Next Article in Journal
Alkoxyalkylation of Electron-Rich Aromatic Compounds
Next Article in Special Issue
The Effect of Maternal High-Fat or High-Carbohydrate Diet during Pregnancy and Lactation on Cytochrome P450 2D (CYP2D) in the Liver and Brain of Rat Offspring
Previous Article in Journal
Candidate Genes and Favorable Haplotypes Associated with Iron Toxicity Tolerance in Rice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Postbiotic Properties of Butyrate in the Modulation of the Gut Microbiota: The Potential of Its Combination with Polyphenols and Dietary Fibers

1
IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy
2
Department of Systems Medicine, University of Rome Tor Vergata, 00133 Roma, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(13), 6971; https://doi.org/10.3390/ijms25136971
Submission received: 28 May 2024 / Revised: 15 June 2024 / Accepted: 19 June 2024 / Published: 26 June 2024
(This article belongs to the Special Issue Nutrition as a Tool for Modulating Cellular Metabolism)

Abstract

:
The gut microbiota is a diverse bacterial community consisting of approximately 2000 species, predominantly from five phyla: Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. The microbiota’s bacterial species create distinct compounds that impact the host’s health, including well-known short-chain fatty acids. These are produced through the breakdown of dietary fibers and fermentation of undigested carbohydrates by the intestinal microbiota. The main short-chain fatty acids consist of acetate, propionate, and butyrate. The concentration of butyrate in mammalian intestines varies depending on the diet. Its main functions are use as an energy source, cell differentiation, reduction in the inflammatory process in the intestine, and defense against oxidative stress. It also plays an epigenetic role in histone deacetylases, thus helping to reduce the risk of colon cancer. Finally, butyrate affects the gut–brain axis by crossing the brain–blood barrier, making it crucial to determine the right concentrations for both local and peripheral effects. In recent years, there has been a significant amount of attention given to the role of dietary polyphenols and fibers in promoting human health. Polyphenols and dietary fibers both play crucial roles in protecting human health and can produce butyrate through gut microbiota fermentation. This paper aims to summarize information on the key summits related to the negative correlation between intestinal microbiota diversity and chronic diseases to guide future research on determining the specific activity of butyrate from polyphenols and dietary fibers that can carry out these vital functions.

1. Introduction

At birth, human organisms are colonized by a global microbiota consisting of bacterial species, viruses, and fungi. The microbiota inhabits various areas of the body, such as the skin, digestive tract, mouth, respiratory system, and more [1]. The largest and most famous microbiota is found in the intestines [2]: the microbiota in the gastrointestinal tract consists of approximately 100 trillion bacteria and 2000 bacterial species that change and adapt throughout life, becoming unique to each person [3]. Microbiota microorganisms outnumber somatic cells by a factor of 10 [4], and their genome is 150 times larger than the human genome [5,6]. The microbiota’s genetic information contains thousands of genes that are not found in the human genome. These genes play crucial roles in the host’s physiological functions [7]. The significance of the gut microbiota cannot be overstated, as it is anticipated to have crucial functions in preserving human health [8]. The microbiota bacteria and the host engage in a mutually beneficial symbiotic co-metabolism. Many factors contribute to the change in microbiota composition throughout life, such as nutrition, age, pH, lifestyle, and more [9,10]. In healthy adults, the intestinal microbiota is made up of five bacterial phyla: Firmicutes (79.4%), Bacteroidetes (16.9%), Actinobacteria (2.5%), Proteobacteria (1%), and Verrucomicrobia (0.1%) [11]. The dietary model suggests that the microbiota can be classified into three enterotypes, with each one dominated by a specific bacterial species. Enterotype 1, for example, is characterized by Bacteroides, which derives energy from fermenting proteins and carbohydrates. Enterotype 1 is linked to a typical Western diet, characterized by its high intake of animal proteins and fats and low fiber and vegetable content [12]. Enterotype 2 is associated with a fiber- and carbohydrate-rich diet, while Enterotype 3 relies on simple sugars for energy and may be connected to weight gain [13]. When the gut microbiota contains a large number and variety of microbial species, it creates a state of physical well-being called “eubiosis”. Conversely, intestinal “dysbiosis” can refer either to a general disruption of the normal bacterial flora or the overgrowth of harmful bacteria, leading to inflammation [14,15]. Intestinal dysbiosis is related to the occurrence of various diseases, such as obesity, hypertension, diabetes mellitus type 2, inflammatory bowel diseases, Crohn’s disease, ulcerative colitis, necrotizing enterocolitis, autoimmune diseases, and colorectal cancer [16,17]. Many scientific studies have shown a two-way relationship between the microbiota and the central nervous system, known as the gut–brain axis, which is now a major area of interest in neuroscience [18]. Various studies have indicated that interventions targeting the microbiota can improve symptoms and conditions of neurological diseases [19,20,21,22]. The microbiota’s bacterial species create numerous distinct metabolites that impact the host’s health. Among these molecules, short-chain fatty acids (SCFAs), products obtained by the digestion of food fibers and the fermentation of undigested carbohydrates by the intestinal microbiota, are well known. SCFAs are saturated fatty acids containing fewer than six carbon atoms. Typically, the colon contains these compounds: acetate, propionate, and butyrate [23]. Valerate, caproate, and formate are other SCFAs found in smaller quantities [24]. Colon cells absorb SCFAs through active transport mediated by monocarboxylate transporters (MCTs) and use them in the citric acid cycle to produce cellular energy [25]. Hepatocytes utilize non-metabolized SCFAs from the colon to generate energy and synthesize glucose, cholesterol, and fatty acids [26]. SCFAs that are left in small quantities enter the systemic circulation and peripheral tissues [27]. The whole metabolic process of SCFAs is represented in Figure 1.
SCFAs have multiple beneficial effects on the epithelial, immune, nervous, and blood vessel systems [28]. A decrease in the production of these metabolites has been linked to various diseases such as intestinal inflammation, diabetes, liver cirrhosis, and atherosclerosis [29]. SCFAs play a crucial role in improving gastrointestinal health by acting locally on the intestine. These metabolites help to preserve the integrity of the intestinal barrier, aiding nutrient absorption and blocking pathogens and harmful substances [30,31]. Additionally, SCFAs enhance mucus production, which helps to lubricate intestinal epithelial cells and aids in digestion. Specifically, acetate and butyrate stimulate mucin secretion [32,33,34]. Research has shown that SCFAs can penetrate the blood–brain barrier (BBB) and access the brain. The three main SCFAs that accumulate in the brain follow this order: butyrate > propionate > acetate, at a ratio of 4.6:3.1:1.4 [35]. The passage of SCFAs through the BBB was verified through experiments involving radio-labeled SCFAs with 14C [36,37]. The expression of SCFA receptors occurs in both the central and peripheral nervous systems [38], playing a crucial role in maintaining the integrity and functioning of the BBB. Close involvement with SCFAs is crucial for maintaining cerebral homeostasis and preventing the passage of toxins and pathogens into the brain due to BBB’s high selectivity [39,40]. Germ-free mice and mice treated with antibiotics [41,42] exhibit lower levels of various BBB junction proteins, including occludin, zonula occludens-1, and claudin-5 [43]. Certain fibers act as “prebiotics”, enhancing intestinal health and favoring beneficial microorganisms while preventing harmful ones. Additionally, certain types of food impact the microbiota by causing competitive interactions, affecting the development of SCAFs, regulating physiological changes, and enhancing the safeguarding of the mucous layer [44,45]. Inulin, galactose derivatives, fructo-oligosaccharides, gluco-oligosaccharides, -glucans, and lactulose are the most recognized prebiotics [46]. This paper aims to compile knowledge on butyrate to guide future research on determining the optimal levels of this metabolite from polyphenols and dietary fibers for its functional roles.

2. Role of Butyrate in the Intestine

Butyrate, propionate, and acetate are the main SCFAs produced in the gut microbiota. In the human large intestine, their concentration typically falls between 50 and 200 mM. The function of these compounds differs: butyrate serves as an energy source for the intestinal mucosa, acetate is closely involved in lipid synthesis (increasing the concentration of acetyl-Coa), regulating metabolic homeostasis, and propionate aids in gluconeogenesis in the liver [47]. SCFA production relies heavily on undigested carbohydrates, including non-starch polysaccharides, resistant starch, sugars, alcohols, non-digestible oligosaccharides, and proteins [48]. Butyrate, a crucial four-carbon SCFA, is found in varying concentrations in the intestines of mammals depending on their dietary fiber intake [49]. Butyrate is the preferred energy source for colon epithelial cells, but it also plays other important roles, such as modulating homeostasis and promoting cell differentiation and proliferation. It can reduce inflammation and oxidative damage and transform neoplastic cells while inhibiting histone deacetylases and binding to various G-protein-coupled receptors [50,51,52]. Carbohydrate glycolysis results in the formation of butyrate; colon cancer cells have a specific behavior that makes them use glucose instead of SCFAs as an energy source, leading to this protective effect. Due to this, butyrate can effectively carry out its histone modification function by inhibiting histone deacetylase and impacting the cell cycle. In contrast, intestinal epithelial cells utilize SCFAs as a source of energy, preventing butyrate from inhibiting histone deacetylase [53]. Cell cycle arrest caused by butyrate mainly happens in the G1 phase [54]. Moreover, cancer cell proliferation is counteracted by increased cell differentiation and cell cycle inhibition [55]. In addition, butyrate is responsible for activating pro-apoptotic genes like Bax and Bak while inhibiting anti-apoptotic genes such as Bcl-2 [56]. Several studies in the literature have found that butyrate improves the integrity of the intestinal barrier by regulating the expression of proteins in the tight junction [57].
The body activates the inflammatory process as a defense mechanism against harmful insults. However, the body has effective mechanisms to regulate inflammation, using self-limiting and negative feedback processes to stop pro-inflammatory signals and prevent prolonged damage [58]. The expression of pro-inflammatory cytokines, inflammatory enzymes, adhesion molecules, chemokines, and growth factors is regulated by NFκB [59]. Inflammatory bowel disease, autoimmune disease, and cancer can result from chronic inflammation in the intestine [60,61]. Numerous studies have shown that butyrate can inhibit NFkB [62] and activate PPARs [63]. The concentration of butyrate in the system is roughly a thousand times lower than in the intestine. It is crucial to recall that butyrate also functions in the gut–brain axis [64]. The intestine and brain communicate bidirectionally via the vagus nerve, neuroendocrine pathways, and neuroimmune pathways [65,66], which involve the central nervous system, enteric nervous system, and various neurons that connect the brain and intestines [67]. In this context, it is worth noting that butyrate can cross the BBB and activate the vagus nerve, thereby impacting the hypothalamus and influencing appetite and eating behavior [68]. Cholinergic enteric neurons can be increased through the epigenetic effect of butyrate [69].

Butyrate Participates in Maintaining the Integrity of the Blood–Brain Barrier

A hypothesis has been proposed about the close relationship between the gut microbiota and the nervous system in recent decades [5]. Researchers have been fascinated by this connection, leading to an increase in knowledge about this topic [70,71]. Although not fully understood, intestinal microbial populations can produce neurotransmitters like serotonin and GABA, which have been shown to affect the nervous system by crossing the blood–brain barrier [72]. Thus, any approach that can impact the makeup of the gut microbiota, like nutrition, can have both positive and negative effects on brain disorders and significant diseases [73]. The BBB specializes in maintaining cerebral homeostasis, regulating ion concentration, protecting against brain toxins, supporting glial and neuronal activity, and defending against infections [5,74]. The BBB becomes more permeable when the barrier malfunctions, disrupting brain tissue homeostasis. Additionally, a higher permeability of the BBB leads to the entry of toxic molecules and the subsequent inflammatory process, both of which contribute to neurodegeneration [75]. Studies have demonstrated that germ-free mice have a higher blood–brain barrier permeability than mice with a healthy microbiota [76]. The loss of BBB integrity is the main cause of several neurological disorders, including Parkinson’s disease, Alzheimer’s disease, depression, epilepsy, multiple sclerosis, mental and behavioral damage, and autism spectrum disorder [77,78]. Moreover, when bacterial strains were reconstituted in animal microbiota, BBB permeability was maintained [79]. Previous studies have demonstrated the significant role of SCFAs, specifically butyrate, in the central nervous system. This includes preventing neuroinflammation, facilitating microglia maturation, aiding neurodevelopment, influencing neurotransmitters, and promoting neurogenesis [80,81]. To validate these discoveries, manipulating the gut microbiome through fecal microbiota transplantation and probiotic use demonstrated that butyrate is crucial for maintaining BBB integrity [82,83]. A reduction in plasma or fecal levels of butyrate can be considered as a biomarker of several neurological disorders [84] and has been observed in many pathological states, such as stroke, multiple sclerosis, vascular dementia, encephalopathy, and traumatic brain injury [85,86].
The rapid uptake of butyrate from plasma to the brain was observed in experiments. The mechanisms of action responsible for the action of butyrate in the health of the BBB are multiple. It promotes the secretion of mucin, which helps to reduce inflammation and the absorption of lipopolysaccharides. Butyrate also increases the integrity of the BBB by improving the activity of antioxidant systems and increasing the expression of tight junction proteins [87,88]. In the inflammatory process linked to intestinal dysbiosis, various inflammatory mediators cause increased BBB permeability by activating microglia and upregulating the expression of adhesion molecules and chemokine receptors [89]. By acting on the immune system, increased butyrate decreases systemic inflammation and suppresses the production of inflammatory mediators.
Figure 2 shows these properties explained for simplicity on the colonocytes; however, it should be remembered that butyrate also practices them specifically on some cell lines of the nervous system.

3. The Protective Role of Polyphenols and Dietary Fibers

The strong connection between lifestyle and health management is widely acknowledged and extensively documented in the scientific literature [90,91]. Overall, eating habits can be modified and have a significant impact on human physiology, health, and cognitive functions [92,93]. The Healthy Eating Index (HEI) was created to measure how well diets align with American dietary guidelines. Initially, the use of the HEI was primarily for scientific purposes to measure diet quality. However, today, the HEI is used to evaluate the adherence of any food group to key dietary recommendations [94]. Many dietary models have been created to address various health conditions, and one of the most prominent is the Mediterranean diet (MD), which is known for its beneficial effects in preventing chronic diseases. The MD is centered around consuming fresh fruits, vegetables, legumes, fiber, vitamins, unrefined cereals, and extra virgin olive oil, with moderate amounts of fish, dairy products, and ethanol (particularly red wine during meals), and a limited intake of red meat [95,96]. Alongside the MD, another well-known dietary model is the Dietary Approach to Stop Hypertension (DASH), which effectively lowers blood pressure. Typically, a complete and varied diet prevents deficiencies and maintains good health in the absence of other diseases. Polyphenols and dietary fiber, known for their numerous health benefits, are especially crucial among all foods. Food polyphenols, numbering around 10,000 compounds, are classified based on various characteristics, like origin, structure, and function. Polyphenols are classified into flavonoids (flavones, flavanones, flavonols, flavanols, isoflavones, and anthocyanins) and non-flavonoid molecules (stilbene, phenolic acids, tannins, lignans, and hydroxycinnamic acids) based on their structural characteristics [97]. Polyphenols offer numerous health benefits, safeguarding against chronic diseases and influencing physiological processes like enzymatic activity and cellular redox. The donation of electrons/hydrogens by polyphenols and the elimination of radical structures are attributed to the phenolic structure [98]. Additionally, polyphenols can inhibit NF-κB, a transcription factor that plays a role in inflammation, cell survival, and growth. NF-κB is also involved in the development of inflammatory bowel diseases and colorectal cancer while promoting antiproliferation and apoptosis [99]. Additionally, polyphenols regulate the activity of kinases like Akt/protein kinase, tyrosine kinase, and mitogen-activated protein kinase (MAPK) [100]. Polyphenols finally hinder certain pro-inflammatory enzymes, like 5-lipoxygenase and cyclooxygenase, which helps to prevent colorectal cancer [101]. The majority of polyphenols are typically found in their glycosylated forms but can undergo structural changes through esterification reactions [102]. Polyphenols have a low bioavailability due to factors like hepatic metabolic processes, interaction with the food matrix, and the action of the intestinal microbiota. Polyphenols are typically considered xenobiotic after being ingested, resulting in a significant decrease in their bioavailability. In fact, in vitro studies have shown that concentrations of 10 to 100 μM of polyphenols have exhibited anticancer or anti-inflammatory effects. Currently, it is understood that only a small percentage (5–10%) of polyphenols are absorbed in the small intestine, while the majority (90–95%) accumulate in the large intestine and undergo enzymatic processes by the gut microbiota [103]. Low-molecular-weight phenolic metabolites are formed through polyphenolic demolition reactions, which are attributed to the gut microbiota. Scientific evidence strongly suggests that polyphenol metabolites have a positive impact, even after undergoing biotransformation [104]. The bioavailability of polyphenols relies on their intake, size, and the composition of intestinal microbiota [105]. Recent research has demonstrated that the gut microbiota can metabolize polyphenols in food. Polyphenols metabolized by gut bacteria produce representative metabolites such as equol, urolithin, and esperitotin [84]. Multiple studies, conducted in animals and humans, indicate that specific amounts of polyphenols can alter the gut’s microbial makeup, either inhibiting or promoting the growth of certain groups [106]. For example, the consumption of polyphenols in wine has significantly increased the abundance of Bacteroides, Bifidobacterium, Enterococci, Prevotella, and Blautia coccoides-E in the human rectale group. The polyphenolic fraction seems to have both prebiotic and selective antimicrobial effects against intestinal pathogenic bacteria [107,108]. Several factors, such as polyphenol structure, dosage, and bacterial strain, influence how polyphenols affect growth and metabolism [109]. As an illustration, the flavonoid B-ring can easily insert itself between nucleic acid bases, disrupting DNA synthesis and RNA [110]. The interaction between polyphenols and gut bacteria impacts SCFA production, leading to a significant increase. In a controlled crossover study, researchers found that polyphenols from freeze-dried cranberry powder increased available butyrate levels by reducing its elimination with feces [111]. The interaction between polyphenols and microbiota has been shown through the incubation of polyphenols with fecal samples and in vivo due to polyphenolic food supplementation [112,113]. Polyphenols and the microbiota have a mutual interaction, allowing for a crosstalk between the two parties.
Fiber is an edible carbohydrate polymer with three or more monomeric units, bound by glycosidic bonds, and is not digested or absorbed in the human small intestine [114]. Currently, defining dietary fiber accurately is a complex task that involves considering various factors like size, solubility, fermentability, viscosity, composition, source, and more [115]. Dietary fiber can be categorized as soluble or insoluble. The absorption of water by insoluble fiber increases the specific weight and volume of feces, making them softer and promoting intestinal motility [116]. Cellulose is the most prevalent insoluble fiber found in nature, typically found alongside hemicellulose, lignin, and pectin. Water is highly attracted to soluble fiber and causes it to dissolve in a watery solution. Moreover, the majority of soluble dietary fibers ferment in water, causing them to swell and form a gel-like structure [117]. Soluble fibers, like pectins, mucilages, galactomannans, and gums, can be consumed through food or dietary supplements [118]. They are primarily present in plant-based foods and have several beneficial effects [119,120,121], including lowering the risk of gastrointestinal diseases like colorectal cancer and irritable bowel syndrome. Including soluble fiber is crucial for maintaining a balanced and healthy dietary model [122,123]. Soluble fiber also helps with controlling appetite, enhancing insulin sensitivity, and reducing weight. This particular fiber can slow down the breakdown and absorption of energy nutrients like starch and triglycerides. This leads to a lower overall energy intake, including glucose and cholesterol, which reduces the risk of type 2 diabetes, obesity, and metabolic diseases [124,125]. Despite the presence of both soluble and insoluble fibers in most plant-based foods, their consumption is notably low, especially in Western countries. Fortified foods were created to boost fiber intake by adding indigestible carbohydrates, polymers, and oligosaccharides to regular foods [126]. Epidemiological studies have shown that African people, who consume a diet rich in dietary fiber, have a lower incidence of colorectal cancer. Certain fibers have a “prebiotic” effect on the gut microbiota by selectively promoting beneficial bacterial populations [127]. SCFAs are formed through the fermentation of dietary fiber by the gut microbiota, as described previously. As a result, a diverse diet high in fiber promotes a healthier gut microbiota and increased production of SCFAs, thus maintaining intestinal health [128]. Conversely, a diet lacking in fiber but high in proteins and sugars can lead to reduced bacterial diversity in the microbiota, decreased SCFAs, and the development of chronic inflammatory diseases. To prevent infections and microbial invasions, the intestinal epithelium is covered and protected by a well-structured and compact mucus layer. Consuming a high-fiber diet promotes the creation of protective mucus that is stable. Animal model studies have shown that the amount of fiber used was higher than recommended for daily consumption. Instead of the recommended daily fiber intake of 30 g, approximately 100 g/day was consumed [129]. Based on these studies, it is reasonable to think that dietary fiber supplementation is inadequate. Currently, the recommended daily intake of dietary fiber for optimal health benefits is over 50 g [130]. Since it may be challenging to consume these quantities through regular food, it is recommended to consider taking supplements for adequate supplementation [131]. It would be intriguing to assess the outcomes of administering butyrate along with polyphenols or fibers, following the discussion on their impact on SCFA production. Positive and regenerative feedback is likely to lead to an increased availability of butyrate and improved intestinal health.
After describing the polyphenols and the dietary fiber and showing how their intake increases the production of SCFAs, it would be interesting to evaluate the results obtained from the administration of butyrate, which is a main ingredient in several dietary supplements (e.g., Colonzak), with the addition of polyphenols or fibers. It is likely to result in positive and regenerative feedback, responsible for a net increase in available butyrate and for achieving intestinal health. Figure 3 illustrates the effects of polyphenols (panel a) and fibers (panel b) on the gut microbiota.

4. Source of Butyrate

Since a greater production of butyrate determines, as already said, a protective effect on the whole organism, it is fundamental to know the main strategies involved in its production. An initial source of butyrate is undoubtedly nutrition: some foods are rich in this compound, including dairy products such as butter (3 g/100 g), goat’s cheese (1 to 1.8 g/100 g), and whole cow’s milk (0.1 g/100 g). However, it is impossible to consume excessive amounts of these foods in the daily diet in order to avoid the occurrence of other metabolic and cardiovascular disorders [144]. As a result, the safest and most immediate pathway for butyrate production remains the fermentation of appropriate foods by the gut microbiota. The amount of butyrate depends on a variety of factors: the quality and quantity of the microflora present in the colon, the type of substrate taken with feeding, and the time of its intestinal transit [145]. To produce butyrate, intestinal bacteria must have the appropriate enzymes to break down substrates. The majority belong to the phylum of the Firmicutes of the genus Clostridium, and, more in particular, to the family of the Ruminococcaceae and the Lachnospiraceae, such as Roseburia intestinalis, Faecalibacterium prausnitzii, Eubacterium rectale, E. Halli, and E. cylindroides [146]. During life, the microbiota is in a state of dynamic equilibrium: in fact, it changes and is very sensitive to dietary, physiological, and/or environmental changes; its composition varies in each individual according to factors such as the place where one lives, personal history, genetic heritage, lifestyle, feeding, and type of birth [73]. The butyrate molecule is produced by the fermentation of different substrates, such as resistant starches (polysaccharides present in whole grains, starchy foods, bananas, and potato starch) and beta-glucans (oats, barley, and rye) [147]. Finally, intestinal transit time is extremely important for the levels of butyrate: numerous studies have shown that a shorter gut transit time leads to reduced development of intestinal bacterial growth and that an insufficient microbiota slows down the production of butyrate [148].
Another strategy to increase the levels of butyrate in the body is to expand the bacteria that produce butyrate; this compound is produced not only in the human colon but also in non-human environments [149]. Many outdoor environments also have conditions that promote the growth of butyrate-producing bacteria, such as the presence of degradable organic materials within bulk soils, intestines of animal carcasses, or plants [150]. These butyrate-producing bacteria can be easily transported into the house, turning themselves from outdoor to indoor sources. Many Gram-positive anaerobic bacteria, producing butyrate, create endospores that allow for their survival in a dormant condition and aerobic outdoor situation. Subsequently, when they come into contact with the human intestine (indoor), they can trigger the germination of the spores, increasing taxa of the gut microbiota and the levels of butyrate [151]. In a randomized controlled mouse study, Liddicoat et al. [152] have demonstrated that butyrate-producing bacteria present in the soil are transferred by dust from the soil to the gut microbiota of mice, increasing the levels of butyrate in the animals. The increase in butyrate-producing bacteria from outside occurs through open doors and windows, clothing, shoes, and pets. In light of these considerations, we can say that the bacteria present in the air inside the house come mainly from a combination of human/pet activity and the outside air that arrives inside [153].

5. Conclusions

Butyrate is a short-chain fatty acid that mammals produce in their gut through the fermentation of dietary fiber by the microbiota. This molecule is versatile and protects against multiple diseases, including diabetes, intestinal inflammation, obesity, colon cancer, and neurological disorders [154,155]. Additionally, butyrate provides multiple approaches to maintain the BBB’s integrity. This review focused on three important topics: identifying the importance of intestinal microbiota and SCFA production; knowing the various roles of butyrate; and understanding the importance of consuming polyphenols and fiber in the diet. In light of the literature cited, butyrate could be considered as a postbiotic compound, able to regulate the immune system by exploiting its metabolic, anti-inflammatory, antioxidant, and antiproliferative properties; it could be indicated for the treatment of gastrointestinal and extra-intestinal diseases. However, to reach these conclusions, further research is needed: first, it would be essential to determine precisely how the bioavailability of butyrate varies after its production. Subsequently, the precise concentrations of butyrate produced after the ingestion of polyphenols and fibers should be identified to determine the exact amount of these foods to be consumed daily. Finally, clinical studies would be suggested to test the use of butyrate for its postbiotic activities.

Author Contributions

J.M. and R.M.B. have conceptualized the manuscript.; J.M. has written the paper; S.R., S.N., R.M. (Roberta Macrì), F.S., C.M. and F.O., have critically reviewed the paper; C.C., M.G. and V.M. (Vincenzo Musolino) have examined the literature; J.M., R.M. (Rocco Mollace) and V.M. (Vincenzo Mollace) have overseen the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research work was supported by public resources from the Italian Ministry of Research and PONa3 00359.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell. Mol. Life Sci. 2019, 76, 473–493. [Google Scholar] [CrossRef] [PubMed]
  2. Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [PubMed]
  3. Zmora, N.; Suez, J.; Elinav, E. You are what you eat: Diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef]
  4. Gomez de Aguero, M.; Ganal-Vonarburg, S.C.; Fuhrer, T.; Rupp, S.; Uchimura, Y.; Li, H.; Steinert, A.; Heikenwalder, M.; Hapfelmeier, S.; Sauer, U.; et al. The maternal microbiota drives early postnatal innate immune development. Science 2016, 351, 1296–1302. [Google Scholar] [CrossRef] [PubMed]
  5. Maiuolo, J.; Gliozzi, M.; Musolino, V.; Carresi, C.; Scarano, F.; Nucera, S.; Scicchitano, M.; Oppedisano, F.; Bosco, F.; Ruga, S.; et al. The Contribution of Gut Microbiota–Brain Axis in the Development of Brain Disorders. Front. Neurosci. 2021, 15, 616883. [Google Scholar] [CrossRef]
  6. Walker, A.W.; Hoyles, L. Human microbiome myths and Misconceptions. Nat. Microbiol. 2023, 8, 1392–1396. [Google Scholar] [CrossRef]
  7. Daliri, E.B.; Tango, C.N.; Lee, B.H.; Oh, D.H. Human microbiome restoration and safety. Int. J. Med. Microbiol. 2018, 308, 487–497. [Google Scholar] [CrossRef]
  8. Kim, S.; Jazwinski, S.M. The gut microbiota and healthy aging: A mini-review. Gerontology 2018, 64, 513–520. [Google Scholar] [CrossRef]
  9. Davenport, E.R.; Mizrahi-Man, O.; Michelini, K.; Barreiro, L.B.; Ober, C.; Gilad, Y. Seasonal variation in human gut microbiome composition. PLoS ONE 2014, 9, e90731. [Google Scholar] [CrossRef]
  10. Hills, R.D.J.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C.; Theberge, C.R. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019, 11, 1613. [Google Scholar] [CrossRef]
  11. Costea, P.I.; Hildebrand, F.; Arumugam, M.; Bäckhed, F.; Blaser, M.J.; Bushman, F.D.; de Vos, W.M.; Ehrlich, S.D.; Fraser, C.M.; Hattori, M.; et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol. 2018, 3, 8–16. [Google Scholar] [CrossRef] [PubMed]
  12. Haran, J.P.; McCormick, B.A. Aging, Frailty, and the Microbiome-How Dysbiosis Influences Human Aging and Disease. Gastroenterology 2021, 160, 507–523. [Google Scholar] [CrossRef]
  13. Maiuolo, J.; Carresi, C.; Gliozzi, M.; Mollace, R.; Scarano, F.; Scicchitano, M.; Macrì, R.; Nucera, S.; Bosco, F.; Oppedisano, F.; et al. The Contribution of Gut Microbiota and Endothelial Dysfunction inthe Development of Arterial Hypertension in Animal Models and in Humans. Int. J. Mol. Sci. 2022, 23, 3698. [Google Scholar] [CrossRef]
  14. Weiss, G.A.; Hennet, T. Mechanisms and consequences of intestinal dysbiosis. Cell. Mol. Life Sci. 2017, 74, 2959–2977. [Google Scholar] [CrossRef] [PubMed]
  15. Allaire, J.M.; Crowley, S.M.; Law, H.T.; Chang, S.-Y.; Ko, H.-J.; Vallance, B.A. The intestinal epithelium: Central coordinator of mucosal immunity. Trends Immunol. 2018, 39, 677–696. [Google Scholar] [CrossRef]
  16. Cryan, J.F.O.; Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
  17. Patel, R.; DuPont, H.L. New approaches for bacteriotherapy: Prebiotics, new-generation probiotics, and synbiotics. Clin. Infect. Dis. 2015, 60 (Suppl. S2), S108–S121. [Google Scholar] [CrossRef] [PubMed]
  18. Fiorentino, M.; Sapone, A.; Senger, S.; Camhi, S.S.; Kadzielski, S.M.; Buie, T.M.; Kelly, D.L.; Cascella, N.; Fasano, A. Blood-brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol. Autism 2016, 7, 49. [Google Scholar] [CrossRef]
  19. Boyton, R.J.; Altmann, D.M. Muco-obstructive lung diseases. N. Engl. J. Med. 2019, 381, e20. [Google Scholar]
  20. Garg, A.; Sui, P.; Verheyden, J.M.; Young, L.R.; Sun, X. Consider the lung as a sensory organ: A tip from pulmonary neuroendocrine cells. Curr. Top. Dev. Biol. 2019, 132, 67–89. [Google Scholar]
  21. Duncan, S.H.; Holtrop, G.; Lobley, G.E.; Calder, A.G.; Stewart, C.S.; Flint, H.J. Contribution of acetate to butyrate formation by human faecal bacteria. Br. J. Nutr. 2004, 91, 915–923. [Google Scholar] [CrossRef] [PubMed]
  22. Macfarlane, S.; Macfarlane, G.T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 67–72. [Google Scholar] [CrossRef] [PubMed]
  23. Schonfeld, P.; Wojtczak, L. Short- and medium- chain fatty acids in energy metabolism: The cellular perspective. J. Lipid Res. 2016, 57, 943–954. [Google Scholar] [CrossRef]
  24. 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]
  25. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  26. Pozuelo, M.; Panda, S.; Santiago, A.; Mendez, S.; Accarino, A.; Santos, J.; Guarner, F.; Azpiroz, F.; Manichanh, C. Reduction of butyrate- and methane-producing microorganisms in patients with irritable bowel syndrome. Sci. Rep. 2015, 5, 12693. [Google Scholar] [CrossRef]
  27. Vatanen, T.; Franzosa, E.A.; Schwager, R.; Tripathi, S.; Arthur, T.D.; Vehik, K.; Lernmark, A.; Hagopian, W.A.; Rewers, M.J.; She, J.-X.; et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature 2018, 562, 589–594. [Google Scholar] [CrossRef]
  28. Ta, J.; Derrien, M.; Törnblom, H.; Brazeilles, R.; Cools-Portier, S.; Doré, J.; Störsrud, S.; Le Nevé, B.; Öhman, L.; Simrén, M. Identification of an Intestinal Microbiota Signature Associated with Severity of Irritable Bowel Syndrome. Gastroenterology. 2017, 152, 111–123. [Google Scholar]
  29. Endesfelder, D.; Engel, M.; Davis-Richardson, A.G.; Ardissone, A.N.; Achenbach, P.; Hummel, S.; Winkler, C.; Atkinson, M.; Schatz, D.; Triplett, E.; et al. Towards a functional hypothesis relating anti-islet cell autoimmunity to the dietary impact on microbial communities and butyrate production. Microbiome 2016, 4, 17. [Google Scholar] [CrossRef]
  30. Rowart, P.; Wu, J.; Caplan, M.J.; Jouret, F. Implications of AMPK in the Formation of Epithelial Tight Junctions. Int. J. Mol. Sci. 2018, 19, 2040. [Google Scholar] [CrossRef]
  31. Miao, W.; Wu, X.; Wang, K.; Wang, W.; Wang, Y.; Li, Z.; Liu, J.; Li, L.; Peng, L. Sodium Butyrate Promotes Reassembly of Tight Junctions in Caco-2 Monolayers Involving Inhibition of MLCK/MLC2 Pathway and Phosphorylation of PKCβ2. Int. J. Mol. Sci. 2016, 17, 1696. [Google Scholar] [CrossRef] [PubMed]
  32. Gaudier, E.; Rival, M.; Buisine, M.P.; Robineau, I.; Hoebler, C. Butyrate enemas upregulate Muc genes expression but decrease adherent mucus thickness in mice colon. Physiol. Res. 2009, 58, 111–119. [Google Scholar] [CrossRef] [PubMed]
  33. Cuciniello, R.; Di Meo, F.; Filosa, S.; Crispi, S.; Bergamo, P. The Antioxidant Effect of Dietary Bioactives Arises from the Interplay between the Physiology of the Host and the Gut Microbiota: Involvement of Short-Chain Fatty Acids. Antioxidants 2023, 12, 1073. [Google Scholar] [CrossRef] [PubMed]
  34. O’Keefe, S.J. Diet, microorganisms and their metabolites and colon cancer. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 691–706. [Google Scholar] [CrossRef] [PubMed]
  35. Fock, E.; Parnova, R. Mechanisms of Blood-Brain Barrier Protection by Microbiota-Derived Short-Chain Fatty Acids. Cells 2023, 12, 657. [Google Scholar] [CrossRef] [PubMed]
  36. Oldendorf, W.H. Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. Am. J. Physiol. 1973, 224, 1450–1453. [Google Scholar] [CrossRef] [PubMed]
  37. Yadav, S.; Dwivedi, A.; Tripathi, A.; Tripathi, A.K. Therapeutic potential of short-chain fatty acid production by gut microbiota in neurodegenerative disorders. Nutr. Res. 2022, 106, 72–84. [Google Scholar] [CrossRef] [PubMed]
  38. Falomir-Lockhart, L.J.; Cavazzutti, G.F.; Gimenez, E.; Toscani, A.M. Fatty acid signaling mechanisms in neural cells: Fatty acid receptors. Front. Cell. Neurosci. 2019, 13, 162. [Google Scholar] [CrossRef]
  39. Maiuolo, J.; Gliozzi, M.; Musolino, V.; Scicchitano, M.; Carresi, C.; Scarano, F.; Bosco, F.; Nucera, S.; Ruga, S.; Zito, M.C.; et al. The “Frail” Brain Blood Barrier in Neurodegenerative Diseases: Role of Early Disruption of Endothelial Cell-to-Cell Connections. Int. J. Mol. Sci. 2018, 19, 2693. [Google Scholar] [CrossRef]
  40. Maiuolo, J.; Gliozzi, M.; Musolino, V.; Carresi, C.; Nucera, S.; Macrì, R.; Scicchitano, M.; Bosco, F.; Scarano, F.; Ruga, S.; et al. The Role of Endothelial Dysfunction in Peripheral Blood Nerve Barrier: Molecular Mechanisms and Pathophysiological Implications. Int. J. Mol. Sci. 2019, 20, 3022. [Google Scholar] [CrossRef]
  41. Sun, N.; Hu, H.; Wang, F.; Li, L.; Zhu, W.; Shen, Y.; Xiu, J.; Xu, Q. Antibiotic-induced microbiome depletion in adult mice disrupts blood-brain barrier and facilitates brain infiltration of monocytes after bone-marrow transplantation. Brain Behav. Immun. 2020, 92, 102–114. [Google Scholar] [CrossRef] [PubMed]
  42. Wu, Q.; Zhang, Y.; Lai, Q.; Dong, Z.; Kuang, W.; Yang, C.; Su, D.; Li, H.; Zhong, Z. Potential effects of antibiotic-induced gut microbiome alteration on blood-brain barrier permeability compromise in rhesus monkeys. Ann. N. Y. Acad. Sci. 2020, 1470, 14–24. [Google Scholar] [CrossRef]
  43. Lee, H.C.; Jenner, A.M.; Low, C.S.; Lee, Y.K. Effect of tea phenolics and their aromatic fecal bacterial metabolites on intestinal microbiota. Res. Microbiol. 2006, 157, 876–884. [Google Scholar] [CrossRef] [PubMed]
  44. Queipo-Ortuño, M.I.; Boto-Ordóñez, M.; Murri, M.; Gomez-Zumaquero, J.M.; Clemente-Postigo, M.; Estruch, R. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am. J. Clin. Nutr. 2012, 95, 1323–1334. [Google Scholar] [CrossRef]
  45. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [PubMed]
  46. Schroeder, B.O.; Birchenough, G.M.H.; Ståhlman, M.; Arike, L.; Johansson, M.E.V.; Hansson, G.C.; Backhed, F. Bifidobacteria or fiber protects against diet-induced microbiota-mediated colonic mucus deterioration. Cell Host Microbe 2018, 23, 27–40. [Google Scholar] [CrossRef]
  47. Zou, J.; Chassaing, B.; Singh, V.; Pellizzon, M.; Ricci, M.; Fythe, M.D.; Kumar, M.V.; Gewirtz, A.T. Fiber-mediated nourishment of gut microbiota protects against diet-induced obesity by restoring IL-22-mediated colonic health. Cell Host Microbe 2018, 23, 41–53. [Google Scholar] [CrossRef]
  48. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.E.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  49. Morrison, D.J.; Preston, T. Formation of shortchain fatty acids by the gut microbiota and their impacton human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef]
  50. Cummings, J.H.; Englyst, H.N. Fermentation in the human large intestine and the available substrates. Am. J. Clin. Nutr. 1987, 45, 1243–1255. [Google Scholar] [CrossRef]
  51. Ingerslev, A.K.; Theil, P.K.; Hedemann, M.S.; Lærke, H.N.; Bach Knudsen, K.E. Resistant starch and arabinoxylan augment SCFA absorption, but affect postprandial glucose and insulin responses differently. Br. J. Nutr. 2014, 111, 1564–1576. [Google Scholar] [CrossRef] [PubMed]
  52. Clarke, J.M.; Young, G.P.; Topping, D.L.; Bird, A.R.; Cobiac, L.; Scherer, B.L.; Winkler, J.G.; Lockett, T.J. Butyrate delivered by butyrylated starch increases distal colonic epithelial apoptosis in carcinogen-treated rats. Carcinogenesis 2011, 33, 197–202. [Google Scholar] [CrossRef] [PubMed]
  53. Siavoshian, S.; Blottière, H.M.; Le Foll, E.; Kaeffer, B.; Cherbut, C.; Galmiche, J.P. Comparison of the effect of different short chain fatty acids on the growth and differentiation of human colonic carcinoma cell lines in vitro. Cell Biol. Int. 1997, 21, 281–287. [Google Scholar] [CrossRef] [PubMed]
  54. Donohoe, D.R.; Collins, L.B.; Wali, A.; Bigler, R.; Sun, W.; Bultman, S.J. The Warburg Effect Dictates the Mechanism of ButyrateMediated Histone Acetylation and Cell Proliferation. Mol. Cell 2012, 48, 612–626. [Google Scholar] [CrossRef] [PubMed]
  55. Saldanha, S.N.; Kala, R.; Tollefsbol, T.O. Molecular mechanisms for inhibition of colon cancer cells by combined epigeneticmodulating epigallocatechin gallate and sodium butyrate. Exp. Cell Res. 2014, 324, 40–53. [Google Scholar] [CrossRef] [PubMed]
  56. Siavoshian, S.; Segain, J.P.; Kornprobst, M.; Bonnet, C.; Cherbut, C.; Galmiche, J.P.; Blottiere, H. Butyrate and trichostatin a effects on the proliferation/differentiation of human intestinal epithelial cells: Induction of cyclin D3 and p21 expression. Gut 2000, 46, 507–514. [Google Scholar] [CrossRef]
  57. Mandal, M.; Olson, D.J.; Sharma, T.; Vadlamudi, R.K.; Kumar, R. Butyric acid induces apoptosis by up-regulating Bax expression via stimulation of the c-Jun N-terminal kinase/activation protein-1 pathway in human colon cancer cells. Gastroenterology 2001, 120, 71–78. [Google Scholar] [CrossRef] [PubMed]
  58. Salvi, P.S.; Cowles, R.A. Butyrate and the Intestinal Epithelium: Modulation of Proliferation and Inflammation in Homeostasis and Disease. Cells 2021, 10, 1775. [Google Scholar] [CrossRef] [PubMed]
  59. Bours, V.; Bonizzi, G.; Bentires-Alj, M.; Bureau, F.; Piette, J.; Lekeux, P.; Merville, M.P. NF-kB activation in response to toxical and therapeutical agents: Role in inflammation and cancer treatment. Toxicology 2000, 153, 27–38. [Google Scholar] [CrossRef]
  60. 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]
  61. Blander, J.M. Death in the intestinal epithelium—Basic biology and implications for inflammatory bowel disease. FEBS J. 2016, 283, 2720–2730. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, P.; Wang, Y.; Yang, G.; Zhang, Q.; Meng, L.; Xin, Y.; Jiang, X. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol. Res. 2021, 165, 105420. [Google Scholar] [CrossRef] [PubMed]
  63. Alex, S.; Lange, K.; Amolo, T.; Grinstead, J.S.; Haakonsson, A.K.; Szalowska, E.; Koppen, A.; Mudde, K.; Haenen, D.; Al-Lahham, S.; et al. Short chain fatty acids stimulate angiopoietin-like 4 synthesis in human colon adenocarcinoma cells by activating ppar. Mol. Cell. Biol. 2013, 33, 1303–1316. [Google Scholar] [CrossRef] [PubMed]
  64. Stilling, R.M.; van de Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. The neuropharmacology of butyrrate: The bread and butter of the microbiota-gut-brain axis? Neurochem. Int. 2016, 99, 110–132. [Google Scholar] [CrossRef] [PubMed]
  65. Sandhu, K.V.; Sherwin, E.; Schellekens, H.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Feeding the microbiota-gut-brain axis: Diet, microbiome, and neuropsychiatry. Transl. Res. 2017, 179, 223–244. [Google Scholar] [CrossRef] [PubMed]
  66. Ghaisas, S.; Maher, J.; Kanthasamy, A. Gut microbiome in health and disease: Linking the microbiome–gut–brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol. Ther. 2016, 158, 52–62. [Google Scholar] [CrossRef]
  67. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef] [PubMed]
  68. van de Wouw, M.; Schellekens, H.; Dinan, T.G.; Cryan, J.F. Microbiota gut-brain axis: Modulator of host metabolism and appetite. J. Nutr. 2017, 147, 727–745. [Google Scholar] [CrossRef] [PubMed]
  69. Li, Y.; Schoufour, J.; Wang, D.D.; Dhana, K.; Pan, A.; Liu, X.; Song, M.; Liu, G.; Shin, H.J.; Sun, Q.; et al. Healthy lifestyle and life expectancy free of cancer, cardiovascular disease, and type 2 diabetes: Prospective cohort study. BMJ 2020, 368, l6669. [Google Scholar] [CrossRef]
  70. Dinan, T.G.; Cryan, J.F. Brain-Gut-Microbiota Axis and Mental Health. Psychosom. Med. 2017, 9, 920–926. [Google Scholar] [CrossRef]
  71. Kim, N.; Yun, M.; Oh, Y.J.; Choi, H.J. Mind-altering with the gut: Modulation of the gut-brain axis with probiotics. J. Microbiol. 2018, 56, 172–182. [Google Scholar] [CrossRef] [PubMed]
  72. Stilling, R.M.; Dinan, T.G.; Cryan, J.F. Microbial genes, brain & behaviour—Epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014, 13, 69–86. [Google Scholar]
  73. Bourassa, M.W.; Alima, I.; Bultmanc, S.J.; Ratana, R.R. Butyrate, neuroepigenetics and the gut microbiome: Can a high fiber diet improve brain health? Neurosci. Lett. 2016, 625, 56–63. [Google Scholar] [CrossRef] [PubMed]
  74. Kadry, H.; Noorani, B.; Cucullo, L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
  75. Zhao, Y.; Gan, L.; Ren, L.; Lin, Y.; Ma, C.; Lin, X. Factors influencing the blood-brain barrier permeability. Brain Res. 2022, 1788, 147937. [Google Scholar] [CrossRef] [PubMed]
  76. Nguyen, T.D.; Hållenius, F.F.; Lin, X.; Nyman, M.; Prykhodko, O. Monobutyrin and Monovalerin Affect Brain Short-Chain Fatty Acid Profiles and Tight-Junction Protein Expression in ApoE-Knockout Rats Fed High-Fat Diets. Nutrients 2020, 12, 1202. [Google Scholar] [CrossRef]
  77. Varatharaj, A.; Galea, I. The blood-brain barrier in systemic inflammation. Brain Behav. Immun. 2017, 60, 1–12. [Google Scholar] [CrossRef] [PubMed]
  78. Takata, F.; Nakagawa, S.; Matsumoto, J.; Dohgu, S. Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. Front. Cell. Neurosci. 2021, 15, 661838. [Google Scholar] [CrossRef] [PubMed]
  79. Wen, J.; Ding, Y.; Wang, L.; Xiao, Y. Gut microbiome improves postoperative cognitive function by decreasing permeability of the blood-brain barrier in aged mice. Brain Res. Bull. 2020, 164, 249–256. [Google Scholar] [CrossRef]
  80. Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
  81. Ahmed, H.; Leyrolle, Q.; Koistinen, V.; Kärkkäinen, O.; Layé, S.; Delzenne, N.; Hanhineva, K. Microbiota-derived metabolites as drivers of gut–brain communication. Gut Microbes 2022, 14, 2102878. [Google Scholar] [CrossRef] [PubMed]
  82. Li, H.; Sun, J.; Du, J.; Wang, F.; Fang, R.; Yu, C.; Xiong, J.; Chen, W.; Lu, Z.; Liu, J. Clostridium butyricum exerts a neuroprotective effect in a mouse model of traumatic brain injury via the gut-brain axis. Neurogastroenterol. Motil. 2018, 30, e13260. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, J.; Sun, J.; Wang, F.; Yu, X.; Ling, Z.; Li, H.; Zhang, H.; Jin, J.; Chen, W.; Pang, M.; et al. Neuroprotective Effects of Clostridium butyricum against Vascular Dementia in Mice via Metabolic Butyrate. BioMed Res. Int. 2015, 2015, 512946. [Google Scholar] [CrossRef] [PubMed]
  84. Gershon, M.D.; Margolis, K.G. The gut, its microbiome, and the brain: Connections and communications. J. Clin. Investig. 2021, 131, e143768. [Google Scholar] [CrossRef] [PubMed]
  85. Saresella, M.; Marventano, I.; Barone, M.; La Rosa, F.; Piancone, F.; Mendozzi, L.; D’Arma, A.; Rossi, V.; Pugnetti, L.; Roda, G. Alterations in Circulating Fatty Acid Are Associated with Gut Microbiota Dysbiosis and Inflammation in Multiple Sclerosis. Front. Immunol. 2020, 11, 1390. [Google Scholar] [CrossRef] [PubMed]
  86. Stanley, D.; Moore, R.J.; Wong, C.H.Y. An insight into intestinal mucosal microbiota disruption after stroke. Sci. Rep. 2018, 8, 568. [Google Scholar] [CrossRef] [PubMed]
  87. Couto, M.R.; Gonçalves, P.; Magro, F.; Martel, F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol. Res. 2020, 159, 104947. [Google Scholar] [CrossRef] [PubMed]
  88. Silveira, A.K.; Gomes, H.M.; Fröhlich, N.T.; Kessler, F.; Martins, A.; Rodrigues, M.S.; De Oliveira, J.; do Nascimento, N.D.; Sirena, D.; Paz, A.H.; et al. Sodium Butyrate Protects Against Intestinal Oxidative Damage and Neuroinflammation in the Prefrontal Cortex of Ulcerative Colitis Mice Model. Immunol. Investig. 2023, 52, 796–814. [Google Scholar] [CrossRef] [PubMed]
  89. van de Wouw, M.; Boehme, M.; Lyte, J.M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain-gut axis alterations. J Physiol. 2018, 596(20), 4923–4944. [Google Scholar] [CrossRef]
  90. Siddiqui, M.T.; Cresci, G.A.M. The Immunomodulatory Functions of Butyrate. J. Inflamm. Res. 2021, 14, 6025–6041. [Google Scholar] [CrossRef]
  91. Wang, J.; Geng, L. Effects of Socioeconomic Status on Physical and Psychological Health: Lifestyle as a Mediator. Int. J. Environ. Res. Public Health 2019, 16, 281. [Google Scholar] [CrossRef] [PubMed]
  92. Yeung, S.S.Y.; Kwan, M.; Woo, J. Healthy Diet for Healthy Aging. Nutrients 2021, 13, 4310. [Google Scholar] [CrossRef] [PubMed]
  93. Kalache, A.; de Hoogh, A.I.; Howlett, S.E.; Kennedy, B.; Eggersdorfer, M.; Marsman, D.S.; Shao, A.; Griffiths, J.C. Nutrition interventions for healthy ageing across the lifespan: A conference report. Eur. J. Nutr. 2019, 58 (Suppl. S1), 1–11. [Google Scholar] [CrossRef] [PubMed]
  94. Reedy, J.; Lerman, J.L.; Krebs-Smith, S.-M.; Kirkpatrick, S.I.; Pannucci, T.R.E.; Wilson, M.M.; Subar, A.M.; Kahle, L.L.; Tooze, J.A. Evaluation of the Healthy Eating Index-2015. J. Acad. Nutr. Diet. 2018, 118, 1622–1633. [Google Scholar] [CrossRef] [PubMed]
  95. Mazza, E.; Ferro, Y.; Pujia, R.; Mare, R.; Maurotti, S.; Montalcini, T.; Pujia, A. Mediterranean Diet In Healthy Aging. J. Nutr. Health Aging 2021, 25, 1076–1083. [Google Scholar] [CrossRef] [PubMed]
  96. Mentella, M.C.; Scaldaferri, F.; Ricci, C.; Gasbarrini, A.; Miggiano, G.A.D. Cancer and Mediterranean Diet: A Review. Nutrients 2019, 11, 2059. [Google Scholar] [CrossRef] [PubMed]
  97. Willcox, B.J.; Willcox, D.C.; Todoriki, H.; Fujiyoshi, A.; Yano, K.; He, Q.; Curb, J.D.; Suzuki, M. Caloric restriction, the traditional okinawan diet, and healthy aging: The diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann. N. Y. Acad. Sci. 2007, 1114, 434–455. [Google Scholar] [CrossRef] [PubMed]
  98. Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and Human Health: The Role of Bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef] [PubMed]
  99. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef]
  100. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef]
  101. Williams, R.J.; Spencer, J.P.; Rice-Evans, C. Flavonoids: Antioxidants or signalling molecules? Free Radic. Biol. Med. 2004, 36, 838–849. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, D.; Dubois, R.N. Eicosanoids and cancer. Nat. Rev. Cancer 2010, 10, 181–193. [Google Scholar] [CrossRef] [PubMed]
  103. Du, Y.X.; Mamun, A.A.; Lyu, A.P.; Zhang, H.J. Natural Compounds Targeting the Autophagy Pathway in the Treatment of Colorectal Cancer. Int. J. Mol. Sci. 2023, 24, 7310. [Google Scholar] [CrossRef] [PubMed]
  104. Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
  105. Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols: The role of metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef] [PubMed]
  106. Gross, G.; Jacobs, D.M.; Peters, S.; Possemiers, S.; van Duynhoven, J.; Vaughan, E.E. In vitro bioconversion of polyphenols from black tea and red wine/grape juice by human intestinal microbiota displays strong interindividual variability. J. Agric. Food Chem. 2010, 58, 10236–10246. [Google Scholar] [CrossRef] [PubMed]
  107. Zhao, L.; Zhang, T.; Zhang, K. Pharmacological effects of ginseng and ginsenosides on intestinal inflammation and the immune system. Front. Immunol. 2024, 15, 1353614. [Google Scholar] [CrossRef] [PubMed]
  108. Cueva, C.; Gil-Sánchez, I.; Ayuda-Durán, B.; González-Manzano, S.; González-Paramás, A.M.; Santos-Buelga, C.; Bartolomé, B.; Moreno-Arribas, M.V. An Integrated View of the Effects of Wine Polyphenols and Their Relevant Metabolites on Gut and Host Health. Molecules. 2017, 22, 99. [Google Scholar] [CrossRef] [PubMed]
  109. Liu, Z.; Lin, X.; Huang, G.; Zhang, W.; Rao, P.; Ni, L. Prebiotic effects of almonds and almond skins on intestinal microbiota in healthy adult humans. Anaerobe 2014, 26, 1–6. [Google Scholar] [CrossRef]
  110. Oliveira, I.; Sousa, A.; Morais, J.S.; Ferreira, I.C.; Bento, A.; Estevinho, L. Chemical composition, and antioxidant and antimicrobial activities of three hazelnut (Corylus avellana L.) cultivars. Food Chem. Toxicol. 2008, 46, 1801–1807. [Google Scholar] [CrossRef]
  111. Zhao, Y.; Jiang, Q. Roles of the Polyphenol-Gut Microbiota Interaction in Alleviating Colitis and Preventing Colitis-Associated Colorectal Cancer. Adv. Nutr. 2021, 12, 546–565. [Google Scholar] [CrossRef]
  112. Parkar, S.G.; Trower, T.M.; Stevenson, D.E. Fecal microbial metabolism of polyphenols and its effects on human gut microbiota. Anaerobe 2013, 23, 12–19. [Google Scholar] [CrossRef] [PubMed]
  113. Tomas-Barberan, F.; Garcia-Villalba, R.; Quartieri, A.; Raimondi, S.; Amaretti, A.; Leonardi, A.; Rossi, M. In vitro transformation of chlorogenic acid by human gut microbiota. Mol. Nutr. Food Res. 2014, 58, 1122–1131. [Google Scholar] [CrossRef] [PubMed]
  114. Veronese, N.; Solmi, M.; Caruso, M.G.; Giannelli, G.; Osella, A.R.; Evangelou, E.; Maggi, S.; Fontana, L.; Stubbs, B.; Tzoulaki, I. Dietary fiber and health outcomes: An umbrella review of systematic reviews and meta-analyses. Am. J. Clin. Nutr. 2018, 107, 436–444. [Google Scholar] [CrossRef] [PubMed]
  115. Fuller, S.; Beck, E.; Salman, H.; TapselL, L. New Horizons for the Study of Dietary Fiber and Health: A Review. Plant Foods Hum. Nutr. 2016, 71, 1–12. [Google Scholar] [CrossRef] [PubMed]
  116. Baky, M.H.; Salah, M.; Ezzelarab, N.; Shao, P.; Elshahed, M.S.; Farag, M.A. Insoluble dietary fibers: Structure, metabolism, interactions with human microbiome, and role in gut homeostasis. Crit. Rev. Food Sci. Nutr. 2022, 64, 1954–1968. [Google Scholar] [CrossRef] [PubMed]
  117. Hamaker, B.R.; Tuncil, Y.E. A perspective on the complexity of dietary fiber structures and their potential effect on the gut microbiota. J. Mol. Biol. 2014, 426, 3838–3850. [Google Scholar] [CrossRef]
  118. Gill, S.K.; Rossi, M.; Bajka, B.; Whelan, K. Dietary fibre in gastrointestinal health and disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 101–116. [Google Scholar] [CrossRef] [PubMed]
  119. Caminiti, R.; Serra, M.; Nucera, S.; Macrì, R.; Muscoli, C.; Palma, E.; Musolino, V.; Statti, G.; Mollace, V.; Maiuolo, J. Antioxidant Activity and Seasonal Variations in the Composition of Insoluble Fiber from the Cladodes of Opuntia ficus-indica (L.) Miller: Development of New Extraction Procedures to Improve Fiber Yield. Plants 2024, 13, 544. [Google Scholar] [CrossRef]
  120. Maiuolo, J.; Oppedisano, F.; Bosco, F.; Macri, R.; Palma, E.; Muscoli, C.; Mollace, V. Involvement of the Intestinal Microbiota in the Appearance of Multiple Sclerosis: Aloe vera and Citrus bergamia as Potential Candidates for Intestinal Health. Nutrients 2022, 14, 2711. [Google Scholar] [CrossRef]
  121. Maiuolo, J.; Bosco, F.; Guarnieri, L.; Nucera, S.; Ruga, S.; Oppedisano, F.; Tucci, L.; Giuffrè, A.M.; Mollace, V. Protective Role of an Extract Waste Product from Citrus bergamia in an In Vitro Model of Neurodegeneration. Plants 2023, 12, 2126. [Google Scholar] [CrossRef]
  122. Staller, K.; Song, M.; Grodstein, F.; Whitehead, W.E.; Matthews, C.A.; Kuo, B.; Chan, A.T. Increased Long-Term Dietary Fiber Intake Is Associated with a Decreased Risk of Fecal Incontinence in Older Women. Gastroenterology 2018, 155, 661–667. [Google Scholar] [CrossRef] [PubMed]
  123. Burton-Freeman, B. Dietary fiber and energy regulation. J. Nutr. 2000, 130, 272S–275S. [Google Scholar] [CrossRef]
  124. Howarth, N.C.; Saltzman, E.; Roberts, S.B. Dietary fiber and weight regulation. Nutr. Rev. 2001, 59, 129–139. [Google Scholar] [CrossRef]
  125. 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]
  126. Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef] [PubMed]
  127. Deehan, E.C.; Duar, R.M.; Armet, A.M.; Perez-Muñoz, M.E.; Jin, M.; Walter, J. Modulation of the gastrointestinal microbiome with nondigestible fermentable carbohydrates to improve human health. Microbiol. Spectr. 2017, 5, 10–1128. [Google Scholar] [CrossRef]
  128. Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef]
  129. Eaton, S.B.; Eaton, S.B., 3rd; Konner, M.J. Paleolithic nutrition revisited: A twelve-year retrospective on its nature and implications. Eur. J. Clin. Nutr. 1997, 51, 207–216. [Google Scholar] [CrossRef]
  130. O’Keefe, S.J.D. The need to reassess dietary fiber requirements in healthy and critically ill patients. Gastroenterol. Clin. N. Am. 2018, 47, 219–229. [Google Scholar] [CrossRef]
  131. Morris, A. Gut microbiota: Fibre restores healthy gut microbiota. Nat. Rev. Endocrinol. 2018, 14, 63. [Google Scholar] [CrossRef] [PubMed]
  132. Mhd Jalil, A.M.; Combet, E.; Edwards, C.A.; Garcia, A.L. Effect of β-Glucan and Black Tea in a Functional Bread on Short Chain Fatty Acid Production by the Gut Microbiota in a Gut Digestion/Fermentation Model. Int. J. Environ. Res. Public Health 2019, 16, 227. [Google Scholar] [CrossRef] [PubMed]
  133. Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; De Lorenzo, A. Influence of Mediterranean Diet on Human Gut Microbiota. Nutrients 2020, 13, 7. [Google Scholar] [CrossRef] [PubMed]
  134. Wan, M.L.Y.; Co, V.A.; El-Nezami, H. Dietary polyphenol impact on gut health and microbiota. Crit. Rev. Food Sci. Nutr. 2021, 61, 690–711. [Google Scholar] [CrossRef] [PubMed]
  135. Mithul Aravind, S.; Wichienchot, S.; Tsao, R.; Ramakrishnan, S.; Chakkaravarthi, S. Role of dietary polyphenols on gut microbiota, their metabolites and health benefits. Food Res. Int. 2021, 142, 110189. [Google Scholar] [CrossRef] [PubMed]
  136. Katsirma, Z.; Dimidi, E.; Rodriguez-Mateos, A.; Whelan, K. Fruits and their impact on the gut microbiota, gut motility and constipation. Food Funct. 2021, 12, 8850–8866. [Google Scholar] [CrossRef] [PubMed]
  137. Bouyahya, A.; Omari, N.E.; El Hachlafi, N.; Jemly, M.E.; Hakkour, M.; Balahbib, A.; El Menyiy, N.; Bakrim, S.; Naceiri Mrabti, H.; Khouchlaa, A. Chemical Compounds of Berry-Derived Polyphenols and Their Effects on Gut Microbiota, Inflammation, and Cancer. Molecules 2022, 27, 3286. [Google Scholar] [CrossRef] [PubMed]
  138. Gates, E.J.; Bernath, A.K.; Klegeris, A. Modifying the diet and gut microbiota to prevent and manage neurodegenerative diseases. Rev. Neurosci. 2022, 33, 767–787. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, L.; Liu, B.; Ren, L.; Du, H.; Fei, C.; Qian, C.; Li, B.; Zhang, R.; Liu, H.; Li, Z.; et al. High-fiber diet ameliorates gut microbiota, serum metabolism and emotional mood in type 2 diabetes patients. Front. Cell. Infect. Microbiol. 2023, 13, 1069954. [Google Scholar] [CrossRef]
  140. Guan, Z.W.; Yu, E.Z.; Feng, Q. Soluble Dietary Fiber, One of the Most Important Nutrients for the Gut Microbiota. Molecules 2021, 26, 6802. [Google Scholar] [CrossRef]
  141. Tomasello, G.; Mazzola, M.; Leone, A.; Sinagra, E.; Zummo, G.; Farina, F.; Damiani, P.; Cappello, F.; Gerges Geagea, A.; Jurjus, A.; et al. Nutrition, oxidative stress and intestinal dysbiosis: Influence of diet on gut microbiota in inflammatory bowel diseases. Biomed. Pap. Med. Fac. Univ. Palacky. Olomouc Czech Repub. 2016, 160, 461–466. [Google Scholar] [CrossRef] [PubMed]
  142. Paone, P.; Cani, P.D. Mucus barrier, mucins and gut microbiota: The expected slimy partners? Gut 2020, 69, 2232–2243. [Google Scholar] [CrossRef] [PubMed]
  143. Cronin, P.; Joyce, S.A.; O’Toole, P.W.; O’Connor, E.M. Dietary Fibre Modulates the Gut Microbiota. Nutrients 2021, 13, 1655. [Google Scholar] [CrossRef] [PubMed]
  144. Pufulete, M. Intake of dairy products and risk of colorectal neoplasia. Nutr. Res. Rev. 2008, 21, 56–67. [Google Scholar] [CrossRef] [PubMed]
  145. Wong, J.M.; De Souza, R.D.; Kendall, W.C.; Emam, M.S.; Jenkins, J.A. Colonic Health: Fermentation and Short Chain Fatty Acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  146. Lewis, J.D.; Abreu, M.T. Diet as a Trigger or Therapy for Inflammatory Bowel Diseases. Gastroenterology 2017, 152, 398–414.e6. [Google Scholar] [CrossRef] [PubMed]
  147. Deehan, E.C.; Yang, C.; Perez-Muñoz, M.E.; Nguyen, N.K.; Cheng, C.C.; Triador, L.; Zhang, Z.; Bakal, J.A.; Walter, J. Precision Microbiome Modulation with Discrete Dietary Fiber Structures Directs Short-Chain Fatty Acid Production. Cell Host Microbe 2020, 27, 389–404.e6. [Google Scholar] [CrossRef] [PubMed]
  148. Roland, B.C.; Ciarleglio, M.M.; Clarke, J.O.; Semler, J.R.; Tomakin, E.; Mullin, G.E.; Pasricha, P.J. Small Intestinal Transit Time Is Delayed in Small Intestinal Bacterial Overgrowth. J. Clin. Gastroenterol. 2015, 49, 571–576. [Google Scholar] [CrossRef]
  149. Levine, U.Y.; Looft, T.; Allen, H.K.; Stanton, T.B. Butyrate-producing bacteria, including mucin degraders, from the swine intestinal tract. Appl. Environ. Microbiol. 2013, 79, 3879–3881. [Google Scholar] [CrossRef]
  150. Brame, J.E.; Liddicoat, C.; Abbott, C.A.; Breed, M.F. The potential of outdoor environments to supply beneficial butyrate-producing bacteria to humans. Sci. Total Environ. 2021, 777, 146063. [Google Scholar] [CrossRef] [PubMed]
  151. Kearney, S.M.; Gibbons, S.M.; Poyet, M.; Gurry, T.; Bullock, K.; Allegretti, J.R.; Clish, C.B.; Alm, E.J. Endospores and other lysis-resistant bacteria comprise a widely shared core community within the human microbiota. ISME J. 2018, 12, 2403–2416. [Google Scholar] [CrossRef] [PubMed]
  152. Liddicoat, C.; Sydnor, H.; Cando-Dumancela, C.; Dresken, R.; Liu, J.; Gellie, N.J.C. Naturally-diverse airborne environmental microbial exposures modulate the gut microbiome and may provide anxiolytic benefits in mice. Sci. Total Environ. 2020, 701, 134684. [Google Scholar] [CrossRef] [PubMed]
  153. Mhuireach, G.Á.; Betancourt-Román, C.M.; Green, J.L.; Johnson, B.R. Spatiotemporal controls on the urban Aerobiome. Front. Ecol. Evol. 2019, 7, 43. [Google Scholar] [CrossRef]
  154. Jalil, A.M.; Edwards, C.A.; Combet, E.; Ibrahim, M.; Garcia, A.L. Combined effects of added beta glucan and black tea in breads on starch functionality. Int. J. Food Sci. Nutr. 2015, 66, 159–165. [Google Scholar] [CrossRef]
  155. Chai, Y.; Wang, M.; Zhang, G. Interaction between amylose and tea polyphenols modulates the postprandial glycemic response to high-amylose maize starch. J. Agric. Food Chem. 2013, 61, 8608–8615. [Google Scholar] [CrossRef]
Figure 1. The fate of SCFAs. Following dietary intake of fiber, the digestive process begins in the intestine, (1)where the microbiota produces SCFAs. (2) Colonic cells quickly absorb these compounds and convert them into CO2 and ATP for energy. (3) The unmetabolized SCFA portion reaches the liver, (4) providing energy to hepatocytes and aiding in the synthesis of glucose, cholesterol, and fatty acids. (5) Finally, only a very small part of SCFAs reaches systemic circulation.
Figure 1. The fate of SCFAs. Following dietary intake of fiber, the digestive process begins in the intestine, (1)where the microbiota produces SCFAs. (2) Colonic cells quickly absorb these compounds and convert them into CO2 and ATP for energy. (3) The unmetabolized SCFA portion reaches the liver, (4) providing energy to hepatocytes and aiding in the synthesis of glucose, cholesterol, and fatty acids. (5) Finally, only a very small part of SCFAs reaches systemic circulation.
Ijms 25 06971 g001
Figure 2. Main functions performed by butyrate on colonocytes. 1. Cellular energy; 2. apoptosis induction; 3. anti-inflammatory effect; 4. histone modification; 5. cellular cycle arrest; 6. maintaining the integrity of BBB; 7. antioxidant effect.
Figure 2. Main functions performed by butyrate on colonocytes. 1. Cellular energy; 2. apoptosis induction; 3. anti-inflammatory effect; 4. histone modification; 5. cellular cycle arrest; 6. maintaining the integrity of BBB; 7. antioxidant effect.
Ijms 25 06971 g002
Figure 3. Panel (a) shows the main effects of polyphenols on the gut microbiota [132,133,134,135,136,137,138], while panel (b) highlights those exerted by fibers [139,140,141,142,143].
Figure 3. Panel (a) shows the main effects of polyphenols on the gut microbiota [132,133,134,135,136,137,138], while panel (b) highlights those exerted by fibers [139,140,141,142,143].
Ijms 25 06971 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Maiuolo, J.; Bulotta, R.M.; Ruga, S.; Nucera, S.; Macrì, R.; Scarano, F.; Oppedisano, F.; Carresi, C.; Gliozzi, M.; Musolino, V.; et al. The Postbiotic Properties of Butyrate in the Modulation of the Gut Microbiota: The Potential of Its Combination with Polyphenols and Dietary Fibers. Int. J. Mol. Sci. 2024, 25, 6971. https://doi.org/10.3390/ijms25136971

AMA Style

Maiuolo J, Bulotta RM, Ruga S, Nucera S, Macrì R, Scarano F, Oppedisano F, Carresi C, Gliozzi M, Musolino V, et al. The Postbiotic Properties of Butyrate in the Modulation of the Gut Microbiota: The Potential of Its Combination with Polyphenols and Dietary Fibers. International Journal of Molecular Sciences. 2024; 25(13):6971. https://doi.org/10.3390/ijms25136971

Chicago/Turabian Style

Maiuolo, Jessica, Rosa Maria Bulotta, Stefano Ruga, Saverio Nucera, Roberta Macrì, Federica Scarano, Francesca Oppedisano, Cristina Carresi, Micaela Gliozzi, Vincenzo Musolino, and et al. 2024. "The Postbiotic Properties of Butyrate in the Modulation of the Gut Microbiota: The Potential of Its Combination with Polyphenols and Dietary Fibers" International Journal of Molecular Sciences 25, no. 13: 6971. https://doi.org/10.3390/ijms25136971

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