutic Targets?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diet-Related Changes of Short-Chain Fatty Acids in Blood and Feces in Obesity and Metabolic Syndrome

Department of Molecular Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
Biology 2022, 11(11), 1556; https://doi.org/10.3390/biology11111556
Submission received: 29 September 2022 / Revised: 17 October 2022 / Accepted: 20 October 2022 / Published: 24 October 2022

Abstract

:

Simple Summary

Short-chain fatty acids are produced by the bacteria present in the large intestine. They are digestion products of fiber-containing foods and have many effects. Short-chain fatty acids appear to decrease weight gain and improve diseases related to obesity, with diet being the most important factor that modifies short-chain fatty acid levels in the body. The purpose of this article is to provide an overview and analysis of the variations of short-chain fatty acids in blood and stool in obesity-related conditions, thus helping to monitor these diseases.

Abstract

Obesity-related illnesses are one of the leading causes of death worldwide. Metabolic syndrome has been associated with numerous health issues. Short-chain fatty acids (SCFAs) have been shown to have multiple effects throughout the body, both directly as well as through specific G protein-coupled receptors. The main SCFAs produced by the gut microbiota are acetate, propionate, and butyrate, which are absorbed in varying degrees from the large intestine, with some acting mainly locally and others systemically. Diet has the potential to influence the gut microbial composition, as well as the type and amount of SCFAs produced. High fiber-containing foods and supplements increase the production of SCFAs and SCFA-producing bacteria in the gut and have been shown to have bodyweight-lowering effects. Dietary supplements, which increase SCFA production, could open the way for novel approaches to weight loss interventions. The aim of this review is to analyze the variations of fecal and blood SCFAs in obesity and metabolic syndrome through a systematic search and analysis of existing literature.

1. Introduction

1.1. Short-Chain Fatty Acids

Short-chain fatty acids (SCFAs) are carboxylic acids with less than five carbon atoms [1]. These include methanolate, acetate (Ac), propionate (Prop), butyrate (Bu), and valerate; however, the three main SCFAs that have been extensively studied are Ac, Prop, and Bu. SCFAs are produced by the microbial fermentation of various dietary compounds, mainly fibers [2,3,4]. Ac, Prop, and Bu represent the majority of SCFAs produced in the colon [5,6].
Acetate is produced by metabolizing ethyl alcohol, by ingesting vinegar directly, or by the fermentation of dietary fiber [2,7]. The main area of synthesis is the cecum, followed by the ascending colon, with minimal production taking place in the distal colon [8]. Acetate is able to pass the blood–brain barrier and is found in varying concentrations in almost all tissues and liquids in the body [9,10]. Due to the fact that Ac is able to cross the blood–brain barrier, it has been shown to have a direct appetite-suppressing role in the central nervous system, more specifically in the hypothalamus [10]. Acetate also appears to be a ligand for the G protein-coupled receptors (GPR) 41 and GPR 43 [11,12].
Propionate is the second most abundant SCFA in the portal circulation after Ac [8]. Similarly to Ac, Prop has been shown to influence hunger and satiety; however, the exact mechanism has not yet been clarified [13]. It has been shown that Prop is the main ligand for GPR 41 and GPR 43, and in mice, Prop increases leptin (the main hormone involved in hunger and satiety regulation) levels, leading to a decrease in energy intake [14].
Butyrate is absorbed the least from the intestine, despite being produced in significant quantities. It acts mainly locally and is a main energy substrate for colonocytes [5,15]. Butyrate has also been shown, along with Prop, to regulate the division and proliferation of the large intestinal mucosa, with a possible role in preventing colon cancer [16,17], as well as other types of cancers [18].
GPR 41 and GPR 43, also known as free fatty acid receptors (FFAR) 3 and 2, respectively, have been shown to be present in significant quantities in adipocytes and colonocytes [19]. GPR 41 and 43 are G protein-coupled receptors, which were discovered earlier than their ligands, being initially called orphan G protein-coupled receptors, until Prop was demonstrated to be a ligand by Brown et al. [20] in 2003.
Figure 1 summarizes the key effects of the three main SCFAs (Ac, Prop, and Bu).

1.2. Obesity and Metabolic Syndrome

Metabolic syndrome is usually defined as a combination of hypertension, dyslipidemia, diabetes, and obesity [21]. Obesity in and of itself does not equal metabolic syndrome, but it is possible to have obesity without hypertension and diabetes. Metabolic syndrome, by increasing atherosclerosis and insulin resistance, increases the risk for stroke and myocardial infarction [22,23,24]. Increased blood glucose levels, independently of diabetes, were also found to be a risk factor for physical impairment [25].
Obesity is a major public health issue, especially in developed countries [26]. Increased body weight and obesity have been associated with multiple diseases that have a significant impact on quality of life and life expectancy [27]. The main causes of death globally are cardiovascular complications (such as myocardial infarction and stroke), as well as cancers of different origins, both being strongly associated with obesity [28,29].
Increased body weight is caused by two processes: excess energy intake and insufficient energy expenditure (due to decreased exercise). Increasing energy expenditure through exercise is the basis of obesity treatment and also of numerous chronic diseases, such as hypertension [30]. Only increasing exercise is not enough for an efficient weight loss, however, so a reduction in energy intake is also needed. There have been numerous diets developed along the years, but compliance to reducing the quantities of food is the most important factor in a successful reduction of energy intake [31].
Diet and exercise remain the predominant cornerstones in preventing and managing obesity and metabolic syndrome, as well as their inherent complications. The main source of SCFAs in the body is a result of microbial activity in the gut, especially the digestion of dietary fibers [2,3]. It is only natural that variations in diet will also influence SCFA production.
Previous reviews published about the effects of SCFAs on energy metabolism have either focused solely on diet [32] or on specific diseases related to obesity and metabolic syndrome [33,34,35]. The aim of this review is to provide an overview and analysis of the variations of SCFAs in feces and blood in obesity and metabolic syndrome and to identify novel ways to approach the monitoring of these diseases. The majority of studies analyzing SCFAs in obesity and metabolic syndrome included in this review were focused on diet and its influence on fecal and blood changes of SCFAs. To date, there have been no reviews to provide a combined and comprehensive overview of blood and fecal SCFA variations in both obesity and metabolic syndrome.

2. Methodology

2.1. Search Strategy

Databases accessible through the National Library of Medicine’s PubMed search engine were scrutinized for this review. Human, animal, and in vitro research papers were all selected. The search terms were constructed using the Medical Subject Headings (MeSH) function of PubMed. The following search terms were jointly selected by the authors: “(“Fatty Acids, Volatile”[Mesh]) AND “Obesity”[Mesh]” and “(“Fatty Acids, Volatile”[Mesh]) AND “Metabolic Syndrome”[Mesh]”. For the first search term, the search was carried out on the 30 June 2022 and, due to the large number of studies, was limited to the last 5 years, while for the second search term, the search was carried out on the 30 April 2022 and was limited to the last 10 years. This strategy yielded 291 and 86 results, respectively, for a total of 377 articles. Lists were made for each set of results which were cross-checked, and duplicates were eliminated. After the initial screening phase, based on the abstracts for each study, full-text articles were obtained, which were evaluated according to the inclusion and exclusion criteria.

2.2. Selection, Screening and Inclusion

The inclusion and exclusion criteria were agreed upon jointly by the authors. Only papers with abstracts were included in the initial screening phase. Human, animal, and in vitro studies were considered, and only articles written in English were included. Studies analyzing the effects of diet and SCFA supplementation were included.
Studies that did not measure total SCFA, Ac, Prop, or Bu levels and studies that lacked a clear description of materials and methods were excluded. Case reports and reviews were excluded as well.
The study identification, selection, and inclusion process are summarized in Figure 2. After eliminating duplicates, in total, 139 articles were included in this review, 112 from the first search term containing obesity [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147] and 27 from the second search term containing metabolic syndrome [148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174]. Of the total number of included articles, the majority of studies (n = 62) analyzed various parameters on high-fat-diet (HFD)-fed mice and rats.

3. Short-Chain Fatty Acids between Diet and Metabolism

3.1. Animal Studies

3.1.1. Diet

Mice with metabolic syndrome have been shown to have lower levels of fecal short-chain fatty acids (fSCFA) [159], and obesity also reduced fecal butyrate (fBu) levels [104]. An HFD is a commonly used method of inducing diet-related obesity in animal test subjects. Mice on HFD were shown to have lower fecal acetate (fAc) [141] and blood acetate (bAc) [49] levels. There are even studies that have shown higher fSCFA levels in HFD-fed mice; this was probably a result of an increase in fiber intake and not a result of the HFD per se [89].
A Western diet also produced a reduction in fSCFA levels; however, this was ameliorated by subsequent exercise [41]. Exercise also increased fAC levels in the case of HFD-fed mice [106].
While there seems to be a connection between SCFA production and exercise levels, the exact nature of this interaction remains unknown. Until further studies analyze the effects of exercise on SCFA production, diet seems to be the main contributing factor to fecal and blood SCFA concentrations. Because HFD was used to induce obesity, the effects of various dietary supplements could be studied on an obese animal model. The effects of dietary supplements on SCFA levels of HFD-fed mice and rats are presented in Table 1.
In addition to HFD-fed mice, in the case of HFD-fed Göttingen Minipigs, whey increased fSCFA and fecal propionate (fProp) levels, and wheat bran increased fBu while decreasing fSCFA and fAc levels [80]. A less-used non-diet-induced obese mouse model is the ob/ob mutant (leptin deficient mice). Similarly to HFD mice, inulin (a dietary fiber from the fructans class) increased fSCFA in ob/ob mice as well [44].
In metabolic syndrome mice, caffeine and chlorogenic acid increased bAc, bProp, and bBu levels [156], while in the case of db/db mice (a type II diabetes and obese mutant mouse model), rutin (a citrus flavonoid) administration increased fSCFA [37]. Similarly, in monosodium glutamate (MSG)-induced obesity, fructo-oligosaccharides increased fAc, fProp, fBu, and bProp concentrations [99].
There was a clear trend in dietary supplements, especially fibers, increasing fecal and blood SCFA levels that were decreased as a result of HFD [56,60,65,66,68,70,72,73,77,81,85,86,103,105,110,111,112,113,114,118,120,122,124,126,130,136,137,138,139,144,158,160,162,167,169].
While an HFD-induced obese model can be used to analyze the effects of dietary supplements on obesity, the effects of dietary supplements are not exclusive to diet-induced obese animal models. Mice fed with a high-fructose diet showed an increase in fSCFA concentrations [161], and inulin-fed mice had increased fBu levels [39].
Even though these studies were carried out on animals that have not been on HFD, due to the close relationship between diet and obesity, the results showed a similar trend of increased SCFA levels after administration, especially fiber-containing dietary supplements [39,44]. This suggests that fiber-containing dietary supplements not only normalize and otherwise alter the SCFA profile due to HFD, but fibers also have the ability to increase SCFA production, regardless of the presence or absence of diet-induced obesity.
Another way of indirectly altering diet, especially in the case of obese subjects, is weight-reducing surgery, which produces a global decrease in food intake. Hypo-absorptive bariatric surgeries in obese rats resulted in higher levels of fProp and fBu, compared to sham surgery [46]. However, there are also conflicting results that show a reduction in fSCFA and blood short-chain fatty acid (bSCFA) levels in obese rats with Roux-en-Y gastric bypass, compared to sham surgery [74]. From the studies carried out so far, a global reduction on energy intake does not seem to have a clear effect on fecal and blood SCFA levels.
Prebiotics are nutrients that increase the concentration of certain bacteria in the gut that are considered beneficial to the host’s metabolism and health [175]. Prebiotics from acorn and sago combined with inulin increased fSCFA levels in HFD mice [119]. Commercial inulin-type fructans prebiotic administration in overweight beagles increased fSCFA levels as well [132]. As expected, prebiotics, especially ones that were fiber-related or that were co-administered with inulin, increased fSCFA levels [119,132].

3.1.2. SCFA Supplementation

Short-chain fatty acids themselves can also be directly utilized as dietary supplements. Consequently, Table 2 contains the effects of SCFA administration on HFD-fed mice and rats. Furthermore, in the case of Western diet-fed mice, co-administration of inulin and Bu was shown to attenuate steatohepatitis [58].
There are studies conducted on non-diet-induced obese mice, in which Bu supplementation reduced body weight increase and reduced food intake [102]. Butyrate supplementation also reduced body weight gain and increased insulin receptor expression in obese Apo E knock-out mice [140]. Toll-like receptor 5 knock-out mice administered with Ac, Prop, and Bu had reduced food intake but increased total serum cholesterol and triglycerides [166].
Table 2. Effects of SCFA supplementation on HFD-fed mice and rats.
Table 2. Effects of SCFA supplementation on HFD-fed mice and rats.
AcetatePropionateButyrate
Ameliorated obesity [88]Reduced body weight and fasting insulin levels [96]Had no significant effect [82]
Normalized weight gain, insulin, TNF-α and leptin levels [48]Increased adiponectin expression [135]Altered gut microbiota to be similar to LFD [117]
Increased adiponectin expression [135]Prevented weight gain [145]Ameliorated obesity, steatohepatitis [38]
Prevented weight gain [145] Reduced body weight gain, improved insulin response [92]
Decreased leptin and insulin levels [146]
Reversed HFD induced dysmetabolism [150]
Increased adiponectin expression [135]
Abbreviations: TNF-α, tumor necrosis factor α; LFD, low-fat diet; HFD, high-fat diet.
Short-chain fatty acids consistently normalized or, in some cases, actually prevented HFD-induced alterations, such as weight gain [38,48,88,92,96,145], elevated insulin levels, and decreased leptin levels [48,92,96,146]. Consequently, SCFAs could be used in the future as a supplement to ameliorate weight gain.

3.1.3. Probiotics and Gut Microbiota

Oral intake of Bifidobacterium longum and Lactobacillus plantarum increased fSCFA in obese, menopausal mice [61]. A similar increase in fSCFA levels was achieved as a result of gut microbiota transplantation from normal to leptin-deficient mice [108]. Contrary to the previous two studies on gut microbiota, antibiotic-induced microbial depletion significantly decreased fSCFA in otherwise healthy mice [133].
Considering the studies that involved probiotics, the majority focused on HFD-fed mice and rats. Thereby, Table 3 presents the effects of probiotics found in these studies.
Even though Bifidobacterium and Clostridium species are considered SCFA-producing bacteria [176], interestingly, their administration has not consistently produced increases in fSCFA [61,69], and in the case of Clostridium, actually decreased fSCFA [40]. Due to the complex and ever-changing nature of the gut microbiota and the numerous factors that influence its composition, it is difficult to correctly assess the ideal gut composition in any given host. The increase in one species, even if it is an SCFA-producing species, can lead to the decrease in other species at the same time, with global SCFA production suffering an imbalance as a result.

3.2. In-Vitro Studies

Using a colon model, it was shown that inulin and resistant starch increased Ac and Bu, but only in the lean model [45]. Similarly, prebiotic bread containing galacto-oligosaccharides increased Ac and Bu [168], and pumpkin skin fermentation produced high amounts of SCFAs [52]. Rice bran arabinoxylan increased total SCFA production in both normal weight and obese human gut microbiota [57]. Obese human gut microbiomes had higher production of Ac and Bu but lower production of Prop, compared to normal weight microbiomes; however, the total SCFA level was still higher in the obese microbiota [55]. In the case of gut microbiota from obese children, commercial prebiotics increased SCFA production; however, the magnitude of this increase was donor specific [94].
In an in vitro digestion model, propolis increased SCFA concentrations [42]. Bianchi et al. [131] showed, using the Simulator of the Human Intestinal Microbial Ecosystem, that citric pectin also increased Ac and Bu.
In precision-cut liver slices from metabolic-associated fatty liver disease from male mice, Bu treatment reduced fibrotic response but decreased fat oxidation [148]. Butyrate enhanced the immunomodulating effect of adipose-derived stem cells from obese and diabetic mice [67].
In vitro studies have the advantage of being able to precisely control all aspects of a given model, compared to animal subjects. The main drawback, however, is the fact that, by controlling all parameters perfectly, in vitro models might not be representative of the real-world conditions. Even though it is difficult to simulate obesity and metabolic syndrome in vitro, studies that assess the effects of nutrients and SCFAs on the gut microbiome still provide valuable insights into the function of this complex symbiosis.

3.3. Human Studies

Levels of fBu and fProp were found to increase progressively with body weight [84], and fSCFA was positively associated with the prevalence of obesity in a Japanese population [59]. Normal-weight individuals also had lower levels of fBu [71], and weight loss interventions lowered fSCFA levels [97]. Non-obese, non-alcoholic steatohepatitis patients with significant fibrosis had elevated fProp, compared to controls [90].
Obesity in pregnancy was associated with a reduction in fSCFA levels [62], while preeclampsia women had lower blood butyrate (bBu) and Bu-producing bacteria [75]. Exclusively breast-feeding mothers had higher milk Bu levels than mixed-feeding mothers, which was negatively associated with changes in infant weight [115].
In children, fAc, fProp, and fBu levels were positively correlated with BMI [51,78]. There were, however, conflicting results as well, which showed that obese children had lower fSCFA levels, compared to normal-weight children [128]. In obese adolescent girls, waist circumference was associated with fSCFA levels [101], while in obese adolescents, Lactobacillus salivarius administration did not alter fSCFA levels [172]. Post-bariatric surgery fAc, fProp, and fBu were significantly reduced and were positively correlated with BMI in obese adults [50].
Men with metabolic syndrome had significantly higher levels of bProp and bBu and lower levels of bAc, compared to healthy, lean men [157], and obese women had significantly higher fAc, fProp, and fBu compared to lean women [173]. Higher fProp was corelated with serum triglycerides, and fBu levels were also associated with metabolic syndrome [149].
Exercise increased fSCFA in lean but not obese subjects [142], behavioral weight loss and metformin treatment increased bAc, and metformin also increased bBu [64].
Butyrate administration reduced inflammation [121] and brain dopamine binding [151]. Levels of bBu were negatively corelated with distal neuropathic pain [91].
The abundance of gut Firmicutes bacteria belonging to the Ruminococcaceae family was positively associated with bAc levels [143], while the levels of Bacteroidetes, such as Allistipes and Bacteroides, were correlated with fProp levels [170]. The concentration of fSCFA was inversely associated with microbiome diversity [123]. Recipients of the metabolic syndrome donor fecal transplant had higher fProp and fBu but not fAc levels [154].

Dietary Supplementation

The effects of dietary supplements and various diets on SCFA levels in humans are summarized in Table 4.
Due to different dietary habits, a pertinent finding was that Ghanaians consumed more fiber and had higher fSCFA levels, compared to United States residents [127]. Levels of fBu were positively associated with starch intake and negatively with non-nutritive sweetener intake [107].
There are significantly fewer human studies compared to animal studies; however, the results of these generally coincide with the results of animal studies. (Fiber-rich diets increase fecal and blood SCAFs.) This suggests that the animal models (most frequently mice and rats) used to analyze the effects of diet on blood and fecal SCFA levels provide valuable information regarding the changes in SCFAs produced by diet.

3.4. Diet and SCFAs

Diet, unequivocally, has a significant influence on both fecal and blood levels of SCFAs, as well as the relative abundance of Ac, Prop, and Bu [36,37,39,41,42,43,44,45,47,52,54,56,57,60,63,65,66,68,70,72,73,76,77,80,81,83,85,86,89,93,95,98,99,100,103,105,110,111,112,113,114,118,120,122,124,125,126,129,130,131,134,136,137,138,139,144,147,149,152,153,155,156,158,160,161,162,163,164,165,167,168,169,171,174]. Diet can alter the SCFA profile both directly, through supplying a substrate for SCFA-producing bacteria, as well as indirectly, by altering the gut microbiome’s composition. HFD reduced SCFA concentrations by varying amounts in multiple studies [49,104,141], as did a Western-type diet [41]. Obesity and metabolic syndrome reduced SCFA levels as well [49,62,104,128,154,159]; however, there is not a clear trend, and obesity has been shown to increase SCFA concentrations in some populations [51,59,71,78,84,101]. Because the main result of HFD consumption is diet-induced obesity, it is not exactly clear if HFD reduced SCFA levels directly or if these alterations are actually due to the resulting obesity.
In terms of diet, supplementation with dietary fiber and/or high-fiber foods had the most profound effect on both fSCFA and bSCFA profiles, generally increasing SCFA concentrations [36,39,44,45,52,54,56,60,70,72,77,80,85,86,93,98,103,105,111,122,125,126,129,130,131,134,137,153,155,158,163,164,165,167,169,171]. This, however, should not come as a surprise, given the fact that the main source of SCFAs is the bacterial fermentation of fibers and resistant sugars [176].
It is of note, however, that there are multiple articles demonstrating that fats increased SCFA production as well [124,174]. This suggests that there might be an alternate mechanism present in the gut microbiota that enables the production of SCFAs from sources other than indigestible fibers.
The effects of fiber supplementation have been extensively studied, but studies on other types of dietary supplements (mainly proteins and lipids) are lacking. Even though it is clear that fibers increase the production of SCFAs, there are multiple studies that have shown this effect in the case of other types of dietary supplements as well [118,124,174]. Further studies should focus on evaluating the effects of other supplements in modifying SCFA production.
Direct supplementation of SCFAs, as well as increases resulting from diet, produces a host of beneficial effects from weight loss to normalization of insulin levels and amelioration of hepatic steatosis (Figure 3).

3.5. Gut Microbiota and SCFAs

Even though the main focus of this review was not the influence of probiotics on the gut microbiota composition and functionality, there are studies that have also analyzed this perspective.
There are many microbes present in the human gut that have been shown to produce SCFAs [176]. Supplementation with probiotics containing such microbes has not produced a consistent increase in SCFA production, in some cases, had the exact opposite effect of reducing fSCFA levels [40,69]. Thus, a study demonstrated that the concentration of fSCFAs was inversely associated with microbiome diversity [123].
This would suggest two possibilities. The first one is that the substrate supplied to the gut microbes actually has a more important role in determining the production and relative abundance of SCFAs. This hypothesis is supported by the fact that, in some conditions, known SCFA-producing bacteria increased fSCFA levels as expected [53,79,87,116], while in others, it actually decreased SCFA production [40,69]. The key to influencing SCFA production might not be artificially increasing the abundance of a certain species of bacteria through probiotics but of increasing the available substrate. This would create an environmental selection pressure that would favor multiple SCFA-producing bacteria on one hand, and on the other hand, increase the production of SCFAs by default, due to a higher substrate supply.
The other possibility is exactly the opposite of the first one, which is that there is actually one or just a small number of species of SCFA-producing bacteria that are actually responsible for the majority of the produced SCFAs. This hypothesis is somewhat supported by the fact that fSCFA levels were inversely associated with gut microbiota diversity [123]. Research that would prove or disprove this hypothesis would need to analyze the effects of various probiotic supplements in modifying the SCFA profile under the same conditions and preferably in the same individuals, both in animals and humans.

4. Conclusions

This is the first review to encompass studies related to diet that analyze both fecal and blood SCFA levels in obesity and metabolic syndrome. The effects of the gut microbiota, while being widespread, are sparsely understood. SCFAs are only a small part of the wide range of metabolites produced by gut bacteria, which have a profound effect on their host’s metabolism and homeostasis.
It is clear that both the gut microbiota functionality, as well as its composition, are closely related to diet, and that dietary fibers increase the production of SCFAs. The exact mechanism by which gut microbiota composition affects this relationship remains unclear.
While it has been shown that increasing fecal and blood SCFA levels lead to numerous beneficial results, especially in the case of obesity, the mechanism by which SCFAs produce these changes is a matter of debate. Short-chain fatty acids, mainly Prop, are known ligands of GPR 41 and GRP 43, through which they have been shown to increase leptin expression. Leptin is responsible for decreasing energy intake through its action on the hunger centers of the central nervous system. This is supported by multiple studies demonstrating that increased SCFA levels decreases weight gain; however, this is only one hypothesis for the mechanism of action of SCFAs. This review envelops the latest research caried out on the effect of diet and dietary supplements on the fecal and blood levels of SCFAs, as well as an overview of the SCFA-mediated effects of the gut microbiota. Diet influences the host microbiome and has a profound effect on energy metabolism regulation. Uncovering the exact nature of this host–microbiota interplay could yield novel ways to approach the monitoring of energy metabolism and treatment of obesity-related conditions, as well as establishing an optimal diet to aid in the treatment of obesity.

Author Contributions

Conceptualization, T.I., C.N.S. and A.M.C.; methodology, T.I. and C.N.S.; resources, A.M.C.; writing—original draft preparation, T.I.; writing—review and editing, T.I., C.N.S. and A.M.C.; supervision, C.N.S. and A.M.C.; project administration, A.M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the Ministry of Research, Innovation, and Digitization, CNCS/CCCDI—UEFISCDI, project number 2/2019 (DARKFOOD), within PNCDI III.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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] [PubMed] [Green Version]
  2. Wong, J.M.W.; de Souza, R.; Kendall, C.W.C.; Emam, A.; Jenkins, D.J.A. Colonic Health: Fermentation and Short Chain Fatty Acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef] [PubMed]
  3. 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] [Green Version]
  4. Rekha, K.; Venkidasamy, B.; Samynathan, R.; Nagella, P.; Rebezov, M.; Khayrullin, M.; Ponomarev, E.; Bouyahya, A.; Sarkar, T.; Shariati, M.A.; et al. Short-Chain Fatty Acid: An Updated Review on Signaling, Metabolism, and Therapeutic Effects. Crit. Rev. Food Sci. Nutr. 2022, 1–29. [Google Scholar] [CrossRef] [PubMed]
  5. Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-Chain Fatty Acids in Control of Body Weight and Insulin Sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577–591. [Google Scholar] [CrossRef]
  6. Topping, D.L.; Clifton, P.M. Short-Chain Fatty Acids and Human Colonic Function: Roles of Resistant Starch and Nonstarch Polysaccharides. Physiol. Rev. 2001, 81, 1031–1064. [Google Scholar] [CrossRef] [Green Version]
  7. Mortensen, P.B.; Holtug, K.; Bonnén, H.; Clausen, M.R. The Degradation of Amino Acids, Proteins, and Blood to Short-Chain Fatty Acids in Colon Is Prevented by Lactulose. Gastroenterology 1990, 98, 353–360. [Google Scholar] [CrossRef]
  8. Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short Chain Fatty Acids in Human Large Intestine, Portal, Hepatic and Venous Blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [Green Version]
  9. Jiang, L.; Gulanski, B.I.; de Feyter, H.M.; Weinzimer, S.A.; Pittman, B.; Guidone, E.; Koretski, J.; Harman, S.; Petrakis, I.L.; Krystal, J.H.; et al. Increased Brain Uptake and Oxidation of Acetate in Heavy Drinkers. J. Clin. Invest. 2013, 123, 1605–1614. [Google Scholar] [CrossRef] [Green Version]
  10. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The Short-Chain Fatty Acid Acetate Reduces Appetite via a Central Homeostatic Mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef]
  11. Karaki, S.-I.; Tazoe, H.; Hayashi, H.; Kashiwabara, H.; Tooyama, K.; Suzuki, Y.; Kuwahara, A. Expression of the Short-Chain Fatty Acid Receptor, GPR43, in the Human Colon. J. Mol. Histol. 2008, 39, 135–142. [Google Scholar] [CrossRef]
  12. Tazoe, H.; Otomo, Y.; Karaki, S.-I.; Kato, I.; Fukami, Y.; Terasaki, M.; Kuwahara, A. Expression of Short-Chain Fatty Acid Receptor GPR41 in the Human Colon. Biomed. Res. 2009, 30, 149–156. [Google Scholar] [CrossRef] [Green Version]
  13. Hosseini, E.; Grootaert, C.; Verstraete, W.; van de Wiele, T. Propionate as a Health-Promoting Microbial Metabolite in the Human Gut. Nutr. Rev. 2011, 69, 245–258. [Google Scholar] [CrossRef]
  14. Xiong, Y.; Miyamoto, N.; Shibata, K.; Valasek, M.A.; Motoike, T.; Kedzierski, R.M.; Yanagisawa, M. Short-Chain Fatty Acids Stimulate Leptin Production in Adipocytes through the G Protein-Coupled Receptor GPR41. Proc. Natl. Acad. Sci. USA 2004, 101, 1045–1050. [Google Scholar] [CrossRef] [Green Version]
  15. Milovic, V.; Teller, I.C.; Turchanowa, L.; Caspary, W.F.; Stein, J. Effect of Structural Analogues of Propionate and Butyrate on Colon Cancer Cell Growth. Int. J. Colorectal Dis. 2000, 15, 264–270. [Google Scholar] [CrossRef]
  16. Jan, G.; Belzacq, A.-S.; Haouzi, D.; Rouault, A.; Métivier, D.; Kroemer, G.; Brenner, C. Propionibacteria Induce Apoptosis of Colorectal Carcinoma Cells via Short-Chain Fatty Acids Acting on Mitochondria. Cell Death Differ. 2002, 9, 179–188. [Google Scholar] [CrossRef]
  17. Scheppach, W. Effects of Short Chain Fatty Acids on Gut Morphology and Function. Gut 1994, 35, S35–S38. [Google Scholar] [CrossRef] [Green Version]
  18. 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, 1–18. [Google Scholar] [CrossRef] [PubMed]
  19. González Hernández, M.A.; 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] [Green Version]
  20. Brown, A.J.; Goldsworthy, S.M.; Barnes, A.A.; Eilert, M.M.; Tcheang, L.; Daniels, D.; Muir, A.I.; Wigglesworth, M.J.; Kinghorn, I.; Fraser, N.J.; et al. The Orphan G Protein-Coupled Receptors GPR41 and GPR43 Are Activated by Propionate and Other Short Chain Carboxylic Acids. J. Biol. Chem. 2003, 278, 11312–11319. [Google Scholar] [CrossRef]
  21. Vega, G.L. Obesity and the Metabolic Syndrome. Minerva Endocrinol. 2004, 29, 47–54. [Google Scholar] [PubMed]
  22. Clavijo, L.C.; Pinto, T.L.; Kuchulakanti, P.K.; Torguson, R.; Chu, W.W.; Satler, L.F.; Kent, K.M.; Suddath, W.O.; Pichard, A.D.; Waksman, R. Metabolic Syndrome in Patients with Acute Myocardial Infarction Is Associated with Increased Infarct Size and In-Hospital Complications. Cardiovasc. Revasc. Med. 2006, 7, 7–11. [Google Scholar] [CrossRef] [PubMed]
  23. Brown, W.V. Metabolic Syndrome and Risk of Stroke. Clin. Cornerstone 2004, 6 (Suppl. 3), S30–S34. [Google Scholar] [CrossRef]
  24. Mone, P.; Morgante, M.; Pansini, A.; Jankauskas, S.S.; Rizzo, M.; Lombardi, A.; Frullone, S.; Santulli, G. Effects of Insulin Resistance on Mitochondrial (Dys)Function. Atherosclerosis 2022, 341, 52–54. [Google Scholar] [CrossRef]
  25. Pansini, A.; Lombardi, A.; Morgante, M.; Frullone, S.; Marro, A.; Rizzo, M.; Martinelli, G.; Boccalone, E.; de Luca, A.; Santulli, G.; et al. Hyperglycemia and Physical Impairment in Frail Hypertensive Older Adults. Front. Endocrinol. 2022, 13, 831556. [Google Scholar] [CrossRef]
  26. Visscher, T.L.; Seidell, J.C. The Public Health Impact of Obesity. Annu. Rev. Public Health 2001, 22, 355–375. [Google Scholar] [CrossRef] [Green Version]
  27. Peeters, A.; Barendregt, J.J.; Willekens, F.; Mackenbach, J.P.; Al Mamun, A.; Bonneux, L. NEDCOM, The Netherlands Epidemiology and Demography Compression of Morbidity Research Group Obesity in Adulthood and Its Consequences for Life Expectancy: A Life-Table Analysis. Ann. Intern. Med. 2003, 138, 24–32. [Google Scholar] [CrossRef]
  28. Suwaidi, J.A.; Wright, R.S.; Grill, J.P.; Hensrud, D.D.; Murphy, J.G.; Squires, R.W.; Kopecky, S.L. Obesity Is Associated with Premature Occurrence of Acute Myocardial Infarction. Clin. Cardiol. 2001, 24, 542–547. [Google Scholar] [CrossRef]
  29. De Pergola, G.; Silvestris, F. Obesity as a Major Risk Factor for Cancer. J. Obes. 2013, 2013, 291546. [Google Scholar] [CrossRef] [Green Version]
  30. Alpsoy, Ş. Exercise and Hypertension. Adv. Exp. Med. Biol. 2020, 1228, 153–167. [Google Scholar] [CrossRef]
  31. Alhassan, S.; Kim, S.; Bersamin, A.; King, A.C.; Gardner, C.D. Dietary Adherence and Weight Loss Success among Overweight Women: Results from the A TO Z Weight Loss Study. Int J. Obes. 2008, 32, 985–991. [Google Scholar] [CrossRef] [Green Version]
  32. Bliesner, A.; Eccles-Smith, J.; Bates, C.; Hayes, O.; Ho, J.Y.; Martins, C.; Truby, H.; Nitert, M.D. Impact of Food-Based Weight Loss Interventions on Gut Microbiome in Individuals with Obesity: A Systematic Review. Nutrients 2022, 14, 1953. [Google Scholar] [CrossRef]
  33. Zaky, A.; Glastras, S.J.; Wong, M.Y.W.; Pollock, C.A.; Saad, S. The Role of the Gut Microbiome in Diabetes and Obesity-Related Kidney Disease. Int. J. Mol. Sci. 2021, 22, 9641. [Google Scholar] [CrossRef]
  34. Nogal, A.; Valdes, A.M.; Menni, C. The Role of Short-Chain Fatty Acids in the Interplay between Gut Microbiota and Diet in Cardio-Metabolic Health. Gut Microbes 2021, 13, 1–24. [Google Scholar] [CrossRef]
  35. Magliocca, G.; Mone, P.; Di Iorio, B.R.; Heidland, A.; Marzocco, S. Short-Chain Fatty Acids in Chronic Kidney Disease: Focus on Inflammation and Oxidative Stress Regulation. Int. J. Mol. Sci. 2022, 23, 5354. [Google Scholar] [CrossRef]
  36. Iversen, K.N.; Dicksved, J.; Zoki, C.; Fristedt, R.; Pelve, E.A.; Langton, M.; Landberg, R. The Effects of High Fiber Rye, Compared to Refined Wheat, on Gut Microbiota Composition, Plasma Short Chain Fatty Acids, and Implications for Weight Loss and Metabolic Risk Factors (the RyeWeight Study). Nutrients 2022, 14, 1669. [Google Scholar] [CrossRef]
  37. Cheng, L.; Shi, L.; He, C.; Wang, C.; Lv, Y.; Li, H.; An, Y.; Dai, H.; Duan, Y.; Zhang, H.; et al. Rutin-Activated Adipose Tissue Thermogenesis Is Correlated with Increased Intestinal Short-Chain Fatty Acid Levels. Phytother. Res. 2022, 36, 2495–2510. [Google Scholar] [CrossRef]
  38. Li, H.; Jia, Y.; Weng, D.; Ju, Z.; Zhao, Y.; Liu, S.; Liu, Y.; Song, M.; Cui, L.; Sun, S.; et al. Clostridium Butyricum Inhibits Fat Deposition via Increasing the Frequency of Adipose Tissue-Resident Regulatory T Cells. Mol. Nutr. Food Res. 2022, 66, e2100884. [Google Scholar] [CrossRef]
  39. Muthyala, S.D.V.; Shankar, S.; Klemashevich, C.; Blazier, J.C.; Hillhouse, A.; Wu, C.-S. Differential Effects of the Soluble Fiber Inulin in Reducing Adiposity and Altering Gut Microbiome in Aging Mice. J. Nutr. Biochem. 2022, 105, 108999. [Google Scholar] [CrossRef]
  40. Yang, F.; Zhu, W.; Edirisuriya, P.; Ai, Q.; Nie, K.; Ji, X.; Zhou, K. Characterization of Metabolites and Biomarkers for the Probiotic Effects of Clostridium Cochlearium on High-Fat Diet-Induced Obese C57BL/6 Mice. Eur. J. Nutr. 2022, 61, 2217–2229. [Google Scholar] [CrossRef]
  41. Huang, W.-C.; Tung, C.-L.; Yang, Y.-C.S.H.; Lin, I.-H.; Ng, X.E.; Tung, Y.-T. Endurance Exercise Ameliorates Western Diet-Induced Atherosclerosis through Modulation of Microbiota and Its Metabolites. Sci. Rep. 2022, 12, 3612. [Google Scholar] [CrossRef] [PubMed]
  42. Garzarella, E.U.; Navajas-Porras, B.; Pérez-Burillo, S.; Ullah, H.; Esposito, C.; Santarcangelo, C.; Hinojosa-Nogueira, D.; Pastoriza, S.; Zaccaria, V.; Xiao, J.; et al. Evaluating the Effects of a Standardized Polyphenol Mixture Extracted from Poplar-Type Propolis on Healthy and Diseased Human Gut Microbiota. Biomed. Pharmacother. 2022, 148, 112759. [Google Scholar] [CrossRef]
  43. Łagowska, K.; Drzymała-Czyż, S. A Low Glycemic Index, Energy-Restricted Diet but Not Lactobacillus Rhamnosus Supplementation Changes Fecal Short-Chain Fatty Acid and Serum Lipid Concentrations in Women with Overweight or Obesity and Polycystic Ovary Syndrome. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 917–926. [Google Scholar] [CrossRef] [PubMed]
  44. Guo, J.; Zhang, M.; Wang, H.; Li, N.; Lu, Z.; Li, L.; Hui, S.; Xu, H. Gut Microbiota and Short Chain Fatty Acids Partially Mediate the Beneficial Effects of Inulin on Metabolic Disorders in Obese Ob/Ob Mice. J. Food Biochem. 2022, 46, e14063. [Google Scholar] [CrossRef] [PubMed]
  45. Canfora, E.E.; Hermes, G.D.A.; Müller, M.; Bastings, J.; Vaughan, E.E.; van Den Berg, M.A.; Holst, J.J.; Venema, K.; Zoetendal, E.G.; Blaak, E.E. Fiber Mixture-Specific Effect on Distal Colonic Fermentation and Metabolic Health in Lean but Not in Prediabetic Men. Gut Microbes 2022, 14, 2009297. [Google Scholar] [CrossRef] [PubMed]
  46. Mukorako, P.; Lemoine, N.; Biertho, L.; Lebel, S.; Roy, M.-C.; Plamondon, J.; Tchernof, A.; Varin, T.V.; Anhê, F.F.; St-Pierre, D.H.; et al. Consistent Gut Bacterial and Short-Chain Fatty Acid Signatures in Hypoabsorptive Bariatric Surgeries Correlate with Metabolic Benefits in Rats. Int. J. Obes. 2022, 46, 297–306. [Google Scholar] [CrossRef]
  47. Daskova, N.; Heczkova, M.; Modos, I.; Videnska, P.; Splichalova, P.; Pelantova, H.; Kuzma, M.; Gojda, J.; Cahova, M. Determination of Butyrate Synthesis Capacity in Gut Microbiota: Quantification of but Gene Abundance by QPCR in Fecal Samples. Biomolecules 2021, 11, 1303. [Google Scholar] [CrossRef]
  48. Olaniyi, K.S.; Owolabi, M.N.; Atuma, C.L.; Agunbiade, T.B.; Alabi, B.Y. Acetate Rescues Defective Brain-Adipose Metabolic Network in Obese Wistar Rats by Modulation of Peroxisome Proliferator-Activated Receptor-γ. Sci. Rep. 2021, 11, 18967. [Google Scholar] [CrossRef]
  49. Huang, S.-W.; Ou, Y.-C.; Tang, K.-S.; Yu, H.-R.; Huang, L.-T.; Tain, Y.-L.; Lin, I.-C.; Sheen, J.-M.; Hou, C.-Y.; Tsai, C.-C.; et al. Metformin Ameliorates Maternal High-Fat Diet-Induced Maternal Dysbiosis and Fetal Liver Apoptosis. Lipids Health Dis 2021, 20, 100. [Google Scholar] [CrossRef]
  50. Juárez-Fernández, M.; Román-Sagüillo, S.; Porras, D.; García-Mediavilla, M.V.; Linares, P.; Ballesteros-Pomar, M.D.; Urioste-Fondo, A.; Álvarez-Cuenllas, B.; González-Gallego, J.; Sánchez-Campos, S.; et al. Long-Term Effects of Bariatric Surgery on Gut Microbiota Composition and Faecal Metabolome Related to Obesity Remission. Nutrients 2021, 13, 2519. [Google Scholar] [CrossRef]
  51. Nandy, D.; Craig, S.J.C.; Cai, J.; Tian, Y.; Paul, I.M.; Savage, J.S.; Marini, M.E.; Hohman, E.E.; Reimherr, M.L.; Patterson, A.D.; et al. Metabolomic Profiling of Stool of Two-Year Old Children from the INSIGHT Study Reveals Links between Butyrate and Child Weight Outcomes. Pediatr. Obes. 2022, 17, e12833. [Google Scholar] [CrossRef]
  52. Gual-Grau, A.; Guirro, M.; Crescenti, A.; Boqué, N.; Arola, L. In Vitro Fermentability of a Broad Range of Natural Ingredients by Fecal Microbiota from Lean and Obese Individuals: Potential Health Benefits. Int. J. Food. Sci. Nutr. 2022, 73, 195–209. [Google Scholar] [CrossRef]
  53. Duan, C.; Huang, L.; Zhang, C.; Zhang, L.; Xia, X.; Zhong, Z.; Wang, B.; Wang, Y.; Man Hoi, M.P.; Ding, W.; et al. Gut Commensal-Derived Butyrate Reverses Obesity-Induced Social Deficits and Anxiety-like Behaviors via Regulation of Microglial Homeostasis. Eur. J. Pharmacol. 2021, 908, 174338. [Google Scholar] [CrossRef]
  54. Deroover, L.; Vázquez-Castellanos, J.F.; Vandermeulen, G.; Luypaerts, A.; Raes, J.; Courtin, C.M.; Verbeke, K. Wheat Bran with Reduced Particle Size Increases Serum SCFAs in Obese Subjects without Improving Health Parameters Compared with a Maltodextrin Placebo. Am. J. Clin. Nutr. 2021, 114, 1328–1341. [Google Scholar] [CrossRef]
  55. Martínez-Cuesta, M.C.; Del Campo, R.; Garriga-García, M.; Peláez, C.; Requena, T. Taxonomic Characterization and Short-Chain Fatty Acids Production of the Obese Microbiota. Front. Cell Infect. Microbiol. 2021, 11, 598093. [Google Scholar] [CrossRef]
  56. Xu, C.; Liu, J.; Gao, J.; Wu, X.; Cui, C.; Wei, H.; Peng, J.; Zheng, R. The Effect of Functional Fiber on Microbiota Composition in Different Intestinal Segments of Obese Mice. Int. J. Mol. Sci. 2021, 22, 6525. [Google Scholar] [CrossRef]
  57. Gu, I.; Lam, W.S.; Marasini, D.; Brownmiller, C.; Savary, B.J.; Lee, J.A.; Carbonero, F.; Lee, S.-O. In Vitro Fecal Fermentation Patterns of Arabinoxylan from Rice Bran on Fecal Microbiota from Normal-Weight and Overweight/Obese Subjects. Nutrients 2021, 13, 2052. [Google Scholar] [CrossRef]
  58. Beisner, J.; Filipe Rosa, L.; Kaden-Volynets, V.; Stolzer, I.; Günther, C.; Bischoff, S.C. Prebiotic Inulin and Sodium Butyrate Attenuate Obesity-Induced Intestinal Barrier Dysfunction by Induction of Antimicrobial Peptides. Front. Immunol. 2021, 12, 678360. [Google Scholar] [CrossRef]
  59. Yamamura, R.; Nakamura, K.; Ukawa, S.; Okada, E.; Nakagawa, T.; Imae, A.; Kunihiro, T.; Kimura, T.; Hirata, T.; Tamakoshi, A. Fecal Short-Chain Fatty Acids and Obesity in a Community-Based Japanese Population: The DOSANCO Health Study. Obes. Res. Clin. Pract. 2021, 15, 345–350. [Google Scholar] [CrossRef]
  60. Kawano, R.; Okamura, T.; Hashimoto, Y.; Majima, S.; Senmaru, T.; Ushigome, E.; Asano, M.; Yamazaki, M.; Takakuwa, H.; Sasano, R.; et al. Erythritol Ameliorates Small Intestinal Inflammation Induced by High-Fat Diets and Improves Glucose Tolerance. Int. J. Mol. Sci. 2021, 22, 5558. [Google Scholar] [CrossRef]
  61. Chen, Q.; Wang, B.; Wang, S.; Qian, X.; Li, X.; Zhao, J.; Zhang, H.; Chen, W.; Wang, G. Modulation of the Gut Microbiota Structure with Probiotics and Isoflavone Alleviates Metabolic Disorder in Ovariectomized Mice. Nutrients 2021, 13, 1793. [Google Scholar] [CrossRef]
  62. Ziętek, M.; Celewicz, Z.; Kikut, J.; Szczuko, M. Implications of SCFAs on the Parameters of the Lipid and Hepatic Profile in Pregnant Women. Nutrients 2021, 13, 1749. [Google Scholar] [CrossRef] [PubMed]
  63. Machado, A.M.; da Silva, N.B.M.; de Freitas, R.M.P.; de Freitas, M.B.D.; Chaves, J.B.P.; Oliveira, L.L.; Martino, H.S.D.; de Cássia Gonçalves Alfenas, R. Effects of Yacon Flour Associated with an Energy Restricted Diet on Intestinal Permeability, Fecal Short Chain Fatty Acids, Oxidative Stress and Inflammation Markers Levels in Adults with Obesity or Overweight: A Randomized, Double Blind, Placebo Controlled Clinical Trial. Arch. Endocrinol. Metab. 2021, 64, 597–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mueller, N.T.; Differding, M.K.; Zhang, M.; Maruthur, N.M.; Juraschek, S.P.; Miller, E.R.; Appel, L.J.; Yeh, H.-C. Metformin Affects Gut Microbiome Composition and Function and Circulating Short-Chain Fatty Acids: A Randomized Trial. Diabetes Care 2021, 44, 1462–1471. [Google Scholar] [CrossRef] [PubMed]
  65. Zhu, M.-Z.; Zhou, F.; Ouyang, J.; Wang, Q.-Y.; Li, Y.-L.; Wu, J.-L.; Huang, J.-A.; Liu, Z.-H. Combined Use of Epigallocatechin-3-Gallate (EGCG) and Caffeine in Low Doses Exhibits Marked Anti-Obesity Synergy through Regulation of Gut Microbiota and Bile Acid Metabolism. Food Funct. 2021, 12, 4105–4116. [Google Scholar] [CrossRef]
  66. Zhang, M.; Zhang, X.; Zhu, J.; Zhao, D.-G.; Ma, Y.-Y.; Li, D.; Ho, C.-T.; Huang, Q. Bidirectional Interaction of Nobiletin and Gut Microbiota in Mice Fed with a High-Fat Diet. Food Funct. 2021, 12, 3516–3526. [Google Scholar] [CrossRef]
  67. Hsu, W.-T.; Huang, W.-J.; Chiang, B.-L.; Tseng, P.-H. Butyrate Modulates Adipose-Derived Stem Cells Isolated from Polygenic Obese and Diabetic Mice to Drive Enhanced Immunosuppression. Cytotherapy 2021, 23, 567–581. [Google Scholar] [CrossRef]
  68. Shen, H.; Huang, L.; Dou, H.; Yang, Y.; Wu, H. Effect of Trilobatin from Lithocarpus Polystachyus Rehd on Gut Microbiota of Obese Rats Induced by a High-Fat Diet. Nutrients 2021, 13, 891. [Google Scholar] [CrossRef]
  69. Wang, B.; Kong, Q.; Cui, S.; Li, X.; Gu, Z.; Zhao, J.; Zhang, H.; Chen, W.; Wang, G. Bifidobacterium Adolescentis Isolated from Different Hosts Modifies the Intestinal Microbiota and Displays Differential Metabolic and Immunomodulatory Properties in Mice Fed a High-Fat Diet. Nutrients 2021, 13, 1017. [Google Scholar] [CrossRef]
  70. Mio, K.; Otake, N.; Nakashima, S.; Matsuoka, T.; Aoe, S. Ingestion of High β-Glucan Barley Flour Enhances the Intestinal Immune System of Diet-Induced Obese Mice by Prebiotic Effects. Nutrients 2021, 13, 907. [Google Scholar] [CrossRef]
  71. Iwaki, M.; Kessoku, T.; Ozaki, A.; Kasai, Y.; Kobayashi, T.; Nogami, A.; Honda, Y.; Ogawa, Y.; Imajo, K.; Yoneda, M.; et al. Gut Microbiota Composition Associated with Hepatic Fibrosis in Non-Obese Patients with Non-Alcoholic Fatty Liver Disease. J. Gastroenterol. Hepatol. 2021, 36, 2275–2284. [Google Scholar] [CrossRef]
  72. Suzuki, S.; Aoe, S. High β-Glucan Barley Supplementation Improves Glucose Tolerance by Increasing GLP-1 Secretion in Diet-Induced Obesity Mice. Nutrients 2021, 13, 527. [Google Scholar] [CrossRef]
  73. Du, J.; Zhang, P.; Luo, J.; Shen, L.; Zhang, S.; Gu, H.; He, J.; Wang, L.; Zhao, X.; Gan, M.; et al. Dietary Betaine Prevents Obesity through Gut Microbiota-Drived MicroRNA-378a Family. Gut Microbes 2021, 13, 1862612. [Google Scholar] [CrossRef]
  74. Seyfried, F.; Phetcharaburanin, J.; Glymenaki, M.; Nordbeck, A.; Hankir, M.; Nicholson, J.K.; Holmes, E.; Marchesi, J.R.; Li, J.V. Roux-En-Y Gastric Bypass Surgery in Zucker Rats Induces Bacterial and Systemic Metabolic Changes Independent of Caloric Restriction-Induced Weight Loss. Gut Microbes 2021, 13, 1875108. [Google Scholar] [CrossRef]
  75. Altemani, F.; Barrett, H.L.; Gomez-Arango, L.; Josh, P.; David McIntyre, H.; Callaway, L.K.; Morrison, M.; Tyson, G.W.; Dekker Nitert, M. Pregnant Women Who Develop Preeclampsia Have Lower Abundance of the Butyrate-Producer Coprococcus in Their Gut Microbiota. Pregnancy Hypertens 2021, 23, 211–219. [Google Scholar] [CrossRef]
  76. Choo, J.M.; Tran, C.D.; Luscombe-Marsh, N.D.; Stonehouse, W.; Bowen, J.; Johnson, N.; Thompson, C.H.; Watson, E.-J.; Brinkworth, G.D.; Rogers, G.B. Almond Consumption Affects Fecal Microbiota Composition, Stool PH, and Stool Moisture in Overweight and Obese Adults with Elevated Fasting Blood Glucose: A Randomized Controlled Trial. Nutr. Res. 2021, 85, 47–59. [Google Scholar] [CrossRef]
  77. Ji, Y.; Ma, N.; Zhang, J.; Wang, H.; Tao, T.; Pei, F.; Hu, Q. Dietary Intake of Mixture Coarse Cereals Prevents Obesity by Altering the Gut Microbiota in High-Fat Diet Fed Mice. Food Chem. Toxicol. 2021, 147, 111901. [Google Scholar] [CrossRef]
  78. Wei, Y.; Liang, J.; Su, Y.; Wang, J.; Amakye, W.K.; Pan, J.; Chu, X.; Ma, B.; Song, Y.; Li, Y.; et al. The Associations of the Gut Microbiome Composition and Short-Chain Fatty Acid Concentrations with Body Fat Distribution in Children. Clin. Nutr. 2021, 40, 3379–3390. [Google Scholar] [CrossRef]
  79. Won, S.-M.; Chen, S.; Lee, S.Y.; Lee, K.E.; Park, K.W.; Yoon, J.-H. Lactobacillus Sakei ADM14 Induces Anti-Obesity Effects and Changes in Gut Microbiome in High-Fat Diet-Induced Obese Mice. Nutrients 2020, 12, 3703. [Google Scholar] [CrossRef]
  80. Xu, Y.; Curtasu, M.V.; Bendiks, Z.; Marco, M.L.; Nørskov, N.P.; Knudsen, K.E.B.; Hedemann, M.S.; Lærke, H.N. Effects of Dietary Fibre and Protein Content on Intestinal Fibre Degradation, Short-Chain Fatty Acid and Microbiota Composition in a High-Fat Fructose-Rich Diet Induced Obese Göttingen Minipig Model. Food Funct. 2020, 11, 10758–10773. [Google Scholar] [CrossRef]
  81. Zhang, X.-Y.; Chen, J.; Yi, K.; Peng, L.; Xie, J.; Gou, X.; Peng, T.; Tang, L. Phlorizin Ameliorates Obesity-Associated Endotoxemia and Insulin Resistance in High-Fat Diet-Fed Mice by Targeting the Gut Microbiota and Intestinal Barrier Integrity. Gut Microbes 2020, 12, 1842990. [Google Scholar] [CrossRef] [PubMed]
  82. Lee, S.; Knotts, T.A.; Goodson, M.L.; Barboza, M.; Wudeck, E.; England, G.; Raybould, H.E. Metabolic Responses to Butyrate Supplementation in LF- and HF-Fed Mice Are Cohort-Dependent and Associated with Changes in Composition and Function of the Gut Microbiota. Nutrients 2020, 12, 3524. [Google Scholar] [CrossRef] [PubMed]
  83. Ramos-Romero, S.; Léniz, A.; Martínez-Maqueda, D.; Amézqueta, S.; Fernández-Quintela, A.; Hereu, M.; Torres, J.L.; Portillo, M.P.; Pérez-Jiménez, J. Inter-Individual Variability in Insulin Response after Grape Pomace Supplementation in Subjects at High Cardiometabolic Risk: Role of Microbiota and MiRNA. Mol. Nutr. Food Res. 2021, 65, e2000113. [Google Scholar] [CrossRef] [PubMed]
  84. Gyarmati, P.; Song, Y.; Dotimas, J.; Yoshiba, G.; Christison, A. Cross-Sectional Comparisons of Gut Microbiome and Short-Chain Fatty Acid Levels among Children with Varied Weight Classifications. Pediatr. Obes. 2021, 16, e12750. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, Z.; Li, L.; Ma, S.; Ye, J.; Zhang, H.; Li, Y.; Sair, A.T.; Pan, J.; Liu, X.; Li, X.; et al. High-Dietary Fiber Intake Alleviates Antenatal Obesity-Induced Postpartum Depression: Roles of Gut Microbiota and Microbial Metabolite Short-Chain Fatty Acid Involved. J. Agric. Food Chem. 2020, 68, 13697–13710. [Google Scholar] [CrossRef]
  86. Ojo, B.A.; Lu, P.; Alake, S.E.; Keirns, B.; Anderson, K.; Gallucci, G.; Hart, M.D.; El-Rassi, G.D.; Ritchey, J.W.; Chowanadisai, W.; et al. Pinto Beans Modulate the Gut Microbiome, Augment MHC II Protein, and Antimicrobial Peptide Gene Expression in Mice Fed a Normal or Western-Style Diet. J. Nutr. Biochem. 2021, 88, 108543. [Google Scholar] [CrossRef]
  87. Hijova, E.; Strojný, L.; Bertková, I.; Bomba, A.; Štofilová, J. Dietary Lactobacillus Plantarum LS/07 and Inulin in the Management of Chronic Disease Risk Factors. Acta Biochim. Pol. 2020, 67, 447–451. [Google Scholar] [CrossRef]
  88. Yelek, C.; Mignion, L.; Joudiou, N.; Terrasi, R.; Gourgue, F.; van Hul, M.; Delzenne, N.; Gallez, B.; Corbet, C.; Muccioli, G.G.; et al. Acetate: Friend or Foe against Breast Tumour Growth in the Context of Obesity? J. Cell Mol. Med. 2020, 24, 14195–14204. [Google Scholar] [CrossRef]
  89. Wang, B.; Kong, Q.; Li, X.; Zhao, J.; Zhang, H.; Chen, W.; Wang, G. A High-Fat Diet Increases Gut Microbiota Biodiversity and Energy Expenditure Due to Nutrient Difference. Nutrients 2020, 12, 3197. [Google Scholar] [CrossRef]
  90. Lee, G.; You, H.J.; Bajaj, J.S.; Joo, S.K.; Yu, J.; Park, S.; Kang, H.; Park, J.H.; Kim, J.H.; Lee, D.H.; et al. Distinct Signatures of Gut Microbiome and Metabolites Associated with Significant Fibrosis in Non-Obese NAFLD. Nat. Commun. 2020, 11, 4982. [Google Scholar] [CrossRef]
  91. Bonomo, R.R.; Cook, T.M.; Gavini, C.K.; White, C.R.; Jones, J.R.; Bovo, E.; Zima, A.V.; Brown, I.A.; Dugas, L.R.; Zakharian, E.; et al. Fecal Transplantation and Butyrate Improve Neuropathic Pain, Modify Immune Cell Profile, and Gene Expression in the PNS of Obese Mice. Proc. Natl. Acad. Sci. USA 2020, 117, 26482–26493. [Google Scholar] [CrossRef]
  92. Sato, F.T.; Yap, Y.A.; Crisma, A.R.; Portovedo, M.; Murata, G.M.; Hirabara, S.M.; Ribeiro, W.R.; Marcantonio Ferreira, C.; Cruz, M.M.; Pereira, J.N.B.; et al. Tributyrin Attenuates Metabolic and Inflammatory Changes Associated with Obesity through a GPR109A-Dependent Mechanism. Cells 2020, 9, 2007. [Google Scholar] [CrossRef]
  93. Nguyen, N.K.; Deehan, E.C.; Zhang, Z.; Jin, M.; Baskota, N.; Perez-Muñoz, M.E.; Cole, J.; Tuncil, Y.E.; Seethaler, B.; Wang, T.; et al. Gut Microbiota Modulation with Long-Chain Corn Bran Arabinoxylan in Adults with Overweight and Obesity Is Linked to an Individualized Temporal Increase in Fecal Propionate. Microbiome 2020, 8, 118. [Google Scholar] [CrossRef]
  94. Holmes, Z.C.; Silverman, J.D.; Dressman, H.K.; Wei, Z.; Dallow, E.P.; Armstrong, S.C.; Seed, P.C.; Rawls, J.F.; David, L.A. Short-Chain Fatty Acid Production by Gut Microbiota from Children with Obesity Differs According to Prebiotic Choice and Bacterial Community Composition. mBio 2020, 11, e00914-20. [Google Scholar] [CrossRef]
  95. 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]
  96. Tengeler, A.C.; Gart, E.; Wiesmann, M.; Arnoldussen, I.A.C.; van Duyvenvoorde, W.; Hoogstad, M.; Dederen, P.J.; Verweij, V.; Geenen, B.; Kozicz, T.; et al. Propionic Acid and Not Caproic Acid, Attenuates Nonalcoholic Steatohepatitis and Improves (Cerebro) Vascular Functions in Obese Ldlr−/−.Leiden Mice. FASEB J. 2020, 34, 9575–9593. [Google Scholar] [CrossRef]
  97. Farup, P.G.; Valeur, J. Changes in Faecal Short-Chain Fatty Acids after Weight-Loss Interventions in Subjects with Morbid Obesity. Nutrients 2020, 12, 802. [Google Scholar] [CrossRef] [Green Version]
  98. Jamar, G.; Santamarina, A.B.; Casagrande, B.P.; Estadella, D.; de Rosso, V.V.; Wagner, R.; Fagundes, M.B.; Pisani, L.P. Prebiotic Potencial of Juçara Berry on Changes in Gut Bacteria and Acetate of Individuals with Obesity. Eur. J. Nutr. 2020, 59, 3767–3778. [Google Scholar] [CrossRef]
  99. Takai, A.; Kikuchi, K.; Ichimura, M.; Tsuneyama, K.; Moritoki, Y.; Matsumoto, K.; Tsunashima, H.; Onda, T.; Kuniyoshi, N.; Nariyama, T.; et al. Fructo-Oligosaccharides Ameliorate Steatohepatitis, Visceral Adiposity, and Associated Chronic Inflammation via Increased Production of Short-Chain Fatty Acids in a Mouse Model of Non-Alcoholic Steatohepatitis. BMC Gastroenterol. 2020, 20, 46. [Google Scholar] [CrossRef]
  100. Sowah, S.A.; Hirche, F.; Milanese, A.; Johnson, T.S.; Grafetstätter, M.; Schübel, R.; Kirsten, R.; Ulrich, C.M.; Kaaks, R.; Zeller, G.; et al. Changes in Plasma Short-Chain Fatty Acid Levels after Dietary Weight Loss Among Overweight and Obese Adults over 50 Weeks. Nutrients 2020, 12, 452. [Google Scholar] [CrossRef]
  101. Miranda, V.P.N.; Dos Santos Amorim, P.R.; Bastos, R.R.; de Faria, E.R.; de Castro Moreira, M.E.; do Carmo Castro Franceschini, S.; do Carmo Gouveia Peluzio, M.; de Luces Fortes Ferreira, C.L.; Priore, S.E. Abundance of Gut Microbiota, Concentration of Short-Chain Fatty Acids, and Inflammatory Markers Associated with Elevated Body Fat, Overweight, and Obesity in Female Adolescents. Mediat. Inflamm. 2019, 2019, 7346863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Oh, T.J.; Sul, W.J.; Oh, H.N.; Lee, Y.-K.; Lim, H.L.; Choi, S.H.; Park, K.S.; Jang, H.C. Butyrate Attenuated Fat Gain through Gut Microbiota Modulation in Db/Db Mice Following Dapagliflozin Treatment. Sci. Rep. 2019, 9, 20300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lee, E.-S.; Song, E.-J.; Nam, Y.-D.; Nam, T.G.; Kim, H.-J.; Lee, B.-H.; Seo, M.-J.; Seo, D.-H. Effects of Enzymatically Modified Chestnut Starch on the Gut Microbiome, Microbial Metabolome, and Transcriptome of Diet-Induced Obese Mice. Int. J. Biol. Macromol. 2020, 145, 235–243. [Google Scholar] [CrossRef] [PubMed]
  104. Wallace, J.G.; Bellissimo, C.J.; Yeo, E.; Fei Xia, Y.; Petrik, J.J.; Surette, M.G.; Bowdish, D.M.E.; Sloboda, D.M. Obesity during Pregnancy Results in Maternal Intestinal Inflammation, Placental Hypoxia, and Alters Fetal Glucose Metabolism at Mid-Gestation. Sci. Rep. 2019, 9, 17621. [Google Scholar] [CrossRef]
  105. Rosado, C.P.; Rosa, V.H.C.; Martins, B.C.; Soares, A.C.; Santos, I.B.; Monteiro, E.B.; Moura-Nunes, N.; da Costa, C.A.; da Rocha Pinheiro Mulder, A.; Daleprane, J.B. Resistant Starch from Green Banana (Musa Sp.) Attenuates Non-Alcoholic Fat Liver Accumulation and Increases Short-Chain Fatty Acids Production in High-Fat Diet-Induced Obesity in Mice. Int. J. Biol. Macromol. 2020, 145, 1066–1072. [Google Scholar] [CrossRef] [PubMed]
  106. Nagano, T.; Yano, H. Effect of Dietary Cellulose Nanofiber and Exercise on Obesity and Gut Microbiota in Mice Fed a High-Fat-Diet. Biosci. Biotechnol. Biochem. 2020, 84, 613–620. [Google Scholar] [CrossRef]
  107. Farup, P.G.; Lydersen, S.; Valeur, J. Are Nonnutritive Sweeteners Obesogenic? Associations between Diet, Faecal Microbiota, and Short-Chain Fatty Acids in Morbidly Obese Subjects. J. Obes. 2019, 2019, 4608315. [Google Scholar] [CrossRef]
  108. Battson, M.L.; Lee, D.M.; Li Puma, L.C.; Ecton, K.E.; Thomas, K.N.; Febvre, H.P.; Chicco, A.J.; Weir, T.L.; Gentile, C.L. Gut Microbiota Regulates Cardiac Ischemic Tolerance and Aortic Stiffness in Obesity. Am. J. Physiol. Heart Circ. Physiol. 2019, 317, H1210–H1220. [Google Scholar] [CrossRef]
  109. Kang, D.; Su, M.; Duan, Y.; Huang, Y. Eurotium Cristatum, a Potential Probiotic Fungus from Fuzhuan Brick Tea, Alleviated Obesity in Mice by Modulating Gut Microbiota. Food Funct. 2019, 10, 5032–5045. [Google Scholar] [CrossRef]
  110. Xiao, S.; Zhang, Z.; Chen, M.; Zou, J.; Jiang, S.; Qian, D.; Duan, J. Xiexin Tang Ameliorates Dyslipidemia in High-Fat Diet-Induced Obese Rats via Elevating Gut Microbiota-Derived Short Chain Fatty Acids Production and Adjusting Energy Metabolism. J. Ethnopharmacol. 2019, 241, 112032. [Google Scholar] [CrossRef]
  111. Liu, H.-Y.; Walden, T.B.; Cai, D.; Ahl, D.; Bertilsson, S.; Phillipson, M.; Nyman, M.; Holm, L. Dietary Fiber in Bilberry Ameliorates Pre-Obesity Events in Rats by Regulating Lipid Depot, Cecal Short-Chain Fatty Acid Formation and Microbiota Composition. Nutrients 2019, 11, 1350. [Google Scholar] [CrossRef] [Green Version]
  112. Ju, M.; Liu, Y.; Li, M.; Cheng, M.; Zhang, Y.; Deng, G.; Kang, X.; Liu, H. Baicalin Improves Intestinal Microecology and Abnormal Metabolism Induced by High-Fat Diet. Eur. J. Pharmacol. 2019, 857, 172457. [Google Scholar] [CrossRef]
  113. Duan, Y.; Zhong, Y.; Xiao, H.; Zheng, C.; Song, B.; Wang, W.; Guo, Q.; Li, Y.; Han, H.; Gao, J.; et al. Gut Microbiota Mediates the Protective Effects of Dietary β-Hydroxy-β-Methylbutyrate (HMB) against Obesity Induced by High-Fat Diets. FASEB J. 2019, 33, 10019–10033. [Google Scholar] [CrossRef]
  114. Liu, J.; Hao, W.; He, Z.; Kwek, E.; Zhao, Y.; Zhu, H.; Liang, N.; Ma, K.Y.; Lei, L.; He, W.-S.; et al. Beneficial Effects of Tea Water Extracts on the Body Weight and Gut Microbiota in C57BL/6J Mice Fed with a High-Fat Diet. Food Funct. 2019, 10, 2847–2860. [Google Scholar] [CrossRef]
  115. 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]
  116. Ji, Y.; Park, S.; Chung, Y.; Kim, B.; Park, H.; Huang, E.; Jeong, D.; Jung, H.-Y.; Kim, B.; Hyun, C.-K.; et al. Amelioration of Obesity-Related Biomarkers by Lactobacillus Sakei CJLS03 in a High-Fat Diet-Induced Obese Murine Model. Sci. Rep. 2019, 9, 6821. [Google Scholar] [CrossRef] [Green Version]
  117. Fang, W.; Xue, H.; Chen, X.; Chen, K.; Ling, W. Supplementation with Sodium Butyrate Modulates the Composition of the Gut Microbiota and Ameliorates High-Fat Diet-Induced Obesity in Mice. J. Nutr. 2019, 149, 747–754. [Google Scholar] [CrossRef]
  118. Hu, S.; Wang, J.; Xu, Y.; Yang, H.; Wang, J.; Xue, C.; Yan, X.; Su, L. Anti-Inflammation Effects of Fucosylated Chondroitin Sulphate from Acaudina Molpadioides by Altering Gut Microbiota in Obese Mice. Food Funct. 2019, 10, 1736–1746. [Google Scholar] [CrossRef]
  119. Ahmadi, S.; Nagpal, R.; Wang, S.; Gagliano, J.; Kitzman, D.W.; Soleimanian-Zad, S.; Sheikh-Zeinoddin, M.; Read, R.; Yadav, H. Prebiotics from Acorn and Sago Prevent High-Fat-Diet-Induced Insulin Resistance via Microbiome-Gut-Brain Axis Modulation. J. Nutr. Biochem. 2019, 67, 1–13. [Google Scholar] [CrossRef]
  120. Xu, J.; Liu, T.; Li, Y.; Liu, W.; Ding, Z.; Ma, H.; Seeram, N.P.; Mu, Y.; Huang, X.; Li, L. Jamun (Eugenia Jambolana Lam.) Fruit Extract Prevents Obesity by Modulating the Gut Microbiome in High-Fat-Diet-Fed Mice. Mol. Nutr. Food Res. 2019, 63, e1801307. [Google Scholar] [CrossRef]
  121. Cleophas, M.C.P.; Ratter, J.M.; Bekkering, S.; Quintin, J.; Schraa, K.; Stroes, E.S.; Netea, M.G.; Joosten, L.A.B. Effects of Oral Butyrate Supplementation on Inflammatory Potential of Circulating Peripheral Blood Mononuclear Cells in Healthy and Obese Males. Sci. Rep. 2019, 9, 775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Okouchi, R.; Shuang, E.; Yamamoto, K.; Ota, T.; Seki, K.; Imai, M.; Ota, R.; Asayama, Y.; Nakashima, A.; Suzuki, K.; et al. Simultaneous Intake of Euglena Gracilis and Vegetables Exerts Synergistic Anti-Obesity and Anti-Inflammatory Effects by Modulating the Gut Microbiota in Diet-Induced Obese Mice. Nutrients 2019, 11, 204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. De la Cuesta-Zuluaga, J.; Mueller, N.T.; Álvarez-Quintero, R.; Velásquez-Mejía, E.P.; Sierra, J.A.; Corrales-Agudelo, V.; Carmona, J.A.; Abad, J.M.; Escobar, J.S. Higher Fecal Short-Chain Fatty Acid Levels Are Associated with Gut Microbiome Dysbiosis, Obesity, Hypertension and Cardiometabolic Disease Risk Factors. Nutrients 2018, 11, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Gart, E.; Souto Lima, E.; Schuren, F.; de Ruiter, C.G.F.; Attema, J.; Verschuren, L.; Keijer, J.; Salic, K.; Morrison, M.C.; Kleemann, R. Diet-Independent Correlations between Bacteria and Dysfunction of Gut, Adipose Tissue, and Liver: A Comprehensive Microbiota Analysis in Feces and Mucosa of the Ileum and Colon in Obese Mice with NAFLD. Int. J. Mol. Sci. 2018, 20, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Mayengbam, S.; Lambert, J.E.; Parnell, J.A.; Tunnicliffe, J.M.; Nicolucci, A.C.; Han, J.; Sturzenegger, T.; Shearer, J.; Mickiewicz, B.; Vogel, H.J.; et al. Impact of Dietary Fiber Supplementation on Modulating Microbiota-Host-Metabolic Axes in Obesity. J. Nutr. Biochem. 2019, 64, 228–236. [Google Scholar] [CrossRef]
  126. Arora, T.; Rudenko, O.; Egerod, K.L.; Husted, A.S.; Kovatcheva-Datchary, P.; Akrami, R.; Kristensen, M.; Schwartz, T.W.; Bäckhed, F. Microbial Fermentation of Flaxseed Fibers Modulates the Transcriptome of GPR41-Expressing Enteroendocrine Cells and Protects Mice against Diet-Induced Obesity. Am. J. Physiol. Endocrinol. Metab. 2019, 316, E453–E463. [Google Scholar] [CrossRef]
  127. Dugas, L.R.; Bernabé, B.P.; Priyadarshini, M.; Fei, N.; Park, S.J.; Brown, L.; Plange-Rhule, J.; Nelson, D.; Toh, E.C.; Gao, X.; et al. Decreased Microbial Co-Occurrence Network Stability and SCFA Receptor Level Correlates with Obesity in African-Origin Women. Sci. Rep. 2018, 8, 17135. [Google Scholar] [CrossRef]
  128. Barczyńska, R.; Litwin, M.; Sliżewska, K.; Szalecki, M.; Berdowska, A.; Bandurska, K.; Libudzisz, Z.; Kapuśniak, J. Bacterial Microbiota and Fatty Acids in the Faeces of Overweight and Obese Children. Pol. J. Microbiol. 2018, 67, 339–345. [Google Scholar] [CrossRef] [Green Version]
  129. Barnes, R.C.; Kim, H.; Fang, C.; Bennett, W.; Nemec, M.; Sirven, M.A.; Suchodolski, J.S.; Deutz, N.; Britton, R.A.; Mertens-Talcott, S.U.; et al. Body Mass Index as a Determinant of Systemic Exposure to Gallotannin Metabolites during 6-Week Consumption of Mango (Mangifera Indica L.) and Modulation of Intestinal Microbiota in Lean and Obese Individuals. Mol. Nutr. Food Res. 2019, 63, e1800512. [Google Scholar] [CrossRef]
  130. Zhai, X.; Lin, D.; Zhao, Y.; Li, W.; Yang, X. Effects of Dietary Fiber Supplementation on Fatty Acid Metabolism and Intestinal Microbiota Diversity in C57BL/6J Mice Fed with a High-Fat Diet. J. Agric. Food Chem. 2018, 66, 12706–12718. [Google Scholar] [CrossRef]
  131. Bianchi, F.; Larsen, N.; de Mello Tieghi, T.; Adorno, M.A.T.; Kot, W.; Saad, S.M.I.; Jespersen, L.; Sivieri, K. Modulation of Gut Microbiota from Obese Individuals by in Vitro Fermentation of Citrus Pectin in Combination with Bifidobacterium Longum BB-46. Appl. Microbiol. Biotechnol. 2018, 102, 8827–8840. [Google Scholar] [CrossRef] [Green Version]
  132. Alexander, C.; Cross, T.-W.L.; Devendran, S.; Neumer, F.; Theis, S.; Ridlon, J.M.; Suchodolski, J.S.; de Godoy, M.R.C.; Swanson, K.S. Effects of Prebiotic Inulin-Type Fructans on Blood Metabolite and Hormone Concentrations and Faecal Microbiota and Metabolites in Overweight Dogs. Br. J. Nutr. 2018, 120, 711–720. [Google Scholar] [CrossRef] [Green Version]
  133. Zarrinpar, A.; Chaix, A.; Xu, Z.Z.; Chang, M.W.; Marotz, C.A.; Saghatelian, A.; Knight, R.; Panda, S. Antibiotic-Induced Microbiome Depletion Alters Metabolic Homeostasis by Affecting Gut Signaling and Colonic Metabolism. Nat. Commun. 2018, 9, 2872. [Google Scholar] [CrossRef] [Green Version]
  134. Van der Beek, C.M.; Canfora, E.E.; Kip, A.M.; Gorissen, S.H.M.; Olde Damink, S.W.M.; van Eijk, H.M.; Holst, J.J.; Blaak, E.E.; Dejong, C.H.C.; Lenaerts, K. The Prebiotic Inulin Improves Substrate Metabolism and Promotes Short-Chain Fatty Acid Production in Overweight to Obese Men. Metabolism 2018, 87, 25–35. [Google Scholar] [CrossRef]
  135. Lu, Y.; Fan, C.; Liang, A.; Fan, X.; Wang, R.; Li, P.; Qi, K. Effects of SCFA on the DNA Methylation Pattern of Adiponectin and Resistin in High-Fat-Diet-Induced Obese Male Mice. Br. J. Nutr. 2018, 120, 385–392. [Google Scholar] [CrossRef] [Green Version]
  136. Liu, J.; Yue, S.; Yang, Z.; Feng, W.; Meng, X.; Wang, A.; Peng, C.; Wang, C.; Yan, D. Oral Hydroxysafflor Yellow A Reduces Obesity in Mice by Modulating the Gut Microbiota and Serum Metabolism. Pharmacol. Res. 2018, 134, 40–50. [Google Scholar] [CrossRef]
  137. Miyamoto, J.; Watanabe, K.; Taira, S.; Kasubuchi, M.; Li, X.; Irie, J.; Itoh, H.; Kimura, I. Barley β-Glucan Improves Metabolic Condition via Short-Chain Fatty Acids Produced by Gut Microbial Fermentation in High Fat Diet Fed Mice. PLoS ONE 2018, 13, e0196579. [Google Scholar] [CrossRef]
  138. Den Hartigh, L.J.; Gao, Z.; Goodspeed, L.; Wang, S.; Das, A.K.; Burant, C.F.; Chait, A.; Blaser, M.J. Obese Mice Losing Weight Due to Trans-10,Cis-12 Conjugated Linoleic Acid Supplementation or Food Restriction Harbor Distinct Gut Microbiota. J. Nutr. 2018, 148, 562–572. [Google Scholar] [CrossRef]
  139. Wu, T.; Yang, L.; Guo, X.; Zhang, M.; Liu, R.; Sui, W. Raspberry Anthocyanin Consumption Prevents Diet-Induced Obesity by Alleviating Oxidative Stress and Modulating Hepatic Lipid Metabolism. Food Funct. 2018, 9, 2112–2120. [Google Scholar] [CrossRef]
  140. Aguilar, E.C.; da Silva, J.F.; Navia-Pelaez, J.M.; Leonel, A.J.; Lopes, L.G.; Menezes-Garcia, Z.; Ferreira, A.V.M.; Capettini, L.D.S.A.; Teixeira, L.G.; Lemos, V.S.; et al. Sodium Butyrate Modulates Adipocyte Expansion, Adipogenesis, and Insulin Receptor Signaling by Upregulation of PPAR-γ in Obese Apo E Knockout Mice. Nutrition 2018, 47, 75–82. [Google Scholar] [CrossRef]
  141. Zeng, H.; Ishaq, S.L.; Liu, Z.; Bukowski, M.R. Colonic Aberrant Crypt Formation Accompanies an Increase of Opportunistic Pathogenic Bacteria in C57BL/6 Mice Fed a High-Fat Diet. J. Nutr. Biochem. 2018, 54, 18–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Allen, J.M.; Mailing, L.J.; Niemiro, G.M.; Moore, R.; Cook, M.D.; White, B.A.; Holscher, H.D.; Woods, J.A. Exercise Alters Gut Microbiota Composition and Function in Lean and Obese Humans. Med. Sci. Sports Exerc. 2018, 50, 747–757. [Google Scholar] [CrossRef] [PubMed]
  143. Moreno-Navarrete, J.M.; Serino, M.; Blasco-Baque, V.; Azalbert, V.; Barton, R.H.; Cardellini, M.; Latorre, J.; Ortega, F.; Sabater-Masdeu, M.; Burcelin, R.; et al. Gut Microbiota Interacts with Markers of Adipose Tissue Browning, Insulin Action and Plasma Acetate in Morbid Obesity. Mol. Nutr. Food Res. 2018, 62, 1700721. [Google Scholar] [CrossRef] [PubMed]
  144. Wu, T.; Guo, X.; Zhang, M.; Yang, L.; Liu, R.; Yin, J. Anthocyanins in Black Rice, Soybean and Purple Corn Increase Fecal Butyric Acid and Prevent Liver Inflammation in High Fat Diet-Induced Obese Mice. Food Funct. 2017, 8, 3178–3186. [Google Scholar] [CrossRef] [PubMed]
  145. Weitkunat, K.; Stuhlmann, C.; Postel, A.; Rumberger, S.; Fankhänel, M.; Woting, A.; Petzke, K.J.; Gohlke, S.; Schulz, T.J.; Blaut, M.; et al. Short-Chain Fatty Acids and Inulin, but Not Guar Gum, Prevent Diet-Induced Obesity and Insulin Resistance through Differential Mechanisms in Mice. Sci. Rep. 2017, 7, 6109. [Google Scholar] [CrossRef] [Green Version]
  146. Pelgrim, C.E.; Franx, B.A.A.; Snabel, J.; Kleemann, R.; Arnoldussen, I.A.C.; Kiliaan, A.J. Butyrate Reduces HFD-Induced Adipocyte Hypertrophy and Metabolic Risk Factors in Obese LDLr−/−.Leiden Mice. Nutrients 2017, 9, 714. [Google Scholar] [CrossRef] [Green Version]
  147. Canfora, E.E.; van der Beek, C.M.; Hermes, G.D.A.; Goossens, G.H.; Jocken, J.W.E.; Holst, J.J.; van Eijk, H.M.; Venema, K.; Smidt, H.; Zoetendal, E.G.; et al. Supplementation of Diet With Galacto-Oligosaccharides Increases Bifidobacteria, but Not Insulin Sensitivity, in Obese Prediabetic Individuals. Gastroenterology 2017, 153, 87–97.e3. [Google Scholar] [CrossRef]
  148. Prins, G.H.; Rios-Morales, M.; Gerding, A.; Reijngoud, D.-J.; Olinga, P.; Bakker, B.M. The Effects of Butyrate on Induced Metabolic-Associated Fatty Liver Disease in Precision-Cut Liver Slices. Nutrients 2021, 13, 4203. [Google Scholar] [CrossRef]
  149. Ratajczak, W.; Mizerski, A.; Rył, A.; Słojewski, M.; Sipak, O.; Piasecka, M.; Laszczyńska, M. Alterations in Fecal Short Chain Fatty Acids (SCFAs) and Branched Short-Chain Fatty Acids (BCFAs) in Men with Benign Prostatic Hyperplasia (BPH) and Metabolic Syndrome (MetS). Aging 2021, 13, 10934–10954. [Google Scholar] [CrossRef]
  150. Adeyanju, O.A.; Badejogbin, O.C.; Areola, D.E.; Olaniyi, K.S.; Dibia, C.; Soetan, O.A.; Oniyide, A.A.; Michael, O.S.; Olatunji, L.A.; Soladoye, A.O. Sodium Butyrate Arrests Pancreato-Hepatic Synchronous Uric Acid and Lipid Dysmetabolism in High Fat Diet Fed Wistar Rats. Biomed. Pharmacother. 2021, 133, 110994. [Google Scholar] [CrossRef]
  151. Hartstra, A.V.; Schüppel, V.; Imangaliyev, S.; Schrantee, A.; Prodan, A.; Collard, D.; Levin, E.; Dallinga-Thie, G.; Ackermans, M.T.; Winkelmeijer, M.; et al. Infusion of Donor Feces Affects the Gut-Brain Axis in Humans with Metabolic Syndrome. Mol. Metab. 2020, 42, 101076. [Google Scholar] [CrossRef]
  152. Hu, B.; Ye, C.; Leung, E.L.-H.; Zhu, L.; Hu, H.; Zhang, Z.; Zheng, J.; Liu, H. Bletilla Striata Oligosaccharides Improve Metabolic Syndrome through Modulation of Gut Microbiota and Intestinal Metabolites in High Fat Diet-Fed Mice. Pharmacol. Res. 2020, 159, 104942. [Google Scholar] [CrossRef]
  153. Polito, R.; Costabile, G.; Nigro, E.; Giacco, R.; Vetrani, C.; Anniballi, G.; Luongo, D.; Riccardi, G.; Daniele, A.; Annuzzi, G. Nutritional Factors Influencing Plasma Adiponectin Levels: Results from a Randomised Controlled Study with Whole-Grain Cereals. Int. J. Food Sci. Nutr. 2020, 71, 509–515. [Google Scholar] [CrossRef]
  154. De Groot, P.; Scheithauer, T.; Bakker, G.J.; Prodan, A.; Levin, E.; Khan, M.T.; Herrema, H.; Ackermans, M.; Serlie, M.J.M.; de Brauw, M.; et al. Donor Metabolic Characteristics Drive Effects of Faecal Microbiota Transplantation on Recipient Insulin Sensitivity, Energy Expenditure and Intestinal Transit Time. Gut 2020, 69, 502–512. [Google Scholar] [CrossRef] [Green Version]
  155. Velikonja, A.; Lipoglavšek, L.; Zorec, M.; Orel, R.; Avguštin, G. Alterations in Gut Microbiota Composition and Metabolic Parameters after Dietary Intervention with Barley Beta Glucans in Patients with High Risk for Metabolic Syndrome Development. Anaerobe 2019, 55, 67–77. [Google Scholar] [CrossRef]
  156. Nishitsuji, K.; Watanabe, S.; Xiao, J.; Nagatomo, R.; Ogawa, H.; Tsunematsu, T.; Umemoto, H.; Morimoto, Y.; Akatsu, H.; Inoue, K.; et al. Effect of Coffee or Coffee Components on Gut Microbiome and Short-Chain Fatty Acids in a Mouse Model of Metabolic Syndrome. Sci. Rep. 2018, 8, 16173. [Google Scholar] [CrossRef] [Green Version]
  157. Bouter, K.; Bakker, G.J.; Levin, E.; Hartstra, A.V.; Kootte, R.S.; Udayappan, S.D.; Katiraei, S.; Bahler, L.; Gilijamse, P.W.; Tremaroli, V.; et al. Differential Metabolic Effects of Oral Butyrate Treatment in Lean versus Metabolic Syndrome Subjects. Clin. Transl. Gastroenterol. 2018, 9, 155. [Google Scholar] [CrossRef]
  158. Si, X.; Shang, W.; Zhou, Z.; Shui, G.; Lam, S.M.; Blanchard, C.; Strappe, P. Gamma-Aminobutyric Acid Enriched Rice Bran Diet Attenuates Insulin Resistance and Balances Energy Expenditure via Modification of Gut Microbiota and Short-Chain Fatty Acids. J. Agric. Food Chem. 2018, 66, 881–890. [Google Scholar] [CrossRef]
  159. Nishitsuji, K.; Xiao, J.; Nagatomo, R.; Umemoto, H.; Morimoto, Y.; Akatsu, H.; Inoue, K.; Tsuneyama, K. Analysis of the Gut Microbiome and Plasma Short-Chain Fatty Acid Profiles in a Spontaneous Mouse Model of Metabolic Syndrome. Sci. Rep. 2017, 7, 15876. [Google Scholar] [CrossRef] [Green Version]
  160. Singh, D.P.; Singh, S.; Bijalwan, V.; Kumar, V.; Khare, P.; Baboota, R.K.; Singh, P.; Boparai, R.K.; Singh, J.; Kondepudi, K.K.; et al. Co-Supplementation of Isomalto-Oligosaccharides Potentiates Metabolic Health Benefits of Polyphenol-Rich Cranberry Extract in High Fat Diet-Fed Mice via Enhanced Gut Butyrate Production. Eur. J. Nutr. 2018, 57, 2897–2911. [Google Scholar] [CrossRef]
  161. Rivero-Gutiérrez, B.; Gámez-Belmonte, R.; Suárez, M.D.; Lavín, J.L.; Aransay, A.M.; Olivares, M.; Martínez-Augustin, O.; Sánchez de Medina, F.; Zarzuelo, A. A Synbiotic Composed of Lactobacillus Fermentum CECT5716 and FOS Prevents the Development of Fatty Acid Liver and Glycemic Alterations in Rats Fed a High Fructose Diet Associated with Changes in the Microbiota. Mol. Nutr. Food Res. 2017, 61, 1600622. [Google Scholar] [CrossRef] [PubMed]
  162. Dai, Z.; Lyu, W.; Xie, M.; Yuan, Q.; Ye, H.; Hu, B.; Zhou, L.; Zeng, X. Effects of α-Galactooligosaccharides from Chickpeas on High-Fat-Diet-Induced Metabolic Syndrome in Mice. J. Agric. Food Chem. 2017, 65, 3160–3166. [Google Scholar] [CrossRef] [PubMed]
  163. Hald, S.; Schioldan, A.G.; Moore, M.E.; Dige, A.; Lærke, H.N.; Agnholt, J.; Bach Knudsen, K.E.; Hermansen, K.; Marco, M.L.; Gregersen, S.; et al. Effects of Arabinoxylan and Resistant Starch on Intestinal Microbiota and Short-Chain Fatty Acids in Subjects with Metabolic Syndrome: A Randomised Crossover Study. PLoS ONE 2016, 11, e0159223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Upadhyaya, B.; McCormack, L.; Fardin-Kia, A.R.; Juenemann, R.; Nichenametla, S.; Clapper, J.; Specker, B.; Dey, M. Impact of Dietary Resistant Starch Type 4 on Human Gut Microbiota and Immunometabolic Functions. Sci. Rep. 2016, 6, 28797. [Google Scholar] [CrossRef] [PubMed]
  165. Vetrani, C.; Costabile, G.; Luongo, D.; Naviglio, D.; Rivellese, A.A.; Riccardi, G.; Giacco, R. Effects of Whole-Grain Cereal Foods on Plasma Short Chain Fatty Acid Concentrations in Individuals with the Metabolic Syndrome. Nutrition 2016, 32, 217–221. [Google Scholar] [CrossRef]
  166. Singh, V.; Chassaing, B.; Zhang, L.; San Yeoh, B.; Xiao, X.; Kumar, M.; Baker, M.T.; Cai, J.; Walker, R.; Borkowski, K.; et al. Microbiota-Dependent Hepatic Lipogenesis Mediated by Stearoyl CoA Desaturase 1 (SCD1) Promotes Metabolic Syndrome in TLR5-Deficient Mice. Cell Metab. 2015, 22, 983–996. [Google Scholar] [CrossRef] [Green Version]
  167. Den Besten, G.; Gerding, A.; van Dijk, T.H.; Ciapaite, J.; Bleeker, A.; van Eunen, K.; Havinga, R.; Groen, A.K.; Reijngoud, D.-J.; Bakker, B.M. Protection against the Metabolic Syndrome by Guar Gum-Derived Short-Chain Fatty Acids Depends on Peroxisome Proliferator-Activated Receptor γ and Glucagon-Like Peptide-1. PLoS ONE 2015, 10, e0136364. [Google Scholar] [CrossRef]
  168. Costabile, A.; Walton, G.E.; Tzortzis, G.; Vulevic, J.; Charalampopoulos, D.; Gibson, G.R. Development of a Bread Delivery Vehicle for Dietary Prebiotics to Enhance Food Functionality Targeted at Those with Metabolic Syndrome. Gut Microbes 2015, 6, 300–309. [Google Scholar] [CrossRef] [Green Version]
  169. Den Besten, G.; Havinga, R.; Bleeker, A.; Rao, S.; Gerding, A.; van Eunen, K.; Groen, A.K.; Reijngoud, D.-J.; Bakker, B.M. The Short-Chain Fatty Acid Uptake Fluxes by Mice on a Guar Gum Supplemented Diet Associate with Amelioration of Major Biomarkers of the Metabolic Syndrome. PLoS ONE 2014, 9, e107392. [Google Scholar] [CrossRef] [Green Version]
  170. Salonen, A.; Lahti, L.; Salojärvi, J.; Holtrop, G.; Korpela, K.; Duncan, S.H.; Date, P.; Farquharson, F.; Johnstone, A.M.; Lobley, G.E.; et al. Impact of Diet and Individual Variation on Intestinal Microbiota Composition and Fermentation Products in Obese Men. ISME J. 2014, 8, 2218–2230. [Google Scholar] [CrossRef]
  171. Hartvigsen, M.L.; Lærke, H.N.; Overgaard, A.; Holst, J.J.; Bach Knudsen, K.E.; Hermansen, K. Postprandial Effects of Test Meals Including Concentrated Arabinoxylan and Whole Grain Rye in Subjects with the Metabolic Syndrome: A Randomised Study. Eur J. Clin. Nutr. 2014, 68, 567–574. [Google Scholar] [CrossRef]
  172. Larsen, N.; Vogensen, F.K.; Gøbel, R.J.; Michaelsen, K.F.; Forssten, S.D.; Lahtinen, S.J.; Jakobsen, M. Effect of Lactobacillus Salivarius Ls-33 on Fecal Microbiota in Obese Adolescents. Clin. Nutr. 2013, 32, 935–940. [Google Scholar] [CrossRef]
  173. Teixeira, T.F.S.; Grześkowiak, Ł.; Franceschini, S.C.C.; Bressan, J.; Ferreira, C.L.L.F.; Peluzio, M.C.G. Higher Level of Faecal SCFA in Women Correlates with Metabolic Syndrome Risk Factors. Br. J. Nutr. 2013, 109, 914–919. [Google Scholar] [CrossRef] [Green Version]
  174. Fava, F.; Gitau, R.; Griffin, B.A.; Gibson, G.R.; Tuohy, K.M.; Lovegrove, J.A. The Type and Quantity of Dietary Fat and Carbohydrate Alter Faecal Microbiome and Short-Chain Fatty Acid Excretion in a Metabolic Syndrome “at-Risk” Population. Int J. Obes. 2013, 37, 216–223. [Google Scholar] [CrossRef] [Green Version]
  175. Hutkins, R.W.; Krumbeck, J.A.; Bindels, L.B.; Cani, P.D.; Fahey, G.; Goh, Y.J.; Hamaker, B.; Martens, E.C.; Mills, D.A.; Rastal, R.A.; et al. Prebiotics: Why Definitions Matter. Curr. Opin. Biotechnol. 2016, 37, 1–7. [Google Scholar] [CrossRef] [Green Version]
  176. Oliphant, K.; Allen-Vercoe, E. Macronutrient Metabolism by the Human Gut Microbiome: Major Fermentation by-Products and Their Impact on Host Health. Microbiome 2019, 7, 91. [Google Scholar] [CrossRef]
Figure 1. Summary of the key effects of the three main SCFAs. Abbreviations: Ac, acetate; Prop, propionate; Bu, butyrate; GRP, G protein-coupled receptor. The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license; https://smart.servier.com/ (accessed on 21 September 2022).
Figure 1. Summary of the key effects of the three main SCFAs. Abbreviations: Ac, acetate; Prop, propionate; Bu, butyrate; GRP, G protein-coupled receptor. The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license; https://smart.servier.com/ (accessed on 21 September 2022).
Biology 11 01556 g001
Figure 2. Flow diagram for the identification, selection, and inclusion process.
Figure 2. Flow diagram for the identification, selection, and inclusion process.
Biology 11 01556 g002
Figure 3. Overview of the effects of dietary supplements and SCFAs. Abbreviations: Ac, acetate; Prop, propionate; Bu, butyrate. The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license; https://smart.servier.com/ (accessed on 21 September 2022).
Figure 3. Overview of the effects of dietary supplements and SCFAs. Abbreviations: Ac, acetate; Prop, propionate; Bu, butyrate. The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license; https://smart.servier.com/ (accessed on 21 September 2022).
Biology 11 01556 g003
Table 1. Effects on SCFA levels of dietary supplements on HFD-fed mice and rats.
Table 1. Effects on SCFA levels of dietary supplements on HFD-fed mice and rats.
Dietary SupplementSCFA VariationsReference
Bilberry↑fSCFA, bSCFA [111]
Guar gum↑fAc, fBu [56]
↑fSCFA [167,169]
Erythritol↑fSCFA, bSCFA [60]
Nobiletin↑fSCFA [66]
Baicalin↑fSCFA [112]
Barley β-glucan↑fSCFA [70]
↑fAc, fProp [72]
↑fBu [137]
Betaine↑fAc, fBu [73]
Coarse cereal mixture↑fSCFA [77]
Phlorizin↑fSCFA [81]
Inulin↑fSCFA [85]
Pinto beans↑fBu [86]
Deinococcus geotermalis modified chestnut starch↑fAc [103]
Green banana↑fSCFA [105]
β-hydroxy-β-methylbutyrate↑fProp [113]
Tea extract↑fSCFA [114]
Chondroitin sulfate↑bSCFA [118]
Jamun fruit extract↑fSCFA [120]
Euglena + vegetables↑fSCFA [122]
Flaxseed fiber↑fSCFA [126]
Bacterial cellulose + konjac glucomannan↑fAc, fProp, fBu [130]
Hydroxysafflor yellow A↑fSCFA [136]
Anthocyanins↑fBu [139,144]
Bletilla striata↑fAc
↓fProp
[152]
Cranberry extract + isomalto-oligosaccharides↑fSCFA, fBu [160]
Chickpea α-galacto-oligosaccharides↑fProp, fBu [162]
Caffeine + epigallocatechin-3-gallate↑fAc, fProp [65]
Trilobatin↑fProp, fBu [68]
Xiexin Tang↑fSCFA [110]
Gamma-aminobutyric acid enriched rice bran↑fProp, fBu [158]
Trans-10,cis-12 Conjugated linoleic acid↑bAc, fBu [138]
Lard fat + sucrose↑bProp [124]
Lard fat + sucrose + fructose↓bAc, bBu [124]
Abbreviations: ↑, increase; ↓, decrease; fSCFA, fecal short-chain fatty acids; bSCFA, blood short-chain fatty acids; N/A, not applicable; fAc, fecal acetate; fBu, fecal butyrate; fProp, fecal propionate; bAc, blood acetate.
Table 3. Effects of probiotics on HFD-fed mice and rats.
Table 3. Effects of probiotics on HFD-fed mice and rats.
ProbioticSCFA VariationsReference
Bifidobacterium adolescentis↓fSCFA [69]
Clostridium cochlearum↓fSCFA [40]
Eurotium cristatum↑fBu [109]
Lactobacillus plantarum↑fSCFA [87]
Lactobacillus reuteri↑fBu, bBu [53]
Lactobacillus sakei↑fSCFA, bSCFA [116]
↑fBu [79]
Abbreviations: ↑, increase; ↓, decrease; fSCFA, fecal short-chain fatty acids; fBu, fecal butyrate; bBu, blood butyrate; bSCFA, blood short-chain fatty acids.
Table 4. Effects of dietary supplements and various diets on SCFA levels in humans.
Table 4. Effects of dietary supplements and various diets on SCFA levels in humans.
Dietary SupplementSCFA VariationsReference
Arabinoxylan↑bAc, bBu [171]
↑fAc, fBu [163]
↑fProp [93]
Inulin↑bAc [134]
Grape pomace↓fBu [83]
Rye↑bBu [36]
Yacon flour↓fSCFA [63]
Energy-restricted dietNo effect on bSCFA [100]
↓fAc, fBu [43]
Intermittent-fasting dietNo effect on bSCFA [100]
Mediterranean diet↑bBu [95]
Vegan diet↑fBu [47]
Barley β-glucan↑fProp [155]
Galacto-oligosaccharidesNo effect on bSCFA or fSCFA [147]
AlmondNo effect on fSCFA [76]
Pea fiber↑fAc [125]
Whole-grain cereal↑bAc, bSCFA [153]
↑bProp [165]
Wheat bran↑bAc, bProp, bSCFA 1 [54]
Refined cereal↑bAc, bSCFA [153]
Mango↑fBu 2 [129]
Resistant starch type 4↑fProp, fBu [164]
Juçara berry↑fAc [98]
Saturated fat↑fSCFA [174]
Monounsaturated fat↑fSCFA [174]
1 only in obese individuals; 2 only in lean individuals. Abbreviations: ↑, increase; ↓, decrease; bAc, blood acetate; bBu, blood butyrate; fAc, fecal acetate; fBu, faecal butyrate; fProp, fecal propionate; fSCFA, fecal short-chain fatty acids; bSCFA, blood short-chain fatty acids; bProp, blood propionate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ilyés, T.; Silaghi, C.N.; Crăciun, A.M. Diet-Related Changes of Short-Chain Fatty Acids in Blood and Feces in Obesity and Metabolic Syndrome. Biology 2022, 11, 1556. https://doi.org/10.3390/biology11111556

AMA Style

Ilyés T, Silaghi CN, Crăciun AM. Diet-Related Changes of Short-Chain Fatty Acids in Blood and Feces in Obesity and Metabolic Syndrome. Biology. 2022; 11(11):1556. https://doi.org/10.3390/biology11111556

Chicago/Turabian Style

Ilyés, Tamás, Ciprian N. Silaghi, and Alexandra M. Crăciun. 2022. "Diet-Related Changes of Short-Chain Fatty Acids in Blood and Feces in Obesity and Metabolic Syndrome" Biology 11, no. 11: 1556. https://doi.org/10.3390/biology11111556

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