Published on
December 20, 2023

Insights into Short Chain Fatty Acids and Gut Microbiota

Short Chain Fatty Acids

Insights into Short Chain Fatty Acids and Gut Microbiota

The interplay of short-chain fatty acids and the microbiota in the gut is of great importance in the field of personalized nutrition and gut health.

  • Short chain fatty acids (SCFAs) are important for our digestive system.
  • SCFAs come from dietary fiber.
  • They have various roles in maintaining health.
  • They influence microbiome diversity.
  • They affect how our body responds to food.

Main Findings

  • Short-chain fatty acids (SCFAs) are crucial for gut health, produced by the microbiota from dietary fiber.
  • SCFA production and metabolism vary significantly between individuals due to genetic and environmental factors.
  • The relationship between dietary fiber, SCFAs, and gut microbiota influences various health outcomes.
  • Different gut microbiota compositions, or enterotypes, affect the body’s response to dietary fiber and SCFA production.
  • Genetic variations (genotypes) influence how the gut microbiota interacts with dietary components and metabolizes SCFAs.
  • SCFAs play a role in immune regulation, with implications for chronic diseases like obesity, diabetes, and heart disease.

The Basics of Microbiota and SCFAs

The Impact of Short-Chain Fatty Acids on Human Health

In the intricate world of our bodies, there’s a hidden community of microbes known as the microbiota, including bacteria, fungi, and more 1.

This microbiota, residing mainly in our colon, collaborates with us in ways we’re just beginning to understand, akin to an ancient partnership.

This fascinating synergy is the foundation of the holobiont theory 2.

Recent research has unveiled the microbiota’s pivotal role in regulating metabolic, immune, and endocrine functions, while also shaping our response to diseases 3.

Imbalances in the gut microbiota have been linked to various chronic health issues, including obesity 4, cardiovascular disease 5, diabetes 6 7, autoimmune disorders 8 9, and even neurodegenerative diseases 10 11 like Parkinson’s 12 and Alzheimer’s 13.

Dietary habits have emerged as a powerful influencer of our gut health and overall well-being 14 15 16 17 18 19 20 21.

The decline in dietary fiber (DF) consumption, for example, has raised concerns.

DF, found in foods like fruits, vegetables, and grains, serves as a bridge between our gut microbiota and our health 22 23 24 25 26 27 28.

It’s a unique substance that our digestive enzymes can’t fully break down but can be utilized by our gut microbes.

They turn DF into short-chain fatty acids (SCFA), such as butyrate, acetate, and propionate 29.

SCFA, particularly butyrate, plays a crucial role in nourishing the cells lining our colon and supporting our immune system 30 31 32 33 34 35.

Additionally, SCFA act as powerful immune regulators, affecting different tissues and systems throughout our body 36 37 38.

The importance of SCFA in maintaining our health cannot be overstated 39 40 41 42 43 44.

But it’s not just our diet that influences our gut microbiota.

Various factors, including genetics, age, medication, and exposure to pathogens, also play significant roles 45 46 47 48 49 50.

Genetic variations, like single nucleotide polymorphisms (SNPs), can impact how our body responds to DF intake and SCFA production 51 52 53 54 55.

Furthermore, genes associated with SCFA receptors and transporters, as well as genes involved in mucus production and antioxidant defense mechanisms, can have far-reaching effects on our health outcomes 56 57.

Short Chain Fatty Acids and Gut Microbiota

Understanding Dietary Fiber (DF)

DF refers to the indigestible carbohydrates found in foods like fruits, vegetables, grains, and legumes 58 59.

It’s the stuff that passes through our digestive system relatively unchanged.

The recommended daily intake for DF varies but generally falls between 25-35 grams for adults, depending on the country and dietary guidelines 60.

Some health-conscious voices, like the Physicians Committee for Responsible Medicine (PCRM), even advocate for a higher intake of around 40 grams per day 61.

DF plays a critical role in our digestive health.

It’s associated with improved bowel function, including regularity and reduced constipation 62.

More importantly, it’s linked to a lower risk of non-communicable diseases (NCDs), such as cardiovascular disease, type 2 diabetes, and colorectal cancer 63.

DF and Precision Nutrition

Interestingly, where you get your DF matters.

The source of DF in your diet influences the composition of your gut microbiota.

For instance, a low-fat diet and a low-carbohydrate diet can lead to different types of DF intake 64.

The former may emphasize whole grains and fruits, while the latter might rely more on vegetables and plant protein sources.

These dietary adaptations can impact your gut microbiota and potentially shape your health outcomes.

Defining Dietary Fiber

DF is a diverse group of compounds, but there’s consensus on its definition 65 66.

It includes carbohydrate polymers with three or more monomeric units that human enzymes can’t digest.

This definition encompasses various types of DF, from insoluble fibers that add bulk to the stool to soluble fibers that serve as fermentation substrates for our gut microbes.

Beyond Carbohydrates

Interestingly, DF isn’t limited to carbohydrates alone.

It can also include non-carbohydrate compounds like lignin and polyphenols.

These compounds, along with resistant proteins, can be substrates for beneficial gut bacteria, leading to the production of metabolites like SCFA.

The Complex World of Fiber Fermentation

Fiber fermentation is a complex process influenced by the chemical and physical structure of DF, as well as the makeup of our gut microbiota.

Some DF types, like oligosaccharides, resistant starches, and pectins, are more likely to contribute to SCFA production.

Others, like lignin and cellulose, tend to pass through our system largely unchanged.

Recent research has spotlighted the significance of Short Chain Fatty Acids (SCFA) in the context of dietary fiber (DF), gut microbiota (GM), and probiotics.

SCFA serve as essential messengers, bridging the communication between the gut microbiota and the host.

This article dives into the multifaceted implications of SCFA metabolism for human health, while staying true to the original citation style 67.

SCFA: The Communicators Between GM and Host

SCFA, characterized as volatile fatty acids with fewer than six carbon atoms, can take straight or branched-chain forms.

The primary SCFA—acetate (C2), propionate (C3), and butyrate (C4)—contribute to the majority (90–95%) of total SCFA produced by the gut microbiota, primarily through carbohydrate fermentation 68 69.

Recently, it has been unveiled that caproate and valerate, once considered dietary components, also stem from the gut microbiota and are notably increased in individuals with severe obesity 70.

Branched-chain SCFA (BCFA), including isobutyrate, isovalerate, and 2-methylbutanoate, make up a smaller portion (up to 5%) of total SCFA production, originating from the metabolism of specific amino acids 71 72.

Interestingly, BCFA levels in fecal samples exhibit an inverse connection with fiber intake, particularly insoluble fiber.

Elevated BCFA levels have been associated with depression and other psychiatric conditions, potentially mediated through vagal afferent nerve signaling.

Additionally, BCFA, especially isobutyrate, have been linked to unfavorable serum lipid profiles in subjects with hypercholesterolemia 73.

Such higher BCFA levels may correspond to increased protein consumption and reduced dietary fiber intake, factors known to contribute to negative health outcomes and age-related complications 74.

DF Fermentation Products: Post-biotics

Recent nomenclature labels the byproducts of DF fermentation as “post-biotics.”

In adults, the primary products of DF fermentation include SCFA and certain gases like CO2, CH4, and H2, which can be absorbed by the host or excreted 75.

SCFA production in the colon coincides with the consumption of ammonia, hydrogen sulfide (H2S), and BCFA by gut bacteria in the synthesis of microbial cell components.

Consequently, the reduction of these metabolites may partly account for the health benefits attributed to SCFA 76.

Notably, ammonia has been associated with negative health outcomes, including neurotoxicity and hepatotoxicity, as well as increased intestinal permeability, loss of tight junction proteins, and elevated pro-inflammatory cytokines, as observed in animal studies 77.

H2S, hydrogen disulfide, when abnormally produced, has been linked to neurological, cardiovascular, and metabolic diseases 78.

How Individual Differences in Gut Microbiota

Dietary fiber (DF) impacts people differently, but understanding this variation is challenging.

Various factors, including genetics and food matrix properties, contribute to the diverse responses observed in nutritional studies 7980.

Bioavailability of DF-derived metabolites, like vitamins, is influenced by the SLAMENGHI factors (molecular species, linkage, amount, matrix, absorption effectors, nutrition status, genetics, and host-related factors) 81.

Inconsistencies also arise from differing DF definitions across studies.

To address this complexity, a systems analysis approach is recommended, similar to clinical oncology 82.

It’s crucial to recognize the baseline variation in the effects of DF consumption on health outcomes, related to individual gut microbiota (enterotype) 8384.

The impact of DF on metabolism and health outcomes varies based on one’s unique gut microbiota composition 85.

While DF is generally beneficial, it may lead to discomfort in those with low prior DF intake.

The following sections delve into three layers of inter-individual variability (enterotypes, genotypes, and phenotypes) among healthy adults 8687.

Gut Microbiota and Factors Influencing It

Gut microbiota composition is shaped by numerous factors.

Host-related aspects like age, genetics, immune status, and medication use, along with environmental elements such as geography, diet, and pollutants, influence the gut microbiota 888990.

Microbial factors, including substrate competition, metabolic collaboration, and species interactions, also play a pivotal role, as do the microbial environment’s local pH, redox potential, and quorum sensing.

The gut microbiota mainly comprises Firmicutes, Bacteroidetes, Proteobacteria, Verrucobacteria, Actinobacteria, and Fusobacteria, with Bacteroidetes and Firmicutes making up 70% of the total composition in adulthood 91.

Early in life, Proteobacteria and Actinobacteria dominate, influenced by delivery and feeding methods in infancy.

Approximately 6.6% of microbial taxa are heritable, while around 48.6% are influenced by cohabitation 92.

Specific bacteria are involved in the production of short-chain fatty acids (SCFA) through dietary fiber fermentation.

SCFA impacts gut microbiota and health outcomes 93.

Glycans and GOS have been shown to increase Bifidobacterium and Lactobacillus abundance, as well as faecal butyrate concentration 94.

Bacteroidetes mainly produce acetate and propionate, while Firmicutes tend to generate butyrate, which may influence the host’s inflammatory state.

Enterotypes, which represent compositional gradients rather than discrete clusters, have been proposed, highlighting distinct genus-level compositions 95.

Prevotella, a key player in one enterotype, is associated with various health outcomes, including inflammation and colitis 96.

However, its role in diseases like autism, rheumatoid arthritis, and HIV remains inconclusive 97.

Akkermansia muciniphila, a mucin-degrading bacterium, contributes to mucus layer regulation 98.

Akkermansia’s abundance decreases with age and has been associated with various health conditions, such as type 1 diabetes 99.

Dysbiosis, an imbalanced gut microbiota, can develop from an early age and may lead to inflammatory or hypotrophic dysbiosis 100.

Hypotrophic dysbiosis can predispose individuals to inflammatory states and diseases like Giardia lamblia infection-induced diarrhea 101.

Genes and Gut Microbiota

The relationship between the human genome and the microbiome is intricate and ever-changing 102.

While the composition of the gut microbiome can shift during a lifetime, influenced by factors like diet, lifestyle choices, and location 103104, the human genome remains relatively stable, with potential epigenetic changes due to environmental exposure 105106107.

In the realm of nutrigenetics, the focus is on how an individual’s genetic makeup, including copy number variations (CNVs) and single nucleotide polymorphisms (SNPs), can impact the fate of different foods in terms of absorption, distribution, metabolism, or excretion (ADME) patterns (Table 3).

Recent discoveries have identified CNVs and SNPs in various regions of the genome as drivers of differences in individuals’ physical traits and health outcomes.

Polygenic risk scores, which combine multiple genetic factors, have been linked to a wide range of human conditions 108109.

When we consider homeostasis at the holobiont level, where the host genome and microbiome interact 110111, polygenic risk becomes particularly relevant 112.

Twin studies have suggested that specific SNPs can influence microbiota colonization from the earliest days of life, acting as a foundation for achieving optimal homeostasis in an ever-changing environment 113114.

Certain SNPs, such as those related to G-protein coupled cellular receptors (GPCR41, GPCR43, GPCR109A), transporter genes like MCT and SMCT, effector genes like MUC2 (involved in mucus layer production), and regulatory genes like NRF2 (which regulates antioxidant defense proteins), may individually or collectively predispose individuals to various health outcomes.

Exploring elderly populations, researchers have identified genes associated with inflammatory responses (IL-6, IL-10, and the IL-1 cluster), genes involved in the insulin/IGF1 pathway, and genes related to oxidative stress management (PON1) as correlated with extreme old age 115.

While bacterial colonization in early life is crucial for various aspects of immune development, the host’s genetic background can modulate the impact of bacteria.

For example, certain genetic modifications, like programmed cell death (PD1) knockout in mice, can lead to changes in microbiota profiles, affecting the abundance of specific bacterial genera 116.

The secretion of immunoglobulin A (IgA) is crucial for the colonization of beneficial bacteria and defense against pathogens.

This highlights the intricate interplay between the host genome and the microbiome, where commensal bacteria require IgA coating for colonization, while the same coating triggers immune responses against pathogenic bacteria 117118.

Microbial acetate in the gut even regulates IgA reactivity to commensal bacteria, influencing microbiota species and colonization in the colon 119.

Antimicrobial peptides (AMPs) produced by intraepithelial cells (IEC) in the gut are another piece of the puzzle.

These peptides, like α-defensins and β-defensin 1, are rapidly activated by IL-22 and IL-17 to inactivate invading microorganisms.

AMPs not only help maintain the separation between the host and microorganisms but also affect microbial composition 120.

Studies with mice deficient in genes like MYD88, NOD2, or those transgenic for α-defensin 5 have shown altered microbiota compositions 121122123124.

When examining Alzheimer’s disease, the APOε4 allele has been linked to higher levels of pro-inflammatory bacteria, while the protective APOε2 allele correlates with SCFA-producing bacteria 125.

SCFAs have even been found to inhibit the aggregation of amyloid β (Aβ), a hallmark of Alzheimer’s disease, in vitro.

Mutations in MEFV, associated with familial Mediterranean fever, have also demonstrated the host’s ability to modulate microbial composition.

This suggests that the host genome and microbiome structure are functionally linked, impacting responses to dietary fiber both directly (via SCFA metabolism) and indirectly (via microbiome modulation) 126.

Genetic Symphony of Short Chain Fatty Acids and Gut Microbiota

In the intricate world of nutrigenomics, where nutrients orchestrate gene expression, Short Chain Fatty Acids (SCFAs) take center stage.

SCFAs can directly influence genetic expression by modulating histone deacetylases 127 128 129 130.

Over time, these epigenetic changes can shape an individual’s observable characteristics, known as their phenotype.

Phenotypes are often measured through various parameters, like age, body mass index (BMI), or blood counts, and their interpretation can vary depending on the type of study, be it epidemiological, clinical, or mechanistic 131132133.

This variability is influenced by genetic predisposition and the individual’s ability to maintain balance in the face of environmental stressors 134135136.

It’s essential to understand that these associations at the phenotype level, such as the link between BMI and metabolic issues or glycated hemoglobin levels and diabetes progression, are influenced by a complex interplay of genetics, microbiome, and diet.

Some genotypes may lead to increased inflammatory function, which is beneficial in defending against infections.

However, when a low-grade chronic inflammation becomes undesirable, as in Non-Communicable Diseases (NCDs) and aging, these genotypes can have detrimental effects 137138139.

Even genes unrelated to immune function can play a crucial role in achieving holobiont homeostasis, with implications for NCDs and aging progression, as seen with genes like FUT-2 and AMY1 140141.

Centenarians, individuals who live beyond 100 years without significant chronic diseases, offer intriguing insights.

Studies suggest that centenarians have distinctive gut microbiome signatures compared to both young adults (20-40 years old) and the general elderly population (60-80 years old) 142143144.

These variations could be attributed to specific gene polymorphisms.

For instance, the FUT2 gene, responsible for regulating glycans in mucosal tissues, has links to conditions like Crohn’s disease 145.

Enterotyping has shown that the secretor phenotype, associated with Firmicutes, Akkermansia, and Ruminococcus spp., may be influenced by certain genes 146.

This underscores the idea that genes not directly linked to immune function can significantly impact health outcomes, including NCDs and survival into old age 147.

In mice, genotype plays a recognized role in microbial colonization 148.

The timing of meals, known as chrononutrition, can also lead to significant changes in gut microbiome composition, immune responses, and metabolic conditions, including Type 2 Diabetes (T2D), Cardiovascular Disease (CVD), and psychological well-being 149150151152.

Recent research highlights the interplay between diet, genetics, and gut microbiota in conditions like neurodegeneration, cancer, menopause symptoms, and Non-Alcoholic Fatty Liver Disease (NAFLD), all characterized by chronic inflammation 153154155156157.

Studies have shown that microbiota heritability exists in humans, particularly in twins 158159.

This suggests that the host can benefit from the intricate interactions between their genetic makeup and microbiome composition, influenced by factors like dietary patterns and environmental exposures 160.

However, there’s a degree of variability in SCFA production among individuals.

For example, while acetate is typically the most abundant SCFA in feces, some individuals may exhibit substantial differences in propionate and butyrate levels.

This variability is an intriguing aspect of the SCFA story 161.

How Our Bodies and Gut Bacteria Work Together

Host-Microbe Interface: Where It All Begins

Imagine your gut as a dynamic landscape.

The stomach’s quick passage time and low acidity initially keep bacterial numbers low.

But as you progress into the small intestine, the scene changes 162 163.

Bacterial populations increase substantially, setting the stage for the grand performance in the colon, where anaerobic bacteria thrive 164 165.

One significant factor shaping this drama is pH levels 166.

The rise in pH from gastric acid neutralization and pancreatic secretions in the small intestine opens doors for specific bacteria.

The type of food you eat also matters, as it influences the fermentation products produced in your gut.

Diverse Diet, Diverse Microbiota

The availability of dietary fiber (DF) depends on what you eat.

DF serves as a substrate 167 168, and its composition influences the fermentation products generated.

In the colon, DF promotes the growth of diverse microbial species that engage in “substrate cross-feeding” 169 170 and “metabolic cross-feeding” 171.

These interactions enhance microbial diversity and create a harmonious environment in your colon.

One remarkable collaboration is between Faecalibacterium prausnitzii (F. prausnitzii) and Bacteroides thetaiotaomicron (B. thetaiotaomicron).

F. prausnitzii derives butyrate from acetate produced by B. thetaiotaomicron 172.

Butyrate, in turn, boosts intestinal barrier function and mucin production, fortifying your gut’s defenses 173 174.

SCFA: Heroes of the Proximal Colon

Short Chain Fatty Acids (SCFA) are the stars of the show in the proximal colon 175.

Acetate, propionate, and butyrate are the primary SCFA, absorbed by colonocytes 176 and entering the bloodstream.

Their absorption and impact on crypt proliferation depend on luminal pH levels.

Butyrate, in particular, is a key player.

It serves as a major energy source for colonocytes, influencing their proliferation, differentiation, and mucus production 177.

Butyrate’s protective role against colorectal carcinoma is well-documented, making it a vital component in gut health 178 179.

Digestive Enzymes

Our genes also influence our gut microbiota.

For instance, the AMY1 gene, which encodes salivary α-amylase, affects the oral and gut microbiota composition 180.

Individuals with low AMY1 copy numbers digest carbohydrates differently from those with high copy numbers, impacting their microbiomes and SCFA production.

Variants in the LCT gene 181, responsible for lactase production, are correlated with Bifidobacterium abundance in the gut.

Lactase persistence can affect dairy consumption and medication responses, making genetics an essential factor in our gut’s story.

Genetic Diversity and Barriers

The gut microbiota has a role in regulating our physical barriers, such as mucin production 182.

Mucin is a vital component of the mucus layer separating enterocytes and microbiota.

Polymorphisms in mucin genes are associated with gastric cancer risk, emphasizing the interplay between genetics and gut health 183.

Maintaining Barrier Integrity

Short Chain Fatty Acids (SCFA) play a crucial role in maintaining the integrity of our gut barriers.

They upregulate tight junction proteins 184, ensuring the gut remains impermeable.

This is especially important in preventing pathogen invasion, inflammation, and diseases like obesity and insulin resistance 185.

Immune Responses and Gut Health

Butyrate influences immune cell populations in the gut.

It promotes the differentiation of intestinal macrophages into tolerant macrophages, reducing pro-inflammatory responses 186.

Natural killer (NK) cell differentiation 187, essential for an impermeable intestinal barrier 188, is also influenced by SCFA 189 190 191].

Antibiotic exposure in early life 192 can predict the risk of conditions like asthma.

Conversely, early exposure to diverse microorganisms may reduce the risk of asthma, allergies, and other disorders 193 194.

Prebiotics, such as GOS, increase beneficial bacteria, boost immune responses, and reduce inflammation 195.

Transporter Genetics: The SCFA Highway

Monocarboxylate transporters (MCTs) 196 197 are the highway for SCFA uptake by colonocytes.

Fourteen transporters, including MCT1 and MCT4, are responsible for SCFA transportation.

These transporters are influenced by genetic variations and play crucial roles in energy supply to different tissues.

MCTs are pleiotropic, meaning they have various downstream effects in different tissues 198 199.

For example, MCT2 is upregulated in neurons following food deprivation and ischemia recovery 200, while MCT4 is reduced in response to fasting 201202.

These transporters also impact drug delivery, making them essential in therapeutic strategies.

SCFA Metabolism

SCFA are known to contribute significantly to human energy needs, comprising approximately 10% of our energy requirements203.

Among the various SCFA, butyrate stands out as the primary energy source for colonocytes, while propionate can be partially converted to glucose in the liver204205.

The GM plays a pivotal role in producing SCFA, generating an estimated 500–600 mmol/d of SCFA daily.

Of this, approximately 60% is acetate, 20% is propionate, and the remaining 20% is butyrate 206.

These numbers translate to around 37 mmol/kg body weight of acetate, 13 mmol/kg body weight of propionate, and 12.4 mmol/kg body weight of butyrate207.

In the human descending colon, SCFA concentration can reach 69–91 mmol/kg luminal content, with acetate accounting for 60–75% of total fecal SCFA.

One notable player in this complex interplay is Methanobrevibacter smithii, found in 70% of humans, and a significant methane producer in the GM.

Studies suggest that methane production is linked to higher BMI scores in obesity, as well as conditions like constipation and antidepressant use208.

Additionally, in the presence of methane, propionate can be elongated to produce valerate209.

Although less is known about valerate and caproate, they exist in small amounts in the gut and may impact health over time by altering the balance of acetate, butyrate, and propionate reaching human cells 210.

Colon: A Hub of SCFA Activity

The colon plays a pivotal role in SCFA utilization, with more than 95% of the SCFA produced by the GM being absorbed here.

Butyrate, in particular, takes the spotlight as it accounts for over 70% of colonocyte energy production211212213.

Butyrate and propionate have been observed to increase cellular proliferation rates, while acetate does not share this effect214215216217.

Butyrate’s significance extends to enterocyte differentiation, primarily driven by factors like FoxO3 and hypoxia-inducible factor α (HIF-α).

FoxO3 is associated with cellular homeostasis and longevity, suggesting a role for SCFA in epigenetic modulation and potential links to cancer development and tissue healing 218.

Butyrate also plays a role in inhibiting DNA-damaged cell proliferation via p53.

In intestinal crypts, a diffusion gradient allows butyrate to be metabolized by mature enterocytes, which, in turn, enhances intestinal permeability and immune function.

This process results in a reduction in intestinal permeability, an increase in TGB-β production promoting Treg differentiation, and a shift towards a more tolerogenic immune profile with increased levels of circulating IL-10219.

Furthermore, butyrate may directly and indirectly improve intestinal permeability and immune function, potentially suppressing pro-inflammatory factors such as TNF-α and IL-6 220.

Interestingly, studies have shown relatively indifferent usage of SCFA by colonic tissue in arterio-venous studies 221.

Acetate, on the other hand, appears to be a potent stimulant of intestinal blood flow and plays a role in regulating the brain-pancreas axis regarding insulin release regulation 222.

Propionate, another SCFA, is gaining attention for its role in regulating colonic motility, and high doses of SCFA have been found to inhibit smooth muscle cell proliferation 223224.

Genetic variations in MCT1 and MCT2, which are responsible for SCFA absorption in the gut, may affect colorectal cancer outcomes 225.

Additionally, ketone bodies produced from SCFA β-oxidation serve as precursors for lipid synthesis in human cells, with glucose and glutamine oxidation being more relevant for energy production in the small intestine and proximal colon226227228.

Liver and Adipose Tissue: SCFA in Action

Studies in humans have revealed that butyrate ratios decrease from 20% in the gut lumen to 8% in portal blood229.

SCFA are taken up by the portal circulation and used as energy substrates by hepatocytes, with propionate being a primary player in the liver230.

Specific transporters, MCT1 and MCT2, are predominantly expressed in cells using lactic acid for lipogenesis and gluconeogenesis, such as the liver, kidney tubules, and adipose tissue231.

Acetate is the only SCFA found in substantial amounts in the systemic circulation, reaching approximately 200 μM in venous serum232.

Arterial concentrations of acetate, propionate, and butyrate have been reported as 173, 3.6, and 7.5 mmol/l, respectively, for acetate, propionate, and butyrate, further underlining the hepatic clearance of SCFA233.

Research indicates that propionate plays a role in gluconeogenesis in the liver, while acetate and butyrate are utilized for fatty acid and cholesterol synthesis234.

The impact of SCFA on the tricarboxylic acid (TCA) cycle and energy production varies depending on cell membrane receptors.

MCTs regulate the influx and efflux of SCFA, lactate, pyruvate, and ketone bodies, influencing nutrient access to different cell tissues235.

Studies have shown that MCT4, more abundant than MCT1 in glycolytic cells, regulates the transport of lactate from glycolyzing cells to respiratory cells, impacting cellular functions236.

Systemic Metabolism: A Complex Web of Effects

The effects of SCFA on the body are intricate, diverse, and sometimes indirect, with potential synergistic interactions.

Acetate and pyruvate, close relatives of SCFA, continually feed the TCA cycle, highlighting the ability to convert and reconvert between SCFA and the importance of acetate in energy production237.

Propionate controls the TCA through its conversion to succinate, while butyrate, after β-oxidation, enters the TCA as acetate.

The impact of SCFA on the TCA and energy production is further regulated by cell membrane receptors, with MCTs playing a central role in the transport of SCFA, lactate, pyruvate, and ketone bodies238.

Recent research has shown that plasma levels of acetate, rather than fecal levels of SCFA, are linked to inflammatory markers and lipid subclasses in the blood, highlighting the importance of absorption239.

Another study suggests that circulating SCFA, particularly propionate, is associated with fasting GLP-1 levels and lipid metabolites, which could have implications for insulin sensitivity and metabolic health 240.

The intricate interplay of SCFA in the body extends to the brain, where acetate, propionate, and butyrate can cross the blood-brain barrier, potentially influencing neurological functions 241.

Key Takeaways and Future Horizons in Gut Health Research

Current dietary fiber (DF) intake in Europe falls below recommended levels.

Increasing DF intake could reduce Non-Communicable Diseases (NCDs) by 15-30%.

SCFA, primarily produced from dietary fiber, has significant systemic effects.

They act as a crucial link between diet, gut microbiome, and health.

However, the causality of microbiota-derived metabolites in human disease remains unclear.

Research should prioritize human studies over in vitro findings.

The enterotype, which clusters gut bacterial communities, can have substantial health impacts.

SCFA effects are dose-dependent and vary among different types.

Colonocytes utilize SCFA for energy, influencing various metabolic pathways.

Future Perspectives:

Studies show that the gut microbiota can shift with dietary changes but remains resilient.

Precision dietary interventions, fecal microbiota transplantation, and synbiotics hold promise for addressing various diseases.

Environmental changes, like increased consumption of ultra-processed foods and decreased DF intake, may contribute to the rise in NCDs.

Complex interactions between the host, GM, and dietary patterns require further research, emphasizing the need for individualized approaches.

Investigating associations between dietary patterns, GM, and host genetics holds potential, but specific mechanisms behind phenotypic differences remain unknown.

Future research may pave the way for personalized therapies based on SCFA measurement, without disrupting overall wellbeing.

Review date not set.

How we reviewed this article:

No related category specified or no posts found.

No related product category specified or no products found.