Next Article in Journal
Biocontrol Potential of Fungal and Oomycete Phytopathogens by Myxobacterial Strains
Previous Article in Journal
Revolutionizing Veterinary Vaccines: Overcoming Cold-Chain Barriers Through Thermostable and Novel Delivery Technologies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioactive Metabolites from Yeasts Presumptively Qualified as Safe as Functional Agents in the Management of Type 2 Diabetes

by
Laverdure Tchamani Piame
Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University, 5117 Nelson Mandela Drive, Mthatha 5100, South Africa
Appl. Microbiol. 2025, 5(3), 84; https://doi.org/10.3390/applmicrobiol5030084
Submission received: 30 June 2025 / Revised: 20 July 2025 / Accepted: 21 July 2025 / Published: 20 August 2025

Abstract

Microbial metabolites offer a multitude of mechanisms for alleviating diabetes, particularly type 2 diabetes (T2D). However, the metabolites of yeasts recognised as safe remain under-explored and are receiving less attention in the treatment of T2D. In addition to the recognised probiotic status of certain yeasts, their genetic feature is responsible for many of the effects observed. Branched and non-branched short-chain fatty acids, bioactive peptides, carotenoids, and polysaccharides (β-glucans, mannans, and peptides derived from them) have vital properties that modulate intestinal permeability, soothe inflammation, and directly influence insulin sensitivity. Their action mechanism ranges from hepatic lipogenesis via the induction of hormone-sensitive lipase and the inhibition of α-glucosidase or DPP-IV to promoting the secretion of GLP-1 (Glucagon-Like Peptide-1) and GIP (Gastric Inhibitory Polypeptide), orchestrating immune modulation, and nourishing the gut microbiota. The richness of the yeast metabolome suggests that a concentrated fermentate could be developed to potentiate the functional effects in vitro in the treatment of T2D. The purpose of this review is to take stock of the current state of knowledge of probiotic yeast metabolites and outline their potential for the treatment of diabetes via the development of food supplements or nutraceuticals.

1. Introduction

Modern dietary habits, combined with an increasing consumption of highly calorific, salty, and fatty processed foods, promote the development of metabolic dysfunctions that underlie chronic diseases. One of the most widespread pathologies to date is type 2 diabetes (T2D), which accounts for around 90% of diabetes cases. It is also the result of hereditary, metabolic, and environmental predispositions [1,2]. The onset of this disease occurs when the pancreatic gland fails to secrete enough insulin, or when the body shows a reduced ability to use the insulin produced. In recent decades, probiotics and their metabolites have been proposed as alternative solutions for improving the management of T2D [3]. Their abilities to modulate the gut microbiota, reduce low-grade inflammation, boost the immune system, and reduce glycaemia and insulin resistance have all been attributed to them [3,4,5]. Although most of the research into anti-diabetic probiotics has focused on lactic acid bacteria, probiotic yeasts and their metabolites represent a particularly interesting but largely underexplored category in the treatment of diabetes [6,7].
A few preclinical studies in rats have demonstrated that the administration of the Saccharomyces boulardii THT 500101 strain moderately improves blood glucose levels, reduces systemic inflammation, normalises the renin-angiotensin II system, and corrects the lipid profile [6,8,9,10]. In addition to Saccharomyces boulardii, the most well-known and commercially available species, this list includes Saccharomyces bayanus, S. pastorianus, Kluyveromyces lactis, Komagataella phaffii, K. pastoris, Schizosaccharomyces pombe, Cyberlindnera jadinii, Hanseniaspora uvarum, Xanthophyllomyces dendrorhous, Ogataea angusta, Limtongozyma cylindracea, Yarrowia lipolytica, and Zygosaccharomyces rouxii [11]. However, their application in the management of diabetes remains very limited or even ineffective, despite the presence in their fermentates of bioactive molecules (organic acids, bioactive peptides, polysaccharides, and carotenoids) known for their ability to alleviate diabetes and/or its complications [12,13,14,15,16]. These metabolites may act as signalling molecules, interpreted by host cells and triggering a cascade of reactions likely to regulate energy metabolism, insulin resistance, and oxidative stress. Therefore, exploring these bioactive compounds could pave the way for a postbiotic approach aimed at formulating food supplements or nutraceuticals that are more stable over time than living cells [17]. The purpose of this review is to take stock of the current state of knowledge of probiotic yeast metabolites and outline their potential for the treatment of diabetes mellitus via the development of food supplements or nutraceuticals

2. Discovery of Probiotics and Characteristics of Probiotic Yeasts

The European Food Safety Authority (EFSA) has determined that a subset of yeast species known as QPS (Qualified Presumption of Safety) yeasts pose little risk of endangering the health of humans or animals. In the absence of probiotic properties, they may still produce bioactive molecules that can be exploited as postbiotics. The exploration of probiotics has long relied on a “top-down” approach, identifying as beneficial the dominant microorganisms in healthy individuals, notably strains of Bifidobacterium and Lactobacillus [18]. An alternative method, based on the exploration of fermented foods to isolate potentially probiotic micro-organisms, corresponds to the initial approach adopted by Metchnikoff for the isolation and identification of Lactobacillus bulgaricus [19]. Advances in molecular techniques have made it possible to extend this research to species that are difficult to cultivate, such as Faecalibacterium prausnitzii, Akkermansia muciniphila, and Microvirga massiliensis [20,21]. At the same time, bottom–up strategies inspired by drug discovery have emerged, divided into phenotypic and targeted approaches [22]. The former involves evaluating probiotic effects in vitro, ex vivo, and in vivo, as with Lactobacillus rhamnosus JB-1, whose benefits observed in mice have not been confirmed in humans. The second identifies candidates who can produce molecular effectors that modulate important host or microbiota pathways using in silico predictions based on multi-omics data [23,24].
Probiotics are live microorganisms that provide health benefits to humans and animals when consumed in adequate amounts. They enhance the activity of the host’s endogenous microflora, modulate the immune system, support digestion and metabolism, and influence the gut–liver axis [25,26,27]. Yeasts represent a valuable alternative to bacterial probiotics, especially in contexts where antibiotics are frequently used in therapeutic treatments to suppress or eliminate bacterial strains [28]. Their strong resistance to gastric acidity ensures their survival through the stomach and enables them to reach the intestine, where they can colonise effectively thanks to specific surface proteins and glycoproteins [29]. Once established, yeast probiotics exert immunomodulatory effects by stimulating cytokine production and enhancing immune cell activity. They also exhibit antimicrobial action by secreting organic acids, hydrogen peroxide, and antifungal peptides [28]. However, the beneficial effects may be attributed either to the yeast cells themselves or to the various metabolites they produce.

3. Metabolites of QPS Yeasts

Contemporary research reveals a growing interest in the constituents of yeast extract and metabolites secreted by probiotic yeasts in the management of chronic diseases such as T2DM [12]. This approach should be based on concrete scientific evidence, demonstrating that the beneficial effects of probiotics come not only from their living presence but also from the bioactive molecules they produce and release into their environment [30,31]. Several studies have indicated that these compounds can have significant effects on glycaemic control, insulin sensitivity, and systemic inflammation, opening new therapeutic prospects for diabetic patients [32,33,34,35]. Unfortunately, the studies demonstrating the action of the metabolites excreted by yeasts are few and do not make it possible to assess the direct effect on humans, which is why this review will focus on the potential effects of antidiabetic yeast metabolites.

3.1. Metabolite Profile of QPS Yeasts

Yeast metabolism can be likened to the components of a factory, where each stage yields an intermediate product and a final product at the end of the chain, including waste. Each of these products or by-products plays a role in the cell, although they can become harmful depending on their concentration. Their nature also varies according to the metabolic pathway employed, which itself is selected based on several factors, such as the type and concentration of substrate, oxygen availability, temperature, and more [36].
In addition to ethanol abundantly produced by species of the genera Saccharomyces, Pichia, Schizosaccharomyces, Kluyveromyces, Candida, among others, yeasts generate a wide range of metabolites. Food-relevant metabolites are the most extensively documented, including glycerol, succinate, butyrate, acetate, α-ketoglutarate, and malate [36,37]. Except for malate, ethanol, and glycerol, many of these compounds can be utilised by colonocytes as an energy source [38]. Yeasts also synthesise a variety of amino acids via the shikimate pathway and produce numerous volatile organic compounds, such as phenyllactic acid, isoamyl alcohols, ethyl acetate, ethyl butyrate, ethyl isobutyrate, acetaldehyde, 1,1-diethoxyethane, 2-hydroxyisocaproic acid, and limonene [39,40,41]. They also release GABA (γ-aminobutyric acid), shikimic acid (a precursor to phenylalanine, tyrosine, and tryptophan), p-aminobenzoic acid, and tyrosol [42]. Yeasts, particularly Saccharomyces boulardii, produce bioactive peptides involved in regulating carbohydrate metabolism of interest for diabetes management and inhibiting pathogenic microorganisms. In addition to these compounds, certain strains are capable of synthesising polysaccharides and carotenoids, which possess antioxidant properties, modulate inflammation, and stimulate the immune system [12,43].
It is important to note that the metabolomic profile is not fixed; it varies depending on the culture medium in which the microorganism is grown. In the following sections of this manuscript, metabolites with antidiabetic potential are grouped into organic acids (including short-chain fatty acids, branched-chain short-chain fatty acids, and other organic acids), bioactive peptides, and carotenoids, to highlight their relevance in diabetes management.

3.2. Antidiabetic Potential of Yeast-Secreted Metabolites

3.2.1. Branched and Non-Branched Short Chain Fatty Acids

Short-chain fatty acids (SCFAs) are generally produced during in vitro and in vivo fermentation. The microorganisms best studied for this purpose are lactic acid bacteria, although yeasts also produce short-chain fatty acids in modest proportions.
Acetic acid is commonly produced by Saccharomyces and non-Saccharomyces yeasts (Saccharomyces cerevisiae, Zygosaccharomyces bailii, Brettanomyces spp.) and is the product of the conversion of pyruvate to acetate via acetaldehyde [44]. It is the product of numerous microbial fermentations and is a SCFA, recognised for its ability to modulate the gut microbiota [4,6]. Although specific data on its anti-diabetic effects is less well documented than for succinic acid, acetate plays an important role in regulating carbohydrate and lipid metabolism. This molecule, produced mainly by fermentation, improves insulin sensitivity by activating G protein-coupled receptors and regulates gene expression linked to glucose and lipid metabolism by inhibiting histone decarboxylases [45]. It has been shown that acetate consumption in rats is marked by a reduction in accumulation in the liver and adipose tissue, demonstrating that acetate can help correct glucose intolerance and reduce fat accumulation, which are dysfunctions that favour the development of diabetes [46]. Although it does not have a direct effect on blood sugar levels, acetate regulates fat mass reduction and steatosis via the repression of lipogenic genes. In addition, acetate is involved in the specific activation of an enzyme involved in maintaining energy balance in cells, AMPK (AMP-activated protein kinase). This activation is linked to an increase in the AMP/ATP ratio due to the increased formation of acetyl-CoA via short-chain acyl-CoA synthetases (ACSS1 and ACSS2). In turn, phosphorylation of ChREBP (carbohydrate-responsive element-binding protein) by AMPK inhibits its activity, leading to a decrease in transcription of these genes [46]. An analysis using Northern blot techniques revealed that the transcripts of several lipogenic genes in the liver were reduced when acetate was administered [46]. All these phenomena work together to activate the β-oxidation of fatty acids and inhibit hepatic glucose synthesis, thereby improving carbohydrate homeostasis [47]. The mechanisms of action of acetate on energy metabolism, immunity, and intestinal integrity are depicted in Figure 1.
Like short-chain fatty acids (SCFAs), their branched-chain counterparts (BSCFAs) also exhibit similar biological effects. Both serve as energy sources for hepatocytes and colonocytes [48]. Their involvement in carbohydrate and lipid metabolism has been associated with improved insulin sensitivity and enhanced lipid catabolism in adipocytes. Notably, it has been shown that branched short-chain fatty acids, particularly isobutyric acid and isovaleric acid, exert an inhibitory effect on lipolysis induced by isoprenaline (a non-selective beta-adrenergic receptor agonist) in primary rat adipocytes. Interestingly, this inhibition occurs independently of the PI3-kinase and phosphodiesterase three pathways, which are the main regulatory routes of lipolysis. Moreover, BSCFAs promote lipolysis by activating protein kinase A (PKA), which in turn phosphorylates and activates hormone-sensitive lipase [49].
Furthermore, BSCFAs have the capacity to inhibit the activation (phosphorylation) of protein kinase B (PKB), a key serine/threonine kinase involved in the insulin-signalling pathway [50]. Mechanistically, insulin binds to its receptor and initiates a signalling cascade that leads to PKB activation, ultimately facilitating glucose uptake into cells via GLUT4 transporters. By inhibiting this process, BSCFAs may act antagonistically to insulin, thereby preventing glucose entry into rat cells [49,51].

3.2.2. Other Organic Acids

Yeasts produce, in addition to SCFAs and BSCFAs, other organic acids, among which succinic and malic acids stand out for their influence on carbohydrate and lipid metabolism. Several studies have underlined the significant role these acids may play in diabetic patients and their associated comorbidities.
An investigation by Ives et al. [52] examined the impact of succinic acid on lipid profiles and insulin resistance in rats rendered obese by a high-fat diet. The authors reported a marked reduction in adipose tissue mass and a decline in the mitochondrial respiratory capacity in oxidative muscle. Paradoxically, a supplementation with succinic acid at 0.75 mg/mL (equivalent to 6.35 mM) led to an increase in the fasting blood glucose in these animals. This finding contrasts with that of Yuxuan Yang et al. [53], who observed that a 40 mM succinate supplementation lowered the fasting glycaemia and improved glucose tolerance. These discrepancies suggest that succinic acid’s effects on glycaemic control are complex and may hinge on its concentration (with improvements in insulin tolerance noted from around 5 mM) as well as on the experimental setting. Indeed, in Ives et al.’s study, no enhancement of the glucose tolerance or insulin sensitivity was detected, indicating that the reduction in fat mass does not necessarily translate into better carbohydrate metabolism [52]. Nevertheless, Yang et al. also found that succinate improved the lipid profile by reducing plasma triglycerides, total cholesterol, and LDL-C, while raising HDL-C levels [53].
The biological action of succinic acid is mediated through the cell-surface receptor SUCNR1 (GPR91), a metabolic sensor expressed in various tissues, including the liver, white adipose tissue, and retina. Upon binding to SUCNR1, succinic acid triggers the p38 MAPK- and PI3K/AKT-signalling cascades, both of which are pivotal for adipocyte differentiation and the maintenance of energy homeostasis [53]. One notable outcome of this pathway activation is the browning of white adipose depots, a process marked by enhanced thermogenesis and elevated energy expenditure [54]. A schematic representation of this mechanism of action is shown in Figure 2.
By contrast, malic acid has been evaluated in the context of xerostomia, a common complication of T2D, via topical application. In a controlled trial involving 52 type 2 diabetic patients, a 1% malic acid formulation applied for four weeks led to a significant increase in unstimulated salivary flow. However, to date, the literature offers no evidence regarding malic acid’s potential to modulate energy metabolism in individuals with T2D [55].

3.2.3. Bioactive Peptides

Bioactive peptides produced from yeast have a wide range of biological functions, including antioxidant, hypotensive, and antimicrobial effects. They are mainly obtained by yeast fermentation, a process in which yeast extracts are used as substrates, and proteolytic enzymes from microorganisms fragment the proteins to generate functional peptides [56].
A study by Aquino et al. [12] on the effect of bioactive peptides extracted from the yeast Saccharomyces cerevisiae highlighted a multitude of beneficial effects in regulating the metabolism of people with diabetes. These include their ability to inhibit enzymes linked to glucose metabolism, such as α-glucosidase and dipeptidyl peptidase IV (DPP-IV). Glycosylation had a differentiating effect on the activity, with peptides carrying mannose in their primary chain being more effective at inhibiting α-glucosidase, whereas non-glycosylated peptides preferentially inhibited DPP-IV. In addition, the impact of these peptides on the gene expression related to glucose metabolism was assessed using mouse jejunum organoids. The results indicated a down-regulation of genes associated with carbohydrate catabolism and glucose uptake, as well as an increased expression of incretin hormones, such as glucose-dependent insulinotropic peptide (GIP) and relevant glucagon-like peptide 1 (GLP-1) [12]. A Cyclo-His-Pro-rich hydrolysate, produced by enzymatic hydrolysis (neutrase, alkalase, protamex, flavourzyme, ficin at 50 °C, W/O 1/100), significantly lowered the blood glucose levels after an oral glucose tolerance test in male ICR mice for up to 120 min [57].
Unlike some anti-diabetic agents, inhibitory peptides have the advantage of not causing hypoglycaemia and are thought to have a direct effect on pancreatic β cells, stimulating their differentiation and proliferation. A meta-analysis conducted by Zhou et al. [58] showed that DPP-IV inhibitors were associated with a significantly lower risk of hypoglycaemia compared with conventional sulphonylureas in type 2 diabetic subjects. Among the outstanding features of these peptides is their ability to neutralise free radicals and prevent lipid peroxidation, which helps to alleviate oxidative stress in the body, thereby reducing chronic inflammation and the dysregulation of oxidative mediators that impair pancreatic β-cell function. They also play an antimicrobial role.
Promising results have shown that certain peptides excreted during yeast fermentation can delay the proliferation of pathogenic microorganisms by disrupting the wall stability or binding to sites essential to the survival of fungi and bacteria; the best known are mycocins, extracellular proteins or glycoproteins with an inhibitory effect against species close to the yeast that produces them. These antimicrobial peptides are effective at pH 4–7 against several pathogenic species, such as Bacillus subtilis, Escherichia coli, Klebsiella aerogenes, and Staphylococcus aureus. Their mode of action varies according to their charge. Cationic peptides bind to the anionic components of membranes, causing them to rupture, while anionic peptides cross the membrane, form pores, and lead to uncontrolled exchange between the intra- and extracellular compartments [59,60]. Branco et al. [61] showed that GAPDH fragments alter the plasma and vacuolar membranes of Hanseniaspora guilliermondii, lowering the intracellular pH from 6.5 to 5.4 in 20 min. In addition, S. cerevisiae K1 toxin creates channels that imbalance H+ and K+ fluxes, leading to cell death [62].

3.2.4. Carotenoids

Carotenoids represent a broad class of naturally occurring pigments found not only in fruits and vegetables but also synthesised by fungi, including yeasts [63,64]. Chemically, these compounds are hydrocarbons divided into two main classes: carotenes, which are non-oxygenated and non-polar, and xanthophylls, which are oxygenated and polar [65]. A distinctive feature of yeast strains capable of carotenoid production is the pigmentation of their colonies, ranging in colour from pale orange to deep red. Among the carotenoids most widely recognised for their biological activity are β-carotene, γ-carotene, astaxanthin, torulene, and torularhodin (native yeasts), lycopene, canthaxanthin, and zeaxanthin (engineered yeasts) (see Table 1). These compounds are associated with a range of health-supporting properties, particularly in relation to the prevention of cancer and cardiovascular diseases, as well as complications linked to diabetes mellitus. Their beneficial effects have been attributed to various mechanisms, including the enhancement of insulin sensitivity and the attenuation of oxidative and inflammatory stress [36,66].
Few investigations have reported an inverse association between the plasma levels of certain carotenoids, such as lycopene and β-carotene, and fasting insulin concentrations or the presence of metabolic syndrome [36,67]. Furthermore, astaxanthin has demonstrated the capacity to improve glucose tolerance and markedly increase insulin responsiveness. The biological mechanisms underpinning these effects are multifactorial. Carotenoids exert protective effects on pancreatic β-cells by mitigating inflammation and neutralising reactive oxygen species that contribute to oxidative injury. In murine models, the administration of astaxanthin was shown to suppress the upregulation of oxidative enzymes. Astaxanthin exhibits particularly potent antioxidant properties, reportedly surpassing those of lutein, β-carotene, and zeaxanthin [68]. Astaxanthin has also been shown to protect pancreatic β-cells from glucotoxic damage and to modulate the expression of key inflammatory proteins, including Nuclear Factor kappa B (NF-κB), Cyclo-oxygenase-2 (COX-2), Intercellular Adhesion Molecule-1 (ICAM-1), and inducible Nitric Oxide Synthase (iNOS) molecules known to sustain inflammatory cascades, nociception, and oxidative stress. In addition, it reduces the levels of pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β. This cytokine downregulation supports the anti-inflammatory response observed in the brain, particularly within the cerebral cortex of diabetic rats [69,70]. These carotenoids are of particular interest for their role in mitigating or preventing complications of diabetes, including nephropathy, retinopathy, and neurological dysfunction. Although little is currently known about the in vitro biological activity of torularhodin, it has found practical use as a natural pigment in the agri-food sector.
Table 1. Yeast carotenoids, antidiabetic properties, and action mechanisms.
Table 1. Yeast carotenoids, antidiabetic properties, and action mechanisms.
CarotenoidProducing YeastClassAntidiabetic PropertiesReference
β-CaroteneRhodotorula glutinis, Sporobolomyces roseusCarotene≈22% reduction in T2D risk, enhanced insulin sensitivity, antioxidant defence[36]
γ-CaroteneSporobolomyces spp., Sporidiobolus spp.CaroteneAntioxidant properties, cardiovascular protection in diabetic individuals[71]
AstaxanthinXanthophyllomyces dendrorhousXanthophyllImproved glucose tolerance, reduced HbA1c, enhanced insulin responsiveness[71]
TorularhodinSporidiobolus pararoseus, Rhodotorula spp.XanthophyllImproved lipid metabolism, reduced hepatic inflammation, enhanced insulin sensitivity[72]
ToruleneSporidiobolus pararoseus, Rhodotorula glutinisCaroteneProtection against hyperglycaemia-induced oxidative stress, superior antioxidant activity[73]
LycopeneCandida utilis (engineered), Saccharomyces cerevisiae (engineered)CaroteneReduced blood glucose, improved lipid profile, protection against diabetic complications[74]
CanthaxanthinSaccharomyces cerevisiae (engineered)XanthophyllOxidative stress tolerance, anti-inflammatory properties[75]
ZeaxanthinSaccharomyces cerevisiae (engineered)XanthophyllProtection of diabetic retina, improved visual function[75]

3.2.5. Polysaccharides

The yeast cell is a natural source of complex polysaccharides, namely mannan oligosaccharides and β-d-glucan, which possess biological properties. Supplementation of these polymers in the diet induces the modulation of energy metabolism, immune response, hormonal balance, and the gastrointestinal tract [7,76].
The aforementioned effects are partly supported by a meta-analysis including three review studies and 273 participants, which demonstrated that the intake of S. cerevisiae hydrolysate (Stetikal® (Quito, Ecuador), Lipigo® (Barcelona, Spain)) induced a generalised weight loss, with a reduction in abdominal circumference and fat mass [77]. The administration of β-glucans (30 mg/kg/d, for 28 d) in type 2 diabetic rats resulted in a reduction in several biochemical markers, such as glycaemia, triglycerides, and alanine aminotransferase by 30, 32, and 41%, respectively [76]. In a randomised, placebo-controlled, phase I pilot study, supplementation with yeast β-glucan (2.5 g/d) significantly reduced the insulin resistance and TNFα after 8 and 4 weeks, respectively, in the treated group, with an increase in bile acid levels, of which tauroursodeoxycholic acid accounted for the majority, without any close link with gut microbiota or SCFAs [7]. In fact, the increase in intestinal bile acid levels is strongly correlated with a reduction in serum cholesterol levels. By binding to bile acids in the intestine, β-glucans promote their excretion in the faeces, encouraging the body to produce more by using the endogenous cholesterol stock [7]. Similarly, β-glucans can be used as a prebiotic by the intestinal flora, whose fermentation generates SCFAs capable of exerting hypocholesterolemic and anti-inflammatory effects that are essential for restoring the integrity of the intestinal barrier and pancreatic β-cells [78].
Mannan oligosaccharides (MOS) also have interesting properties for alleviating the subjacent complications of diabetes. MSOs supplementation in obese rats had no significant effect on carbohydrate homeostasis and inflammation, whereas in lean rats, it led to an increase in the number of immune cells (macrophages and eosinophils) in mesenteric white adipose tissue [79]. MSOs are also thought to have prebiotic effects that promote the modulation of the gut microbiota and therefore the reversal of dysbiosis. A study by Reiko Tanihiro et al. [80] showed that the consumption of MSOs increased the relative abundance of bacteroides, specifically Bacteroides thetaiotaomicron and Bacteroides ovatus, and reduced the concentration of faecal p-cresol, indole, and skatole, harmful metabolites associated with intestinal dysbiosis.
Yeast polysaccharides stand out for their low production cost, simple extraction technology, and stable and potentially inexhaustible supply, ensuring their strong dominant position in the market for various essential carbohydrate functional ingredients for years to come [31,81,82]. They exert multiple actions that, although not favourable for maintaining carbohydrate homeostasis, modulate and reduce immunity, inflammation, and intestinal dysbiosis and could be used as dietary supplements. The combined effects of these polysaccharides allow us to speculate on the possibilities of formulating effective nutritional supplements or nutraceuticals. Figure 3 shows the roles of yeast polysaccharides (β-glucans and mannan oligosaccharides) in regulating glycaemia, preventing metabolic syndromes, and boosting immunity.

4. Potential of Postbiotic Formulations, Knowledge Gaps and Perspectives

4.1. Potential of Postbiotic Formulations from Yeast

The translation of fundamental research towards therapeutic applications should generate more interest in exploring the metabolites of microorganisms, in this case, yeast, for the development of the postbiotic approach. This approach is sustainable insofar as it presents concentrates of pure or combined molecules that remain stable when combined with other formulations, have a fairly long shelf life (generally two years), and require less stringent storage conditions than probiotics, whose loss of viability during storage remains a challenge to overcome for certain formulations [17,83].
Unfortunately, postbiotic forms of yeast metabolites (Stetikal, Lipigo, EpiCor) are virtually non-existent in the world market. However, the best-known is EpiCor, a dehydrated fermentate of the yeast S. cerevisiae, renowned for its antioxidant content. It has been tested for its ability to protect cells against oxidative damage and its ability to improve immune health [84,85]. Several randomised, placebo-controlled clinical trials in which participants received a dose of 500 mg/d for 12 weeks showed significant curative effects on flu and colds compared with the placebo group [86,87]. Another 12-week trial showed that PE significantly reduced the average severity of specific symptoms of allergic rhinitis, particularly nasal congestion and rhinorrhoea [88].
More specifically, the consumption of EpiCor has antioxidant and immunomodulatory properties, transiently reducing the number of circulating T and NK lymphocytes and increasing interferon C levels [87,88,89]. Such mechanisms are thought to alleviate the basis of several inflammatory diseases, such as inflammatory bowel disease, irritable bowel disease, gout, and diabetes.

4.2. Knowledge Gaps and Perspectives

Synthetic biology is an interdisciplinary science that applies the principles of engineering (modular design, standardisation, automation) to biological systems. Thus, thanks to synthetic biology, it is possible to precisely reprogramme the metabolic pathways of yeast organisms for the overproduction of molecules of interest by optimising key genes, strengthening limiting enzymes, and adjusting the pool of co-factors (NADH/NAD+, ATP, coenzyme, etc.) [90]. These authors, respectively, described how the introduction of four metabolic reactions into the S. cerevisiae genome reduced ATP consumption and CO2 losses and rebalanced the NADH/NAD+ redox couple for farnesene production. In addition, the establishment of synthetic consortia will enable bioactive molecules to be synthesised, thanks to the synergistic action of strains which will sequentially degrade substrates to mutually synthesise another.
A lasting solution to the challenge of treating metabolic disorders such as diabetes lies in the strategy for disseminating bioactive formulations. Researchers could focus on the supplementation of dried fermentates in appropriate foods to suit the diets of their subjects. In this way, the range of foods and beverages that are widely consumed, accessible, and without adverse effects on long-term health can be the subject of serious and relevant studies. In addition, bars, capsules, or powders could be proposed to prevent or manage the pathological components that support diabetes.

5. Conclusions

Exploring the under-explored metabolic profile of probiotic yeasts in the context of type 2 diabetes reveals a promising therapeutic potential. The metabolites identified (SCFAs, BSCFAs, bioactive peptides, carotenoids, and polysaccharides) have complementary anti-diabetic effects. SCFAs and BSCFAs improve the insulin sensitivity and glucose tolerance whilst modulating the lipid and immune homeostasis. Bioactive peptides inhibit α-glucosidase and DPP-IV, stimulate the secretion of incretins (GLP-1, GIP), and protect β-cells without inducing hypoglycaemia. Carotenoids bolster antioxidant defences, reduce oxidative stress and inflammation (NF-κB, COX-2), and lower HbA1c whilst improving the lipid profile. Finally, β-glucans and mannan oligosaccharides regulate blood glucose, triglycerides, insulin resistance, and inflammation (TNF-α); promote bile acid excretion; and positively modulate the gut microbiota. Exploiting these metabolites for postbiotic purposes could provide a standardised and stable alternative to probiotics. Nevertheless, the validation of these results via complementary in vitro tests will enable us to gain a deeper understanding of the mechanisms of action and to envisage clinical trials, the data from which, via meta-analyses, will enable us to make more generalised recommendations.

Funding

The article processing charge was funded by the Directorate of Research and Innovation of Walter Sisulu University, South Africa.

Data Availability Statement

No data was generated by this article.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Fletcher, B.; Gulanick, M.; Lamendola, C. Risk factors for type 2 diabetes mellitus. J. Cardiovasc. Nurs. 2002, 16, 17–23. [Google Scholar] [CrossRef] [PubMed]
  2. Dai, M.; Guo, W.; Zhu, S.; Gong, G.; Chen, M.; Zhong, Z.; Guo, J.; Zhang, Y. Type 2 diabetes mellitus and the risk of abnormal spermatozoa: A Mendelian randomization study. Front. Endocrinol. 2022, 13, 1035338. [Google Scholar] [CrossRef] [PubMed]
  3. Ayesha, I.E.; Monson, N.R.; Klair, N.; Patel, U.; Saxena, A.; Patel, D.; Venugopal, S.; Ayesha, I.E. Probiotics and their role in the management of type 2 diabetes mellitus (Short-term versus long-term effect): A systematic review and meta-analysis. Cureus 2023, 15, 10. [Google Scholar] [CrossRef] [PubMed]
  4. Shen, X.; Ma, C.; Yang, Y.; Liu, X.; Wang, B.; Wang, Y.; Zhang, G.; Bian, X.; Zhang, N. The Role and Mechanism of Probiotics Supplementation in Blood Glucose Regulation: A Review. Foods 2024, 13, 2719. [Google Scholar] [CrossRef] [PubMed]
  5. Piame, L.T. Potential of Non-Dairy Probiotic Beverages to Modulate Gut Microbiota and Improve Management of Non-Communicable Diseases. Nov. Res. Microbiol. J. 2025, 9, 198–213. [Google Scholar] [CrossRef]
  6. Everard, A.; Matamoros, S.; Geurts, L.; Delzenne, N.M.; Cani, P.D. Saccharomyces boulardii administration changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and type 2 diabetic db/db mice. MBio 2014, 5, e01011-14. [Google Scholar] [CrossRef] [PubMed]
  7. Cronin, P.; Hurley, C.; Ryan, A.; Zamora-Úbeda, M.; Govindan, A.; Stanton, C.; Lane, G.P.; Joyce, S.A.; O’Toole, P.W.; O’Connor, E.M. Yeast β-glucan supplementation lowers insulin resistance without altering microbiota composition compared with placebo in subjects with type II diabetes: A phase I exploratory study. Br. J. Nutr. 2024, 132, 1161–1172. [Google Scholar] [CrossRef] [PubMed]
  8. Albuquerque, R.C.M.F.; Brandão, A.B.P.; De Abreu, I.C.M.E.; Ferreira, F.G.; Santos, L.B.; Moreira, L.N.; Taddei, C.R.; Cunha, F.A.A.T.S. Saccharomyces boulardii Tht 500101 changes gut microbiota and ameliorates hyperglycaemia, dyslipidaemia, and liver inflammation in streptozotocin-diabetic mice. Benef. Microbes 2019, 10, 12. [Google Scholar] [CrossRef] [PubMed]
  9. Abreu, I.; Albuquerque, R.; Brandão, A.B.P.; Barssotti, L.; de Souza, L.B.; Ferreira, F.G.; Oliveira, L.C.G.; Yokota, R.; Sparvoli, L.G.; Dias, D.D.S.; et al. Saccharomyces boulardii exerts renoprotection by modulating oxidative stress, renin angiotensin system and uropathogenic microbiota in a murine model of diabetes. Life Sci. 2022, 301, 120616. [Google Scholar] [CrossRef] [PubMed]
  10. Barssotti, L.; Abreu, I.C.M.E.; Brandão, A.B.P.; Albuquerque, R.C.M.F.; Ferreira, F.G.; Salgado, M.A.C.; Dias, D.D.S.; De Angelis, K.; Yokota, R.; Casarini, D.E.; et al. Saccharomyces boulardii modulates oxidative stress and renin angiotensin system attenuating diabetes-induced liver injury in mice. Sci. Rep. 2021, 11, 9189. [Google Scholar] [CrossRef] [PubMed]
  11. Hazards, E.P.o.B.; Koutsoumanis, K.; Allende, A.; Alvarez-Ordóñez, A.; Bolton, D.; Bover-Cid, S.; Chemaly, M.; De Cesare, A.; Hilbert, F.; Lindqvist, R. Update of the list of qualified presumption of safety (QPS) recommended microbiological agents intentionally added to food or feed as notified to EFSA 19: Suitability of taxonomic units notified to EFSA until September 2023. EFSA J. 2024, 22, e8517. [Google Scholar] [CrossRef] [PubMed]
  12. Aquino, M.E.; Drago, S.R.; de Medina, F.S.; Martínez-Augustin, O.; Cian, R.E. Anti-diabetic properties of brewer’s spent yeast peptides. In vitro, in silico and ex vivo study after simulated gastrointestinal digestion. Food Funct. 2024, 15, 3778–3790. [Google Scholar] [CrossRef] [PubMed]
  13. Cao, Y.; Sun, Y.; Zou, S.; Li, M.; Xu, X. Orally Administered Baker’s Yeast β-Glucan Promotes Glucose and Lipid Homeostasis in the Livers of Obesity and Diabetes Model Mice. J. Agric. Food Chem. 2017, 65, 9665–9674. [Google Scholar] [CrossRef] [PubMed]
  14. Poorniammal, R.; Sarathambal, C.; Sakthi, A.; Arun, S. Yeast carotenoids importance in food and feed industries and its health benefits–A review. Agric. Rev. 2013, 34, 307–312. [Google Scholar] [CrossRef]
  15. Noh, Y.-H.; Lee, D.-B.; Lee, Y.-W.; Pyo, Y.-H. In vitro inhibitory effects of organic acids identified in commercial vinegars on α-amylase and α-glucosidase. Prev. Nutr. Food Sci. 2020, 25, 319. [Google Scholar] [CrossRef] [PubMed]
  16. Baek, K.-R.; Lee, H.-J.; Jeong, K.-H.; Park, B.-R.; Kim, S.-J.; Seo, S.-O. Parabiotic immunomodulatory activity of yeast cell wall polysaccharides from Saccharomyces cerevisiae and S. boulardii. J. Funct. Foods 2024, 123, 106577. [Google Scholar] [CrossRef]
  17. Prajapati, N.; Patel, J.; Singh, S.; Yadav, V.K.; Joshi, C.; Patani, A.; Prajapati, D.; Sahoo, D.K.; Patel, A. Postbiotic production: Harnessing the power of microbial metabolites for health applications. Front. Microbiol. 2023, 14, 1306192. [Google Scholar] [CrossRef] [PubMed]
  18. O’Toole, P.W.; Marchesi, J.R.; Hill, C. Next-generation probiotics: The spectrum from probiotics to live biotherapeutics. Nat. Microbiol. 2017, 2, 17057. [Google Scholar] [CrossRef] [PubMed]
  19. Mackowiak, P.A. Recycling metchnikoff: Probiotics, the intestinal microbiome and the quest for long life. Front. Public Health 2013, 1, 52. [Google Scholar] [CrossRef] [PubMed]
  20. Lagier, J.C.; Armougom, F.; Million, M.; Hugon, P.; Pagnier, I.; Robert, C.; Bittar, F.; Fournous, G.; Gimenez, G.; Maraninchi, M.; et al. Microbial culturomics: Paradigm shift in the human gut microbiome study. Clin. Microbiol. Infect. 2012, 18, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
  21. Bilen, M.; Dufour, J.C.; Lagier, J.C.; Cadoret, F.; Daoud, Z.; Dubourg, G.; Raoult, D. The contribution of culturomics to the repertoire of isolated human bacterial and archaeal species. Microbiome 2018, 6, 94. [Google Scholar] [CrossRef] [PubMed]
  22. Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 2019, 25, 716–729. [Google Scholar] [CrossRef] [PubMed]
  23. Kelly, J.R.; Allen, A.P.; Temko, A.; Hutch, W.; Kennedy, P.J.; Farid, N.; Murphy, E.; Boylan, G.; Bienenstock, J.; Cryan, J.F. Lost in translation? The potential psychobiotic Lactobacillus rhamnosus (JB-1) fails to modulate stress or cognitive performance in healthy male subjects. Brain Behav. Immun. 2017, 61, 50–59. [Google Scholar] [CrossRef] [PubMed]
  24. Lucas, N.; Legrand, R.; Deroissart, C.; Dominique, M.; Azhar, S.; Le Solliec, M.-A.; Léon, F.; Rego, J.-C.D.; Déchelotte, P.; Fetissov, S.O. Hafnia alvei HA4597 strain reduces food intake and body weight gain and improves body composition, glucose, and lipid metabolism in a mouse model of hyperphagic obesity. Microorganisms 2019, 8, 35. [Google Scholar] [CrossRef] [PubMed]
  25. Gu, Y.; Wang, C.; Qin, X.; Zhou, B.; Liu, X.; Liu, T.; Xie, R.; Liu, J.; Wang, B.; Cao, H. Saccharomyces boulardii, a yeast probiotic, inhibits gut motility through upregulating intestinal serotonin transporter and modulating gut microbiota. Pharmacol. Res. 2022, 181, 106291. [Google Scholar] [CrossRef] [PubMed]
  26. Pothoulakis, C. anti-inflammatory mechanisms of action of Saccharomyces boulardii. Aliment. Pharmacol. Ther. 2009, 30, 826–833. [Google Scholar] [CrossRef] [PubMed]
  27. Stier, H.; Bischoff, S.C. Influence of Saccharomyces boulardii CNCM I-745on the gut-associated immune system. Clin. Exp. Gastroenterol. 2016, 13, 269–279. [Google Scholar] [CrossRef] [PubMed]
  28. Staniszewski, A.; Kordowska-Wiater, M. Probiotic and potentially probiotic yeasts—Characteristics and food application. Foods 2021, 10, 1306. [Google Scholar] [CrossRef] [PubMed]
  29. Alkalbani, N.S.; Osaili, T.M.; Al-Nabulsi, A.A.; Olaimat, A.N.; Liu, S.-Q.; Shah, N.P.; Apostolopoulos, V.; Ayyash, M.M. Assessment of yeasts as potential probiotics: A review of gastrointestinal tract conditions and investigation methods. J. Fungi 2022, 8, 365. [Google Scholar] [CrossRef] [PubMed]
  30. Tullio, V. Probiotic yeasts: A developing reality? J. Fungi 2024, 10, 489. [Google Scholar] [CrossRef] [PubMed]
  31. Tchamani Piame, L.; Kaktcham, P.M.; Kouam, E.M.F.; Techeu, U.D.F.; Ngouénam, R.J.; Ngoufack, F.Z. Technological characterisation and probiotic traits of yeasts isolated from Sha’a, a Cameroonian maize-based traditional fermented beverage. Heliyon 2022, 8, e10850. [Google Scholar] [CrossRef] [PubMed]
  32. Shruthi, B.; Deepa, N.; Somashekaraiah, R.; Adithi, G.; Divyashree, S.; Sreenivasa, M. Exploring biotechnological and functional characteristics of probiotic yeasts: A review. Biotechnol. Rep. 2022, 34, e00716. [Google Scholar] [CrossRef] [PubMed]
  33. Moslehi-Jenabian, S.; Pedersen, L.L.; Jespersen, L. Beneficial effects of probiotic and food borne yeasts on human health. Nutrients 2010, 2, 449–473. [Google Scholar] [CrossRef] [PubMed]
  34. Hosseinzadeh, P.; Djazayery, A.; Mostafavi, S.-A.; Javanbakht, M.H.; Derakhshanian, H.; Rahimiforoushani, A.; Djalali, M. Brewer’s yeast improves blood pressure in type 2 diabetes mellitus. Iran. J. Public Health 2013, 42, 602. [Google Scholar] [PubMed]
  35. Horn, P.A.; Zeni, A.L.B.; Herkenhoff, M.E.; Curbani, L.; Gonçalves, G.H.P.; Rutkoski, C.F.; Israel, N.G.; de Almeida, E.A. Brewer’s spent yeast improves human gut microbiota and ameliorates clinical blood parameters: A randomized, double-blind, placebo-controlled trial. Bioact. Carbohydr. Diet. Fibre 2024, 32, 100442. [Google Scholar] [CrossRef]
  36. Roohbakhsh, A.; Karimi, G.; Iranshahi, M. Carotenoids in the treatment of diabetes mellitus and its complications: A mechanistic review. Biomed. Pharmacother. 2017, 91, 31–42. [Google Scholar] [CrossRef] [PubMed]
  37. Alkalbani, N.S.; Osaili, T.M.; Al-Nabulsi, A.A.; Obaid, R.S.; Olaimat, A.N.; Liu, S.-Q.; Ayyash, M.M. In vitro characterization and identification of potential probiotic yeasts isolated from fermented dairy and non-dairy food products. J. Fungi 2022, 8, 544. [Google Scholar] [CrossRef] [PubMed]
  38. Chapot-Chartier, M.-P.; Kulakauskas, S. Cell wall structure and function in lactic acid bacteria. Microb. Cell Factories 2014, 13 (Suppl. 1), S9. [Google Scholar] [CrossRef] [PubMed]
  39. Hahn-Hägerdal, B.; Karhumaa, K.; Larsson, C.U.; Gorwa-Grauslund, M.; Görgens, J.; Van Zyl, W.H. Role of cultivation media in the development of yeast strains for large scale industrial use. Microb. Cell Factories 2005, 4, 31. [Google Scholar] [CrossRef] [PubMed]
  40. Pan, L.; Yu, J.; Mi, Z.; Mo, L.; Jin, H.; Yao, C.; Ren, D.; Menghe, B. A metabolomics approach uncovers differences between traditional and commercial dairy products in Buryatia (Russian Federation). Molecules 2018, 23, 735. [Google Scholar] [CrossRef] [PubMed]
  41. Sarkar, S.; Sha, S.; Ghatani, K. Metabolomics of ethnic fermented foods and beverages: Understanding new aspects through omic techniques. Front. Sustain. Food Syst. 2023, 7, 1040567. [Google Scholar] [CrossRef]
  42. De Miranda, N.M.Z.; De Souza, A.C.; de Souza Costa Sobrinho, P.; Dias, D.R.; Schwan, R.F.; Ramos, C.L. Novel yeasts with potential probiotic characteristics isolated from the endogenous ferment of artisanal Minas cheese. Braz. J. Microbiol. 2023, 54, 1021–1033. [Google Scholar] [CrossRef] [PubMed]
  43. Gibson, G.R.; Hutkins, R.W.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D. 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]
  44. Vilela-Moura, A.; Schuller, D.; Mendes-Faia, A.; Silva, R.D.; Chaves, S.R.; Sousa, M.J.; Côrte-Real, M. The impact of acetate metabolism on yeast fermentative performance and wine quality: Reduction of volatile acidity of grape musts and wines. Appl. Microbiol. Biotechnol. 2011, 89, 271–280. [Google Scholar] [CrossRef] [PubMed]
  45. Abildinova, G.Z.; Benberin, V.V.; Vochshenkova, T.A.; Afshar, A.; Mussin, N.M.; Kaliyev, A.A.; Zhussupova, Z.; Tamadon, A. Global trends and collaborative networks in gut microbiota-insulin resistance research: A comprehensive bibliometric analysis (2000–2024). Front. Med. 2024, 11, 1452227. [Google Scholar] [CrossRef] [PubMed]
  46. Yamashita, H.; Fujisawa, K.; Ito, E.; Idei, S.; Kawaguchi, N.; Kimoto, M.; Hiemori, M.; Tsuji, H. Improvement of obesity and glucose tolerance by acetate in Type 2 diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Biosci. Biotechnol. Biochem. 2007, 71, 1236–1243. [Google Scholar] [CrossRef] [PubMed]
  47. Moffett, J.R.; Puthillathu, N.; Vengilote, R.; Jaworski, D.M.; Namboodiri, A.M. Acetate revisited: A key biomolecule at the nexus of metabolism, epigenetics and oncogenesis—Part 1: Acetyl-CoA, acetogenesis and acyl-CoA short-chain synthetases. Front. Physiol. 2020, 11, 580167. [Google Scholar] [CrossRef] [PubMed]
  48. Nakamura, Y.K.; Janowitz, C.; Metea, C.; Asquith, M.; Karstens, L.; Rosenbaum, J.T.; Lin, P. Short chain fatty acids ameliorate immune-mediated uveitis partially by altering migration of lymphocytes from the intestine. Sci. Rep. 2017, 7, 11745. [Google Scholar] [CrossRef] [PubMed]
  49. Heimann, E.; Nyman, M.; Pålbrink, A.-K.; Lindkvist-Petersson, K.; Degerman, E. Branched short-chain fatty acids modulate glucose and lipid metabolism in primary adipocytes. Adipocyte 2016, 5, 359–368. [Google Scholar] [CrossRef] [PubMed]
  50. Toker, A.; Kucuksen, S.; Kucuk, A.; Cicekler, H. Serum ischemia-modified albumin and malondialdehyde levels and superoxide dismutase activity in patients with fibromyalgia. Clin. Lab. 2014, 60, 1609–1615. [Google Scholar] [CrossRef] [PubMed]
  51. Zaibi, M.S.; Stocker, C.J.; O’Dowd, J.; Davies, A.; Bellahcene, M.; Cawthorne, M.A.; Brown, A.J.; Smith, D.M.; Arch, J.R. Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett. 2010, 584, 2381–2386. [Google Scholar] [CrossRef] [PubMed]
  52. Ives, S.J.; Zaleski, K.S.; Slocum, C.; Escudero, D.; Sheridan, C.; Legesse, S.; Vidal, K.; Lagalwar, S.; Reynolds, T.H. The effect of succinic acid on the metabolic profile in high-fat diet-induced obesity and insulin resistance. Physiol. Rep. 2020, 8, e14630. [Google Scholar] [CrossRef] [PubMed]
  53. Yang, Y.; Luo, L.; Li, Y.; Shi, X.; Li, C.; Chai, J.; Jiang, S.; Zheng, R. Succinic Acid Improves the Metabolism of High-Fat Diet-Induced Mice and Promotes White Adipose Browning. Nutrients 2024, 16, 3828. [Google Scholar] [CrossRef] [PubMed]
  54. Glastras, S.J.; Chen, H.; Teh, R.; McGrath, R.T.; Chen, J.; Pollock, C.A.; Wong, M.G.; Saad, S. Mouse models of diabetes, obesity and related kidney disease. PloS ONE 2016, 11, e0162131. [Google Scholar] [CrossRef] [PubMed]
  55. Muhamed, S.A.; Moussa, E.M.; Aboasy, N.K.; Gaweesh, Y.Y. Effect of 1% malic acid spray on diabetes mellitus-induced xerostomia: A randomized clinical trial. Oral Dis. 2024, 30, 631–638. [Google Scholar] [CrossRef] [PubMed]
  56. Mirzaei, M.; Shavandi, A.; Mirdamadi, S.; Soleymanzadeh, N.; Motahari, P.; Mirdamadi, N.; Moser, M.; Subra, G.; Alimoradi, H.; Goriely, S. Bioactive peptides from yeast: A comparative review on production methods, bioactivity, structure-function relationship, and stability. Trends Food Sci. Technol. 2021, 118, 297–315. [Google Scholar] [CrossRef]
  57. Jung, E.Y.; Lee, H.S.; Choi, J.W.; Ra, K.S.; Kim, M.R.; Suh, H.J. Glucose tolerance and antioxidant activity of Spent Brewer’s yeast hydrolysate with a high content of cyclo-his-pro (CHP). J. Food Sci. 2011, 76, C272–C278. [Google Scholar] [CrossRef] [PubMed]
  58. Zhou, J.B.; Bai, L.; Wang, Y.; Yang, J.K. The benefits and risks of DPP 4-inhibitors vs. sulfonylureas for patients with type 2 diabetes: Accumulated evidence from randomised controlled trial. Int. J. Clin. Pract. 2016, 70, 132–141. [Google Scholar] [CrossRef] [PubMed]
  59. Branco, P.; Francisco, D.; Chambon, C.; Hébraud, M.; Arneborg, N.; Almeida, M.G.; Caldeira, J.; Albergaria, H. Identification of novel GAPDH-derived antimicrobial peptides secreted by Saccharomyces cerevisiae and involved in wine microbial interactions. Appl. Microbiol. Biotechnol. 2014, 98, 843–853. [Google Scholar] [CrossRef] [PubMed]
  60. Lis, M.; Bhatt, S.; Schoenly, N.E.; Lee, A.Y.; Nislow, C.; Bobek, L.A. Chemical genomic screening of a Saccharomyces cerevisiae genomewide mutant collection reveals genes required for defense against four antimicrobial peptides derived from proteins found in human saliva. Antimicrob. Agents Chemother. 2013, 57, 840–847. [Google Scholar] [CrossRef] [PubMed]
  61. Branco, P.; Viana, T.; Albergaria, H.; Arneborg, N. Antimicrobial peptides (AMPs) produced by Saccharomyces cerevisiae induce alterations in the intracellular pH, membrane permeability and culturability of Hanseniaspora guilliermondii cells. Int. J. Food Microbiol. 2015, 205, 112–118. [Google Scholar] [CrossRef] [PubMed]
  62. Gier, S.; Lermen, M.; Schmitt, M.J.; Breinig, F. Substitution of cysteines in the yeast viral killer toxin K1 precursor reveals novel insights in heterodimer formation and immunity. Sci. Rep. 2019, 9, 13127. [Google Scholar] [CrossRef] [PubMed]
  63. Fiedor, J.; Burda, K. Potential role of carotenoids as antioxidants in human health and disease. Nutrients 2014, 6, 466–488. [Google Scholar] [CrossRef] [PubMed]
  64. Watcharawipas, A.; Runguphan, W. Red yeasts and their carotenogenic enzymes for microbial carotenoid production. FEMS Yeast Res. 2023, 23, foac063. [Google Scholar] [CrossRef] [PubMed]
  65. Kotake-Nara, E.; Yonekura, L.; Nagao, A. Effect of glycerophospholipid class on the β-carotene uptake by human intestinal Caco-2 cells. Biosci. Biotechnol. Biochem. 2010, 74, 209–211. [Google Scholar] [CrossRef] [PubMed]
  66. Roehrs, M.; Conte, L.; da Silva, D.T.; Duarte, T.; Maurer, L.H.; de Carvalho, J.A.M.; Moresco, R.N.; Somacal, S.; Emanuelli, T. Annatto carotenoids attenuate oxidative stress and inflammatory response after high-calorie meal in healthy subjects. Food Res. Int. 2017, 100, 771–779. [Google Scholar] [CrossRef] [PubMed]
  67. Rajaei, Z.; Hadjzadeh, M.-A.-R.; Nemati, H.; Hosseini, M.; Ahmadi, M.; Shafiee, S. Antihyperglycemic and antioxidant activity of crocin in streptozotocin-induced diabetic rats. J. Med. Food 2013, 16, 206–210. [Google Scholar] [CrossRef] [PubMed]
  68. Duzguner, V.; Kucukgul, A.; Erdogan, S.; Celik, S.; Sahin, K. Effect of lycopene administration on plasma glucose, oxidative stress and body weight in streptozotocin diabetic rats. J. Appl. Anim. Res. 2008, 33, 17–20. [Google Scholar] [CrossRef]
  69. Xi, L.; Qian, Z.; Xu, G.; Zheng, S.; Sun, S.; Wen, N.; Sheng, L.; Shi, Y.; Zhang, Y. Beneficial impact of crocetin, a carotenoid from saffron, on insulin sensitivity in fructose-fed rats. J. Nutr. Biochem. 2007, 18, 64–72. [Google Scholar] [CrossRef] [PubMed]
  70. Muriach, M.; Bosch-Morell, F.; Alexander, G.; Blomhoff, R.; Barcia, J.; Arnal, E.; Almansa, I.; Romero, F.J.; Miranda, M. Lutein effect on retina and hippocampus of diabetic mice. Free. Radic. Biol. Med. 2006, 41, 979–984. [Google Scholar] [CrossRef] [PubMed]
  71. Buzzini, P.; Innocenti, M.; Turchetti, B.; Libkind, D.; van Broock, M.; Mulinacci, N. Carotenoid profiles of yeasts belonging to the genera Rhodotorula, Rhodosporidium, Sporobolomyces, and Sporidiobolus. Can. J. Microbiol. 2007, 53, 1024–1031. [Google Scholar] [CrossRef] [PubMed]
  72. Li, X.; Cheng, Y.; Li, J.; Liu, C.; Qian, H.; Zhang, G. Torularhodin Alleviates Hepatic Dyslipidemia and Inflammations in High-Fat Diet-Induced Obese Mice via PPARα Signaling Pathway. Molecules 2022, 27, 6398. [Google Scholar] [CrossRef] [PubMed]
  73. Shi, Q.; Wang, H.; Du, C.; Zhang, W.; Qian, H. Tentative identification of torulene cis/trans geometrical isomers isolated from Sporidiobolus pararoseus by high-performance liquid chromatography-diode array detection-mass spectrometry and preparation by column chromatography. Anal. Sci. 2013, 29, 997–1002. [Google Scholar] [CrossRef] [PubMed]
  74. Elfeky, N.; Elmahmoudy, M.; Zhang, Y.; Guo, J.; Bao, Y. Lipid and Carotenoid Production by Rhodotorula glutinis with a Combined Cultivation Mode of Nitrogen, Sulfur, and Aluminium Stress. Appl. Sci. 2019, 9, 2444. [Google Scholar] [CrossRef]
  75. Promdonkoy, P.A.; Watcharawipas, S.; Bubphasawan, K.; Sansatchanon, N.; Suwanakitti; Kocharin, K.; Runguphan, W. Metabolic Engineering of Saccharomyces cerevisiae for Production of Canthaxanthin, Zeaxanthin, and Astaxanthin. J. Fungi 2024, 10, 433. [Google Scholar]
  76. Lobato, R.V.; Silva, V.O.; Andrade, E.F.; Orlando, D.R.; Zangeronimo, M.G.; de Souza, R.V.; Pereira, L.J. Metabolic effects of β-glucans (Saccharomyces cerevisae) per os administration in rats with streptozotocin-induced diabetes. Nutr. Hosp. 2015, 32, 256–264. [Google Scholar]
  77. Palacios-Garcia, A.A.; Yamamoto-Cuevas, J.V.; Abreu-Rosario, C.G.; Moreno-Higareda, R.; Ceron-Trujillo, B.; Ramirez-Ramirez, E. Systematic Review and Meta-Analysis of the Effects of a Polysaccharide-Rich Hydrolysate Derived from Saccharomyces cerevisiae on Body Weight Reduction. 2025. Available online: https://www.preprints.org/manuscript/202505.0491/v1 (accessed on 20 July 2025).
  78. Wang, W.; Zhao, J.; Gui, W.; Sun, D.; Dai, H.; Xiao, L.; Chu, H.; Du, F.; Zhu, Q.; Schnabl, B. Tauroursodeoxycholic acid inhibits intestinal inflammation and barrier disruption in mice with non-alcoholic fatty liver disease. Br. J. Pharmacol. 2018, 175, 469–484. [Google Scholar] [CrossRef] [PubMed]
  79. Hoving, L.R.; van der Zande, H.J.; Pronk, A.; Guigas, B.; van Dijk, K.W.; van Harmelen, V. Dietary yeast-derived mannan oligosaccharides have immune-modulatory properties but do not improve high fat diet-induced obesity and glucose intolerance. PloS ONE 2018, 13, e0196165. [Google Scholar] [CrossRef] [PubMed]
  80. Tanihiro, R.; Sakano, K.; Oba, S.; Nakamura, C.; Ohki, K.; Hirota, T.; Sugiyama, H.; Ebihara, S.; Nakamura, Y. Effects of yeast mannan which promotes beneficial Bacteroides on the intestinal environment and skin condition: A randomized, double-blind, placebo-controlled study. Nutrients 2020, 12, 3673. [Google Scholar] [CrossRef] [PubMed]
  81. Günal-Köroğlu, D.; Karabulut, G.; Mohammadian, F.; Karaca, A.C.; Capanoglu, E.; Esatbeyoglu, T. Production of yeast cell wall polysaccharides-β-glucan and chitin by using food waste substrates: Biosynthesis, production, extraction, and purification methods. Compr. Rev. Food Sci. Food Saf. 2025, 24, e70161. [Google Scholar] [CrossRef] [PubMed]
  82. Chioru, A.; Chirsanova, A.; Dabija, A.; Avrămia, I.; Boiştean, A.; Chetrariu, A. Extraction Methods and Characterization of β-Glucans from Yeast Lees of Wines Produced Using Different Technologies. Foods 2024, 13, 3982. [Google Scholar] [CrossRef] [PubMed]
  83. Duysburgh, C.; Miclotte, L.; Green, J.B.; Watts, K.T.; Sardi, M.I.; Chakrabarti, A.; Khafipour, E.; Marzorati, M. Saccharomyces cerevisiae derived postbiotic alters gut microbiome metabolism in the human distal colon resulting in immunomodulatory potential in vitro. Front. Microbiol. 2024, 15, 1358456. [Google Scholar] [CrossRef] [PubMed]
  84. Honzel, D.; Carter, S.G.; Redman, K.A.; Schauss, A.G.; Endres, J.R.; Jensen, G.S. Comparison of chemical and cell-based antioxidant methods for evaluation of foods and natural products: Generating multifaceted data by parallel testing using erythrocytes and polymorphonuclear cells. J. Agric. Food Chem. 2008, 56, 8319–8325. [Google Scholar] [CrossRef] [PubMed]
  85. Jensen, G.S.; Hart, A.N.; Schauss, A.G. An antiinflammatory immunogen from yeast culture induces activation and alters chemokine receptor expression on human natural killer cells and B lymphocytes in vitro. Nutr. Res. 2007, 27, 327–335. [Google Scholar] [CrossRef] [PubMed]
  86. Moyad, M.A.; Robinson, L.E.; Zawada, E.T., Jr.; Kittelsrud, J.M.; Chen, D.-G.; Reeves, S.G.; Weaver, S.E. Effects of a modified yeast supplement on cold/flu symptoms. Urol. Nurs. 2008, 28, 50–55. [Google Scholar] [PubMed]
  87. Moyad, M.A.; Robinson, L.E.; Zawada, E.T., Jr.; Kittelsrud, J.; Chen, D.-G.; Reeves, S.G.; Weaver, S. Immunogenic yeast-based fermentate for cold/flu-like symptoms in nonvaccinated individuals. J. Altern. Complement. Med. 2010, 16, 213–218. [Google Scholar] [CrossRef] [PubMed]
  88. Moyad, M.A.; Robinson, L.E.; Kittelsrud, J.M.; Reeves, S.G.; Weaver, S.E.; Guzman, A.I.; Bubak, M.E. Immunogenic yeast-based fermentation product reduces allergic rhinitis-induced nasal congestion: A randomized, double-blind, placebo-controlled trial. Adv. Ther. 2009, 26, 795–804. [Google Scholar] [CrossRef] [PubMed]
  89. Moyad, M.A. Brewer’s/baker’s yeast (Saccharomyces cerevisiae) and preventive medicine: Part II. Urol. Nurs. 2008, 28, 73–75. [Google Scholar] [PubMed]
  90. Meadows, A.L.; Hawkins, K.M.; Tsegaye, Y.; Antipov, E.; Kim, Y.; Raetz, L.; Dahl, R.H.; Tai, A.; Mahatdejkul-Meadows, T.; Xu, L. Rewriting yeast central carbon metabolism for industrial isoprenoid production. Nature 2016, 537, 694–697. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of action of acetate on energy metabolism, immunity, and intestinal integrity. Acetate can act directly or indirectly in the intestine. Acetate is converted into Acetyl-CoA (ACoA) by Acyl-CoA Synthetase Short-Chain Family Member 2 (ACSS2), or it can be directly integrated into the Krebs cycle to generate energy, particularly by colon cells, or used as a co-substrate by lysine acetyltransferase to modulate the activity of transcription factors. When AMP levels rise, AMP-activated protein kinase is activated and then, in turn, phosphorylates carbohydrate-responsive element-binding proteins (ChREBPs), leading to a reduction in the transcription of genes involved in carbohydrate metabolism. In addition, acetate induces the proliferation of immune cells with increased levels of interleukin (IL-10) and modulation of the gut microbiota (reduction in the number of pathogenic bacteria). All these phenomena regulate energy metabolism by inhibiting hepatic glucose synthesis, boosting immunity, and helping maintain intestinal integrity.
Figure 1. Mechanisms of action of acetate on energy metabolism, immunity, and intestinal integrity. Acetate can act directly or indirectly in the intestine. Acetate is converted into Acetyl-CoA (ACoA) by Acyl-CoA Synthetase Short-Chain Family Member 2 (ACSS2), or it can be directly integrated into the Krebs cycle to generate energy, particularly by colon cells, or used as a co-substrate by lysine acetyltransferase to modulate the activity of transcription factors. When AMP levels rise, AMP-activated protein kinase is activated and then, in turn, phosphorylates carbohydrate-responsive element-binding proteins (ChREBPs), leading to a reduction in the transcription of genes involved in carbohydrate metabolism. In addition, acetate induces the proliferation of immune cells with increased levels of interleukin (IL-10) and modulation of the gut microbiota (reduction in the number of pathogenic bacteria). All these phenomena regulate energy metabolism by inhibiting hepatic glucose synthesis, boosting immunity, and helping maintain intestinal integrity.
Applmicrobiol 05 00084 g001
Figure 2. Effect of succinic acid on energy metabolism and insulin resistance. UCP1: Uncoupling Protein 1; SUCNR1: Succinate Receptor 1; LDL-C: Low-Density Lipoprotein Cholesterol; TG: Triglycerides; PGC-1α: Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha; p38 MAPK: p38 Mitogen-Activated Protein Kinase; PI3K/PKB: Phosphoinositide 3-Kinase/protein Kinase B.
Figure 2. Effect of succinic acid on energy metabolism and insulin resistance. UCP1: Uncoupling Protein 1; SUCNR1: Succinate Receptor 1; LDL-C: Low-Density Lipoprotein Cholesterol; TG: Triglycerides; PGC-1α: Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha; p38 MAPK: p38 Mitogen-Activated Protein Kinase; PI3K/PKB: Phosphoinositide 3-Kinase/protein Kinase B.
Applmicrobiol 05 00084 g002
Figure 3. Functional impact of yeast β-glucans and mannans on glycaemia, cholesterol, and immune responses. On the left (light blue frame) and right (light pink frame) are the effects of β-glucans and mannans, respectively, in the body. TNF-α: Tumour Necrosis Factor α; ALT: Alanine aminotransferase.
Figure 3. Functional impact of yeast β-glucans and mannans on glycaemia, cholesterol, and immune responses. On the left (light blue frame) and right (light pink frame) are the effects of β-glucans and mannans, respectively, in the body. TNF-α: Tumour Necrosis Factor α; ALT: Alanine aminotransferase.
Applmicrobiol 05 00084 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

Tchamani Piame, L. Bioactive Metabolites from Yeasts Presumptively Qualified as Safe as Functional Agents in the Management of Type 2 Diabetes. Appl. Microbiol. 2025, 5, 84. https://doi.org/10.3390/applmicrobiol5030084

AMA Style

Tchamani Piame L. Bioactive Metabolites from Yeasts Presumptively Qualified as Safe as Functional Agents in the Management of Type 2 Diabetes. Applied Microbiology. 2025; 5(3):84. https://doi.org/10.3390/applmicrobiol5030084

Chicago/Turabian Style

Tchamani Piame, Laverdure. 2025. "Bioactive Metabolites from Yeasts Presumptively Qualified as Safe as Functional Agents in the Management of Type 2 Diabetes" Applied Microbiology 5, no. 3: 84. https://doi.org/10.3390/applmicrobiol5030084

APA Style

Tchamani Piame, L. (2025). Bioactive Metabolites from Yeasts Presumptively Qualified as Safe as Functional Agents in the Management of Type 2 Diabetes. Applied Microbiology, 5(3), 84. https://doi.org/10.3390/applmicrobiol5030084

Article Metrics

Back to TopTop