Next Article in Journal
Oral Intake of L-Ornithine-L-Aspartate Is Associated with Distinct Microbiome and Metabolome Changes in Cirrhosis
Next Article in Special Issue
Combined Evaluation of Geriatric Nutritional Risk Index and Modified Creatinine Index for Predicting Mortality in Patients on Hemodialysis
Previous Article in Journal
A Food-Based Intervention in a Military Dining Facility Improves Blood Fatty Acid Profile
Previous Article in Special Issue
Dietary Exposure to Polychlorinated Biphenyls and Dioxins and Its Relationship to Telomere Length in Subjects Older Than 55 Years from the SUN Project
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential Roles of Probiotics, Resistant Starch, and Resistant Proteins in Ameliorating Inflammation during Aging (Inflammaging)

Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(4), 747; https://doi.org/10.3390/nu14040747
Submission received: 30 December 2021 / Revised: 29 January 2022 / Accepted: 7 February 2022 / Published: 10 February 2022
(This article belongs to the Special Issue Public Health Nutrition and Healthy Aging)

Abstract

:
Aging is typically accompanied by biological and physiological changes that alter cellular functions. Two of the most predominant phenomena in aging include chronic low-grade inflammation (inflammaging) and changes in the gut microbiota composition (dysbiosis). Although a direct causal relationship has not been established, many studies have reported significant reductions in inflammation during aging through well-maintained gut health and microbial balance. Prebiotics and probiotics are known to support gut health and can be easily incorporated into the daily diet. Unfortunately, few studies specifically focus on their significance in reducing inflammation during aging. Therefore, this review summarizes the scientific evidence of the potential roles of probiotics and two types of prebiotics, resistant starch and resistant proteins, in later age. Studies have demonstrated that the oral consumption of bacteria that may contribute to anti-inflammatory response, such as Bifidobacterium spp., Akkermansia munichipilla, and Faecalis praunitzii, contributes significantly to the suppression of pro-inflammatory markers in elderly humans and aged animals. Colonic fermentation of resistant starch and proteins also demonstrates anti-inflammatory activity owing to the production of butyrate and an improvement in the gut microbiota composition. Collectively, probiotics, resistant starch, and resistant proteins have the potential to promote healthy aging.

Graphical Abstract

1. Introduction

Research interest in aging is gradually increasing, as the scientific community attempts to alleviate its detrimental effects by acquiring a more comprehensive understanding of this natural yet unstoppable process. As humans age, cellular functions decline, which leads to various physiological degradations, such as deteriorated cardiovascular structures and functions [1]; increased gut permeability, which increases susceptibility to pathogenic bacterial infections [2]; gastrointestinal tract disorders [3], which alter nutrition absorption and cause malnutrition in the elderly; degraded immune functions [4]; cognitive decline [5]; muscle frailty [6]; and gut dysbiosis [7]. Interestingly, the gut microbiota also coevolve as humans age [8]. Unfortunately, this leads to an imbalance in microbial composition and dominance of pathogenic and unbeneficial bacteria, a condition known as dysbiosis.
Some researchers postulate that dysbiosis also gives rise to chronic low-grade inflammation [7,9], which is linked to age-related chronic diseases, including cancer, dementia, and type 2 diabetes [8], while other researchers suggest that low-grade inflammation leads to dysbiosis [10,11]. Although the causal relationship between chronic low-grade inflammation and microbiota dysbiosis has yet to be definitively elucidated, the consensus is that they are strongly correlated as many studies have shown that a well-maintained gut health can reduce inflammation [7,12,13,14,15].
The gut microbiota composition is determined by many factors, with diet as the most predominant one. Our diet is the main source of nutrition, which is significant for daily bodily functions and the major contributing factor to gut microbiome composition and activity. A diet with low nutritional value tends to imbalance the gut microbial composition and trigger many health problems later in life.
Prebiotics are well established as one of the important substrates for gut microbial fermentation. Among the many sources of prebiotics, dietary fiber is considerably the most common. Resistant starch (RS), which has been classified as a new type of dietary fiber by the Food and Agriculture Organization (FAO), is characterized by a higher butyrate production from its fermentation compared with other dietary fibers [16,17,18,19,20]. Butyrate plays an essential role in providing energy to human colonocytes [21], preventing dysbiosis [22], and suppressing inflammation [23].
On the other hand, resistant proteins (RP), found in soybean, buckwheat, rice, potato, sericin, and eggshell membrane (ESM), also possess similar properties to RS but originate from proteins that are resistant to human proteases. Although they have not been scrutinized as widely as RS yet, several studies have reported their significant contributions to gut health [24,25,26]. Considering the limited reports on RP and gut health, this review is, to our knowledge, the first to highlight their prospects as elderly diet constituents to promote healthy aging.
In addition to prebiotics, probiotics (beneficial living bacteria) can be consumed orally. Some probiotics, such as Lactobacillus acidophilus, Streptococcus thermophilus, and Bifidobacterium lactis, are commonly known. Notably, Bifidobacterium spp., Akkermansia muciniphila, and Faecalibacterium prausnitzii also have high anti-inflammatory activities [27,28], and their oral intervention efficacies in the elderly have been reported.
As recently proclaimed by the United Nations (UN), 2021–2030 is the decade of healthy aging, highlighting the importance of well-being in later life [29]. By introducing and analyzing the potential roles of probiotics that may be involved in anti-inflammatory mechanisms, resistant starch, and resistant proteins in abating chronic low-grade inflammation that occurs in aging, this review is expected to contribute to the UN’s campaign on healthy aging.

2. Methods

For writing this narrative review, we performed a comprehensive search through PubMed and Google Scholar databases until December 2021. The keywords were “inflammation in aging”, “inflammaging”, “chronic low-grade inflammation”, “anti-inflammation” and “probiotics”, “resistant starch” and “aging”, “resistant starch” and “gut health”, “resistant protein” and “inflammation”, “resistant protein” and “gut health”. All in vivo studies in elderly and aged animals, review articles, and clinical studies reporting changes in inflammatory response (pro- and anti-inflammation cytokines) and gut microbiota composition were included. In addition, the World Health Organization or WHO’s website was also referred regarding the proclamation of the United Nations decade of healthy aging [29].

3. The Importance of Gut Health

Over the past decade, concerns regarding gut health have grown. Our intestinal tract, particularly the colon, is the dwelling site for both beneficial and pathogenic microbiota. Approximately 100 trillion microorganisms, mostly bacteria, are present in the human gastrointestinal tract [30]. They are considered master regulators of immune homeostasis, as their absence caused an impaired immune system [31]. While their role in immune system regulation is well established, they also protect the host from pathogens and unwanted microorganisms and produce thousands of metabolites that contribute to fitness and health [32].
The microbiota composition is regarded as a key factor in healthy gut physiology. According to Arumugam et al., the gut microbiota of adult humans is dominated by the phyla Firmicutes and Bacteroidetes, accounting for 90% of the total gut microbiota, with the rest being made up by other groups of bacteria with distinct functions and health benefits [33]. For instance, Akkermansia muciniphila was reported to protect human intestinal epithelial integrity via its outer membrane pili-like protein Amuc 1100 [34]. Studies in animals also showed that A. municiphila has notable anti-inflammatory activity in mice [28], as it supports the colonization of beneficial SCFA-producing bacteria in mice and macaque [35]. Bifidobacterium spp. generates lactate and acetate and reduces the population of inflammation-related microbes in human colon [36]. Toward et al. reported that the population of Bifidobacterium spp. was inversely correlated with the serum levels of inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) [37]. Butyrate-producing bacteria include Clostridium cluster XIVa, Lachnospiraceae bacterium [38], Faecalibacterium prausnitzii [27], Coprococcus spp. [39], and Roseburia spp. [39]. Among these, F. prausnitzii and Roseburia intestinalis have strong anti-inflammatory properties [27,40]. On the other hand, the abundance of some family of bacteria may be associated with health conditions, as lower population of Christensenellaceae family was reported in patients with Crohn’s disease [41,42,43,44], patients with ulcerative colitis [41,42], and people with body mass index (BMI) ≥30 [45].
In contrast, several pathogens and bacteria that may cause inflammation live in the gut, such as Esterichia coli [46], Eggerthella lenta [47], Streptococcusgallolyticus [48], and Enterococcus spp. [49]. An unhealthy diet and lifestyle significantly support the growth of these undesired bacteria, which suppresses the population of beneficial bacteria and increases the risk of various diseases. Hence, maintenance of the beneficial bacteria dominance in the gut is considered a strategy to sustain a healthy life considering their important roles and functions in the body (Table 1).

4. Aging-Related Changes: Gut Microbiota Composition and Chronic Low-Grade Inflammation

There is a considerable amount of literature published on the significant aging-related changes in gut microbiota composition, which greatly influences the general health of the host. Previously, the Western-style diet was the presumed cause of dysbiosis and the consequent aging-related inflammation [54]; in other words, external factors were thought to be the primary cause. However, Ragonnaud and Biragyn suggested recently that microbiota changes could be intrinsic to the aging process after their observations in Chinese and Italian elderly [8]. More interestingly, O’Toole and Jeffrey reported that despite the shift in gut microbiota balance, the core of microbiota did not age per se [55].
As indicated previously, several types of bacteria provide health benefits. Unfortunately, their population tends to decrease with age, and bacteria that promote chronic inflammation may replace them. The likely decrease or even disappearance of bifidobacteria with age has been well documented [56]. A study by Biagi et al. supported this by confirming the populations of bifidobacteria, some members of Firmicutes, including Clostridium clusters IV (Ruminococcus obeum et rel., Roseburia intestinalis et rel., Eubacterium ventriosum et rel., E. rectale et rel., and E. hallii et rel.), and some members of Clostridium cluster XIVa, including Papillibacter cinnamovorans et rel. and F. prausnitzii et rel., decreased in aged individuals and centenarians [12]. Typical beneficial bacteria, which are indicators of healthy aging, identified in Italian centenarians (99–104 years old) and semi-supercentenarians (105–109 years old) decreased during aging [57]. These included the Ruminococcaceae, Lachnospiraceae, and Bacteridaceae families.
In addition to changes in gut microbiota composition, other physiological features also change significantly with age. Reduced organ functions due to aging, such as reduced cardiovascular structures and functions [1], increased gut permeability [2], gastrointestinal tract disorder [3], and degradation of immune function [4], have been widely reported. The degradation of immune function during aging is also termed immunosenescence and is suggested to be related to chronic low-grade inflammation or inflammaging. Chronic low-grade inflammation refers to an extended increase in pro-inflammatory factors at a level lower than typically observed in acute infections [58]. Figure 1 illustrates some mechanisms by which aging may contribute to chronic low-grade inflammation.
While studies on inflammation commonly use the pro-inflammatory cytokine interleukin-6 (IL-6) as a marker, the majority of studies investigating low-grade inflammation have utilized the surrogate marker high-sensitivity C-reactive protein (hsCRP) because of its extreme sensitivity to systemic inflammation [59,60,61]. Elevated IL-6 or hsCRP levels in the absence of an acute infection or other inflammatory stimuli indicate maladaptive chronic low-grade inflammation [62]. With advancing age, higher hsCRP and IL-6 levels are observed [61,63].
Figure 1. Illustration of the various mechanisms by which aging may activate inflammatory pathways [64]. An elevated NF-κB DNA binding activity increases the expression of the upstream kinases of NF-κB [65,66]. (2) Mitophagy and mtDNA mutation [67]. (3) Cell senescence activates DNA damage response (DDR) and senescence-associated secretory phenotype (SASP, which is a variation in phenotype induced by cell senescence) [68]. (4) Immunosenescence increases susceptibility of foreign materials invasion and activate the inflammatory response [69].
Figure 1. Illustration of the various mechanisms by which aging may activate inflammatory pathways [64]. An elevated NF-κB DNA binding activity increases the expression of the upstream kinases of NF-κB [65,66]. (2) Mitophagy and mtDNA mutation [67]. (3) Cell senescence activates DNA damage response (DDR) and senescence-associated secretory phenotype (SASP, which is a variation in phenotype induced by cell senescence) [68]. (4) Immunosenescence increases susceptibility of foreign materials invasion and activate the inflammatory response [69].
Nutrients 14 00747 g001
Chronic inflammation is a potential driver of metabolic disorders and age-related decline in physical functions [6], such as reduced mobility and impaired cognitive functions [5], and age-related diseases, such as cancer, depression, sarcopenia, and disability [70,71,72].
Many natural changes in the human body are hallmarks of aging, while chronic low-grade inflammation is one of the major contributing factors for various age-related diseases and disabilities in the elderly. Microbial composition dominated by bacteria that may promote inflammation also aggravates chronic inflammation, highlighting the profound effects of the gut microbiota on the health of the elderly.

5. How Gut Health Influences Inflammaging

The cause-and-effect relationship between inflammaging and gut health, that is, whether gut dysbiosis is the cause or consequence of inflammaging, remains unclear. Although there have been many studies, results are still inconclusive due to inconsistency. A study by Byndloss et al., for instance, suggested that the decrease in SCFAs altered the growth of beneficial bacteria and thus represented an opportunity for pathogens and bacteria that may induce inflammation to take over the gut colony [22]. In an aged mouse model, this microbiota transformation promoted dysbiosis and gut leakage, resulting in activated innate immune response or inflammation [9]. Age-related microbial dysbiosis is considered to contribute to systemic inflammation in humans [7]. A study by Nastasi et al. supported this finding, as the gut microbiota metabolites, particularly butyrate and propionate, reduced the pro-inflammatory chemokines by modulating the immune system in dendritic cells [73].
However, a contrasting result was described in a study by Thevaranjan et al. [11]. TNF-knockout old mice (>18 months old, equivalent to 60 years old human [74]) did not experience dysbiosis when the wild-type (WT) did, and the microbiome of old (but not young) mice changed after anti-TNF treatment. Similarly, Chen et al. indicated that dysbiosis occurred in mice that were deficient in the negative regulator of inflammatory signaling in a TNF- and IL-6-dependent manner [10]. Both studies proposed that inflammation triggered dysbiosis, notably in old mice.
Notwithstanding the cause-and-effect relationship between inflammaging and gut dysbiosis, the consensus is that they are intertwined. Although the causal relationship is complex, evidence supports the notion that healthy gut environment can reduce inflammation during aging.

6. Reducing Inflammaging by Improving Gut Health

There are multiple ways to improve gut health. Here, however, we focus on the consumption of butyrate-producing probiotics or probiotics with potential anti-inflammatory activities, resistant starch, and resistant proteins. Notably, their prospective impacts on aging-related chronic low-grade inflammation have been elaborated on. To the best of our knowledge, no previous review has addressed this topic. Based on this discussion, we expect to introduce a new approach to promote gut health through probiotics, resistant starch, and resistant proteins (Figure 2), which can be intensively investigated in the future.

6.1. Intervention of Probiotics with Potential Anti-Inflammatory Effects or Butyrate-Producing Probiotics

One type of beneficial metabolite produced by gut bacteria is short-chain fatty acids (SCFAs), which are produced by colonic fermentation of indigestible fibers [75]. The major SCFAs produced are acetate, propionate, and butyrate. Acetate is the most abundant and essential metabolite for the growth of other bacteria [52]. Butyrate mostly functions as the main energy source for human colonocytes [21], and it is essential for epithelial cells to prevent dysbiosis by maintaining the oxygen balance in the gut [22]. Liu et al. and Nastasi et al. also reported the anti-inflammatory properties of butyrate [23,73].
There is ample evidence that a high-fiber diet increases the production of SCFAs via microbial fermentation [76,77,78]. These organic acids were proven to improve survival against influenza in mice by inhibiting neutrophil recruitment and airway inflammation [50]. This may be explained by direct stimulation of immune cells, for example, by elevating both intestinal IgA and systemic IgG responses to curb pathogens [51].
SCFAs provide energy for beneficial bacteria, enterocytes, and immune cells. Ragonnaud and Biragyn stated that SCFAs could cross the gut epithelium layer to induce regulatory T cells (Tregs) and IL-10-producing T cells [8]. This activates anti-inflammatory responses in antigen-presenting cells and promotes the production of IgA and IgG by B cells, which increases immune tolerance.
As noted in the previous section, several species of bacteria with different functions are typically found in centenarians. In addition to bacteria that may attenuate inflammation, bacteria that produce SFCAs also contribute to healthy aging. Several recent reports revealed that the supplementation of some strains in the diet ameliorated age-related loss of mucin and demonstrated health and immune benefits in elderly and aged animals [28,79,80]. In addition to probiotic supplementation, fecal microbiota transplantation also effectively extends the lifespan of progeroid mice [81].
On the other hand, some probiotics, such as Lactobacillus spp. and bifidobacteria, are common supplements in dairy products or other functional foods. Detailed work by Maneerat et al. highlighted that the consumption of Bifidobacterium lactis Bi-07 could potentially strengthen the innate immune response in an elderly population without contributing to inflammation-related disorders [82]. The intervention by another species, B. longum, in healthy elderly people (65–90 years old) also contributed significantly to the suppression of inflammatory markers TNF-α and the increase in bifidobacteria (specifically B. adolescentis, B. angulatum, B. bifidum, and B. longum), fecal butyrate, and acetate concentrations [83]. The suppressing activities of B. bifidum BGN4 and B. longum BORI against bacteria with pro-inflammatory effects in the elderly (≥65 years old) led to the improvement of probiotic composition in the gut, as reported recently by Kim et al. [84].
Akkermansia muciniphila is another butyrate-producing bacterium with notable anti-inflammatory activities. Previous reports have established its anti-inflammatory activity in glucose-intolerant, obese, and aged animal models [79,85,86], as well as in humans [87,88]. In a study of accelerated aging Ercc1−/Δ7 mice, a decrease in mucus thickness was observed as an outcome of aging, whereas mice supplemented with A. muciniphila MucT (ATTC BAA-835) for 10 weeks (three times/week) showed a thicker layer due to the activation of colonic mucus production [28]. A thick mucus layer acts as a vital barrier against bacterial penetration, which causes inflammation. A. muciniphila supplementation also downregulated the expression of multiple genes and pathways related to inflammation and immune functions in both the colon and ileum. Ingenuine Pathway Analysis (IPA) predicted the inhibition of several inflammation-related factors upon supplementation, including pro-inflammatory IL-1, TNF receptor superfamily members 1B and 12, nuclear factor kappa B (NF-κB) inhibitor α, T-cell receptor (TCR), and Toll-like receptor adaptor molecule 1 (TICAM1). Although the anti-inflammatory properties of A. muciniphila have been extensively described previously [89], the study confirmed its protection against aging-related inflammation by reducing colonic expression of pro-inflammatory genes [28].
Like A. muciniphila, Faecalibacterium praunitzii is a butyrate-producing bacterium that may potentially contribute to the anti-inflammatory response. It mitigates inflammation in vitro and in vivo by blocking NF-κB activation and IL-8 production [90]. This is supported by significantly lower concentrations of IL-6, interferon gamma (IFN-γ), and IL-4 in the colon, and IL-6 and IL-22 in serum after 10-day supplementation in mice with dinitrobenzene sulfonic acid (DNBS)-induced chronic low-grade inflammation [91]. An association between this anti-inflammatory activity and butyrate production was hypothesized [91]. While the anti-inflammatory effects of F. praunitzii were widely reported previously, its protective activity against chronic low-grade inflammation was first revealed in mice by this seminal research.
However, the supplementation of probiotics may lead to safety issues, as not all bacteria have been clinically tested for oral consumption. Furthermore, some bacteria function in a diet-dependent manner, such as Prevotella copri [92,93], and the best way to enrich their colonization in the colon is by consuming more prebiotics. A study of gut microbiota composition in 168 long-lived Chinese (>90 years old) suggested alpha diversity, which depicts the abundance and evenness of species within a host [94], as a reasonable predictor of longevity [95]. This indicates that dietary and other interventions to maintain or promote diversity might be worth pursuing for healthy aging [7]. However, this view requires further scientific support. Another concern in probiotic supplementation is the viability of probiotics, which can survive in the human digestive system only long enough to reach the colon. Considering the above, incorporating prebiotics into our diets in addition to the oral consumption of probiotics may be a good strategy to increase gut microbiota diversity.

6.2. Prebiotics Intervention

Dietary fiber is widely regarded as an excellent source of prebiotics that can be naturally obtained from food, such as fruits, vegetables, whole grains, legumes, and nuts. Data from multiple sources have shown that a high-fiber diet is positively correlated with the enrichment of beneficial bacteria [96,97,98]. Similar to fermenting fiber, gastrointestinal microflora degrades other indigestible starch and protein fractions to produce SCFAs. These materials are referred to as resistant starches and resistant proteins. In recent years, research interests in resistant starch and resistant protein have been on the rise, and their potential benefits to human healthy aging have been demonstrated and warrant further discussion.

6.2.1. Resistant Starch and Its Consumption in Elderly and Aged Animals

Most previous studies have defined resistant starch (RS) as a type of dietary fiber resistant to hydrolysis by human digestive enzymes. As it is indigestible, RS can reach the large intestine and is fermented by the gut microbiota to produce SCFAs. In other words, RS is a type of prebiotic. It is worth noting that the fermentation of resistant starch produces more butyrate than dietary fiber [16,17,18,19,20].
A recent in vitro study utilizing static batch fermentation with fecal slurry method re-confirmed that gut microbial fermentation of RS resulted in the highest accumulation of butyric acid compared to other dietary fibers [19]. This is because only RS enriches butyrate-producing bacteria that belong to Bifidobacterium, Ruminococcus, and Roseburia genera [19]. This is supported by research findings that Ruminococcus bromii and Bifidobacterium spp. degrade resistant starch more efficiently than other species [99,100,101]. Based on these findings and our focus on aging-related inflammation, we discuss resistant starch instead of other common prebiotics. The benefits of resistant starch in improving gut health have been reported, but to our knowledge, a review on its consumption and effects in the elderly has never been conducted previously.
RS is a common natural component of several types of food that can be categorized into four types based on its physical and chemical characteristics. RS1, generally found in whole grains and legumes, is an entrapped starch in a non-digestible matrix [102]. RS2 refers to ungelatinized starch granules, such as starch in raw potato and high-amylose cornstarch. The FDA has approved Hi-Maize resistant starch (naturally produced from modified high-amylose corn) for use in patients with type 2 diabetes [103]. RS3 consists of starch that has already undergone retrogradation (starch is cooled down after gelatinization). RS4, found in bread, includes starch that is chemically modified by adding ester or ether groups [102].
The correlation between RS consumption and gut health, inflammatory markers, insulin response, and lipid metabolism has been well documented. However, studies with elderly subjects are limited. We summarize relevant research findings here (Table 2) to provide an overview of the benefits of RS at later age.
RS should be considered for elderly diets because it can increase the population of beneficial bacteria and butyrate production according to multiple studies. In a serial study, MSPrebiotic®, a commercial RS containing 70% RS2 from Solanum tuberosum extract, promoted the growth of bifidobacteria and ameliorated dysbiosis related to the high abundance of Proteobacteria in subjects ≥70 years old [104]. Accordingly, changes in the levels of inflammatory markers (IL-10, C-reactive protein, and TNF-α) in blood were observed. Other investigations have shown that even though RS was able to reduce dysbiosis in the elderly, the inflammatory levels remained elevated at the end of the study [105]. It was hypothesized that three-month consumption of RS is not enough to alleviate the increased inflammatory response caused by colonocyte apoptosis, and earlier intervention of prebiotics in the diet (before 70 years of age) is required to prevent bowel damage, permeability, and the associated increase in non-specific inflammatory markers.
A similar result was found in a recent study using 18-month-old mice, reporting the therapeutic effects of RS2 against high-fat diet-induced and aging-related dysfunctions [103]. According to this study, RS2 effectively decreased the expression of systemic endotoxemia and pro-inflammatory cytokines, as evidenced by lower levels of serum and fecal lipopolysaccharides (LPS), which is an endotoxic component in the cell membrane of Gram-negative bacteria that induces inflammatory response, colonic IL-2, and hepatic IL-4. This corroborated the anti-inflammatory properties of RS2 against aging-related chronic low-grade inflammation. RS2 also enhanced gut barrier function, which was marked by increased expression of colonic mucin 2 at both the mRNA and protein levels. Moreover, this study revealed that RS2 reduced the abundance of pathogenic taxa associated with obesity, inflammation, and aging, such as Desulfovibrio (phylum Proteobacteria), Ruminiclostridium 9, Lachnoclostridium, Helicobacteria, Oscillibacter, Alistipes, Peptococcus, and Rikenella [103]. Collectively, studies both in humans and in mice support the view that RS2 can significantly improve gut health at a later age.
Changes in gut microbiota diversity in aged organisms are presumed to alter the microbial fermentation ability in the colon. To confirm this, Zhou et al. conducted a comprehensive study to examine RS tolerability in healthy aged mice (18 to 20 months old, analogous to 60–66 years old in humans [74]) and concluded that up to 36% of high-amylose-maize-resistant starch type 2 (HAMRS2) diets were well tolerated and, more importantly, could be fermented by aged mice in a dose-responsive manner as in young mice [106]. Furthermore, HAMRS2 increased colonic proglucagon expression and adiponectin levels in visceral fat, indicating improved insulin sensitivity in visceral fat. Increased cecal proglucagon expression is always linked to the elevation of circulating glucagon-like peptide 1 (GLP-1) in young rats, and GLP-1 can recuperate aging-related decline in glucose tolerance [109]. An identical outcome was established in a study conducted by Tachon et al. with the same mouse model [107]. They claimed that the improved microbiota composition (higher levels of Bacteroidetes, Bifidobacterium spp., Akkermansia spp., and Allobaculum spp.) in aged mice after 10-week supplementation with HAMRS2 was positively correlated with the expression levels of proglucagon.
Peixoto et al. investigated the effects of corn-based RS consumption in 11.5-year-old dogs [108]. The results showed that butyrate and total SCFA concentrations increased by approximately 49% and 36%, respectively, in the feces of dogs fed with high RS. Inflammation levels in the gut mucosa were also examined, but the differences were not significant. Nevertheless, this does not diminish the finding, as Swanson et al. pointed out that not all fermentable substrates, when included in diets, induce higher fecal butyrate levels [110]. This claim is supported by a study showing that the consumption of whole wheat-based RS infused into biscuits together with a canned diet did not elevate fecal butyrate levels in dogs [111].
Previous studies both in aged humans and in animals showed that RS consumption may contribute to the improvement of gut health and gut microbiota diversity; however, the results may depend on the host’s gut microbiota composition since it may vary between humans and between humans and animals. Moreover, individual intake should be monitored due to variance in intestinal physiology as the overconsumption of RS may cause diarrhea, vomiting, or soft stools. In other words, RS tolerance may vary among individuals. While some previous studies included tolerance tests for subjects, they were for reference only because the tolerance may vary drastically between animals and humans.

6.2.2. Resistant Proteins

Resistant protein is not as well defined as resistant starch in the literature. In this review, resistant proteins (RP) refer to protein fractions indigestible by the human enzymatic digestive system that enter the colon and are fermented by the gut microbiota. They are different from undigested proteins, which refer to dietary proteins that escaped digestion or absorption in the small intestine. It is important to discern these two types of proteins because the metabolites and effects on the host are significantly different even though both are fermented in the colon. The fermentation of undigested dietary proteins primarily results in toxic products, such as ammonia, amines, phenols, and sulfides, which are associated with gastrointestinal tract diseases, particularly colorectal cancer and ulcerative colitis [112]. On the other hand, RP are fermented in the same manner as dietary fiber, which produces SCFAs and other metabolites derived from protein fractions that are indigestible by the human digestive system.
RP are found naturally in food, particularly plant-based foods, such as soybean, buckwheat, rice, and potato. Research on RP is scarce to date; thus the commonly known RPs are still limited to buckwheat protein, sericin, and the recently revealed eggshell membrane (ESM). Interest in RPs grew initially when the undigested high molecular fraction (HMF) of soy protein isolate (SPI) was found to be 25% less effective as a protein source compared to the SPI, and its consumption led to substantial bile acid binding by the post-digestive remnants of HMF and the subsequent elimination together with other excreta [113]. This bile acid-binding property is responsible for the hypocholesterolemic effect of RP, with a similar mechanism to that of dietary fiber and resistant starch.
Further investigation of RP led to the identification of sericin. Sericin is a natural macromolecular protein derived from the silkworm Bombyx mori and constitutes 15–35% of silk proteins [114]. Sasaki et al. reported that sericin, a silk protein, had low digestibility in the digestive system and a high water-holding capacity, and these properties led to the successful reduction of atropine-induced constipation in rats [26]. This suggested the effects of sericin, as a resistant protein, in mitigating constipation that were similar to those of dietary fiber.
According to Okazaki et al., sericin can be considered a prebiotic because it promotes colon health by modulating the immune response and intestinal barrier function [115]. They investigated the effects of sericin supplementation at 40 g/kg body weight in rats in a high-fat diet and observed elevated levels of fecal IgA and the presence of fecal mucins. As the gut barrier function weakens with age, sericin may be used to stimulate the immune system in the elderly. However, other typical prebiotic properties, such as promoting gut microbiota balance or acting as a substrate for beneficial microbial fermentation, have not been further investigated in sericin.
Recently, our research group identified eggshell membrane (ESM) as a resistant protein because of its low digestibility (approximately 46%) based on a study in rats [24]. ESM is a by-product of egg with interesting bioactivities, including anti-inflammatory activity, skin- and joint-health-promoting functions, and wound-healing properties [116,117,118]. We showed that ESM, as a resistant protein, could stimulate cecal fermentation and alter intestinal bacterial composition [24]. Dextran sodium sulphate (DSS)-induced IBD mice exhibited reduced richness in colon microbiota and dysbiosis. This condition, however, was alleviated by the ingestion of 8% ESM for 7 days. The populations of SCFA-producing bacteria, notably from the family of Ruminococcaceae and Porphyromonadaceae, which were principally decreased by intestinal inflammation, were restored. Considering the significant improvements in the gut environment after ESM intake, microbial diversity may be a major contributor to the anti-inflammatory activity of ESM.
Consistent with this finding, our research further confirmed that ESM could improve the inflammatory response by modulating gut microbiota composition [25]. Supplementation with 8% ESM in high-fat diet (HFD)-fed mice significantly suppressed the proliferation of inflammation-related bacteria (Roseburia faecis, Ruminococcus callidus, and Blautia hydrogenotrophica) and obesity-related bacteria (Blautia coccoides and Parabacteroides goldsteinii) at 20 weeks. Although the concentrations of cecal acetate, lactate, butyrate, and propionate were not affected by either the high-fat or the ESM diet, cecal isobutyrate levels were higher in the ESM group than in the HFD and control groups. The negative correlations between the abundance of P. goldsteinii and B. hydrogenotrophica and isobutyrate concentration suggest that high isobutyrate levels are unfavorable to the obesity-related bacteria, which is consistent with previous findings by Wang et al. [119]. Collectively, these studies provide important evidence that ESM, as a novel source of resistant proteins, has the potential to reduce dysbiosis, which is a determining factor for healthy aging. In the future, we are interested in investigating the anti-inflammatory effectiveness of ESM against aging-related chronic low-grade inflammation.

7. Future Studies

In studies using aged humans and animals, probiotics, resistant starch, and resistant proteins show potential effects in promoting gut health and mitigating inflammation in aging. However, there are differences in the gut microbiota composition between humans and animals. Even between humans, the gut microbiota may be influenced by gender, lifestyle, and geographical factors. Therefore, studies investigating the role of probiotics, resistant starch, and resistant proteins by considering these factors can be addressed in the future. The findings in animal experiments we have summarized in this review can be a basis for further clinical studies to provide more empirical data. Underscoring the prospective role of resistant protein as a prebiotic, we have interest in continuing our work on ESMs. Future studies may investigate the mechanism of how ESMs may improve gut microbiota composition or how they may contribute to the production of beneficial metabolites, such as short-chain fatty acids. Considering its potential anti-inflammatory activity, the relationship between ESM intake and immune system, particularly in relation to aging to promote healthy aging, is worth a further investigation.

8. Conclusions

Multiple studies have demonstrated that typical probiotics possessing anti-inflammatory activities and resistant starch can abate aging-related chronic low-grade inflammation in humans and animal models. The main mechanisms include the direct increase of bacterial population, particularly bifidobacteria, A. muciniphila, and F. prautnitzii, and the supply of substrates for colonic microbial fermentation in the form of indigestible starch. Besides the efficacy, the dosage and consumption frequency of resistant starch should be monitored, particularly for individuals with sensitive digestive tracts or low tolerance to dietary fiber. On the other hand, despite limited studies on resistant proteins and their interventions in the elderly, recent studies have disseminated their fermentabilities to produce SCFAs and promising potential in ameliorating gut dysbiosis and thus their consequent protection against inflammation. All findings elaborated in this review are expected to trigger further exploration of resistant proteins, probiotics, and resistant starch in maintaining gut health, targeting inflammaging, and promoting healthy aging.

Author Contributions

Conceptualization, H.J.; writing original draft preparation, D.J.W.; writing review and editing, H.J. and H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fleg, J.L.; Strait, J. Age-associated changes in cardiovascular structure and function: A fertile milieu for future disease. Heart Fail. Rev. 2012, 17, 545–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Finlay, B.B.; Pettersson, S.; Melby, M.K.; Bosch, T.C.G. The Microbiome Mediates Environmental Effects on Aging. Bioessays 2019, 41, e1800257. [Google Scholar] [CrossRef]
  3. Dumic, I.; Nordin, T.; Jecmenica, M.; Stojkovic Lalosevic, M.; Milosavljevic, T.; Milovanovic, T. Gastrointestinal Tract Disorders in Older Age. Can. J. Gastroenterol. Hepatol. 2019, 2019, 6757524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
  5. Greco, E.A.; Pietschmann, P.; Migliaccio, S. Osteoporosis and Sarcopenia Increase Frailty Syndrome in the Elderly. Front. Endocrinol. (Lausanne) 2019, 10, 255. [Google Scholar] [CrossRef]
  6. Xu, Y.; Liu, X.; Liu, X.; Chen, D.; Wang, M.; Jiang, X.; Xiong, Z. The Roles of the Gut Microbiota and Chronic Low-Grade Inflammation in Older Adults with Frailty. Front. Cell Infect. Microbiol. 2021, 11, 675414. [Google Scholar] [CrossRef]
  7. DeJong, E.N.; Surette, M.G.; Bowdish, D.M.E. The Gut Microbiota and Unhealthy Aging: Disentangling Cause from Consequence. Cell Host Microbe 2020, 28, 180–189. [Google Scholar] [CrossRef]
  8. Ragonnaud, E.; Biragyn, A. Gut microbiota as the key controllers of “healthy” aging of elderly people. Immun Ageing 2021, 18, 2. [Google Scholar] [CrossRef]
  9. Bauernfeind, F.; Niepmann, S.; Knolle, P.A.; Hornung, V. Aging-Associated TNF Production Primes Inflammasome Activation and NLRP3-Related Metabolic Disturbances. J. Immunol. 2016, 197, 2900–2908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Chen, L.; Wilson, J.E.; Koenigsknecht, M.J.; Chou, W.C.; Montgomery, S.A.; Truax, A.D.; Brickey, W.J.; Packey, C.D.; Maharshak, N.; Matsushima, G.K.; et al. NLRP12 attenuates colon inflammation by maintaining colonic microbial diversity and promoting protective commensal bacterial growth. Nat. Immunol. 2017, 18, 541–551. [Google Scholar] [CrossRef] [PubMed]
  11. Thevaranjan, N.; Puchta, A.; Schulz, C.; Naidoo, A.; Szamosi, J.C.; Verschoor, C.P.; Loukov, D.; Schenck, L.P.; Jury, J.; Foley, K.P.; et al. Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe 2017, 21, 455–466.e454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Biagi, E.; Nylund, L.; Candela, M.; Ostan, R.; Bucci, L.; Pini, E.; Nikkila, J.; Monti, D.; Satokari, R.; Franceschi, C.; et al. Through ageing, and beyond: Gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE 2010, 5, e10667. [Google Scholar] [CrossRef]
  13. Kim, M.S.; Hwang, S.S.; Park, E.J.; Bae, J.W. Strict vegetarian diet improves the risk factors associated with metabolic diseases by modulating gut microbiota and reducing intestinal inflammation. Environ. Microbiol. Rep. 2013, 5, 765–775. [Google Scholar] [CrossRef] [PubMed]
  14. Moro-Garcia, M.A.; Alonso-Arias, R.; Baltadjieva, M.; Fernandez Benitez, C.; Fernandez Barrial, M.A.; Diaz Ruisanchez, E.; Alonso Santos, R.; Alvarez Sanchez, M.; Saavedra Mijan, J.; Lopez-Larrea, C. Oral supplementation with Lactobacillus delbrueckii subsp. bulgaricus 8481 enhances systemic immunity in elderly subjects. Age (Dordr.) 2013, 35, 1311–1326. [Google Scholar] [CrossRef]
  15. Roager, H.M.; Vogt, J.K.; Kristensen, M.; Hansen, L.B.S.; Ibrugger, S.; Maerkedahl, R.B.; Bahl, M.I.; Lind, M.V.; Nielsen, R.L.; Frokiaer, H.; et al. Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: A randomised cross-over trial. Gut 2019, 68, 83–93. [Google Scholar] [CrossRef] [Green Version]
  16. Cummings, J.H.; Macfarlane, G.T.; Englyst, H.N. Prebiotic digestion and fermentation. Am. J. Clin. Nutr. 2001, 73, 415S–420S. [Google Scholar] [CrossRef]
  17. Smith, J.G.; Yokoyama, W.H.; German, J.B. Butyric acid from the diet: Actions at the level of gene expression. Crit. Rev. Food Sci. Nutr. 1998, 38, 259–297. [Google Scholar] [CrossRef]
  18. Topping, D.L.; Clifton, P.M. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiol. Rev. 2001, 81, 1031–1064. [Google Scholar] [CrossRef]
  19. Tsitko, I.; Wiik-Miettinen, F.; Mattila, O.; Rosa-Sibakov, N.; Seppanen-Laakso, T.; Maukonen, J.; Nordlund, E.; Saarela, M. A Small In Vitro Fermentation Model for Screening the Gut Microbiota Effects of Different Fiber Preparations. Int. J. Mol. Sci. 2019, 20, 1925. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, X.; Gibson, G.R. Effects of the in vitro fermentation of oligofructose and inulin by bacteria growing in the human large intestine. J. Appl. Bacteriol. 1993, 75, 373–380. [Google Scholar] [CrossRef]
  21. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Backhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Byndloss, M.X.; Olsan, E.E.; Rivera-Chavez, F.; Tiffany, C.R.; Cevallos, S.A.; Lokken, K.L.; Torres, T.P.; Byndloss, A.J.; Faber, F.; Gao, Y.; et al. Microbiota-activated PPAR-gamma signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017, 357, 570–575. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, X.; Wu, Y.; Li, F.; Zhang, D. Dietary fiber intake reduces risk of inflammatory bowel disease: Result from a meta-analysis. Nutr. Res. 2015, 35, 753–758. [Google Scholar] [CrossRef] [PubMed]
  24. Jia, H.; Hanate, M.; Aw, W.; Itoh, H.; Saito, K.; Kobayashi, S.; Hachimura, S.; Fukuda, S.; Tomita, M.; Hasebe, Y.; et al. Eggshell membrane powder ameliorates intestinal inflammation by facilitating the restitution of epithelial injury and alleviating microbial dysbiosis. Sci. Rep. 2017, 7, 43993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ramli, N.S.; Jia, H.; Sekine, A.; Lyu, W.; Furukawa, K.; Saito, K.; Hasebe, Y.; Kato, H. Eggshell membrane powder lowers plasma triglyceride and liver total cholesterol by modulating gut microbiota and accelerating lipid metabolism in high-fat diet-fed mice. Food Sci. Nutr. 2020, 8, 2512–2523. [Google Scholar] [CrossRef] [Green Version]
  26. Sasaki, M.Y.H.; Kato, N. A resistant protein, sericin improves atropine-induced constipation in rats. Food Sci. Technol. Res. 2000, 6, 280–283. [Google Scholar] [CrossRef] [Green Version]
  27. Ferreira-Halder, C.V.; Faria, A.V.S.; Andrade, S.S. Action and function of Faecalibacterium prausnitzii in health and disease. Best Pract. Res. Clin. Gastroenterol. 2017, 31, 643–648. [Google Scholar] [CrossRef]
  28. van der Lugt, B.; van Beek, A.A.; Aalvink, S.; Meijer, B.; Sovran, B.; Vermeij, W.P.; Brandt, R.M.C.; de Vos, W.M.; Savelkoul, H.F.J.; Steegenga, W.T.; et al. Akkermansia muciniphila ameliorates the age-related decline in colonic mucus thickness and attenuates immune activation in accelerated aging Ercc1 (-/Delta7) mice. Immun Ageing 2019, 16, 6. [Google Scholar] [CrossRef]
  29. World Health Organization. UN Decade of Healthy Aging 2021-2030. Available online: https://www.who.int/initiatives/decade-of-healthy-ageing#:~:text=The%20United%20Nations%20Decade%20of,improve%20the%20lives%20of%20older (accessed on 10 December 2021).
  30. Bull, M.J.; Plummer, N.T. Part 1: The Human Gut Microbiome in Health and Disease. Integr. Med. (Encinitas) 2014, 13, 17–22. [Google Scholar]
  31. Honda, K.; Littman, D.R. The microbiota in adaptive immune homeostasis and disease. Nature 2016, 535, 75–84. [Google Scholar] [CrossRef]
  32. Rath, C.M.; Dorrestein, P.C. The bacterial chemical repertoire mediates metabolic exchange within gut microbiomes. Curr. Opin. Microbiol. 2012, 15, 147–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef] [PubMed]
  34. Ottman, N.; Reunanen, J.; Meijerink, M.; Pietila, T.E.; Kainulainen, V.; Klievink, J.; Huuskonen, L.; Aalvink, S.; Skurnik, M.; Boeren, S.; et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 2017, 12, e0173004. [Google Scholar] [CrossRef]
  35. Bodogai, M.; O’Connell, J.; Kim, K.; Kim, Y.; Moritoh, K.; Chen, C.; Gusev, F.; Vaughan, K.; Shulzhenko, N.; Mattison, J.A.; et al. Commensal bacteria contribute to insulin resistance in aging by activating innate B1a cells. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef]
  36. O’Callaghan, A.; van Sinderen, D. Bifidobacteria and Their Role as Members of the Human Gut Microbiota. Front. Microbiol. 2016, 7, 925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Toward, R.; Montandon, S.; Walton, G.; Gibson, G.R. Effect of prebiotics on the human gut microbiota of elderly persons. Gut Microbes 2012, 3, 57–60. [Google Scholar] [CrossRef] [Green Version]
  38. Haran, J.P.; Bucci, V.; Dutta, P.; Ward, D.; McCormick, B. The nursing home elder microbiome stability and associations with age, frailty, nutrition and physical location. J. Med. Microbiol. 2018, 67, 40–51. [Google Scholar] [CrossRef]
  39. Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [Green Version]
  40. Shen, Z.; Zhu, C.; Quan, Y.; Yang, J.; Yuan, W.; Yang, Z.; Wu, S.; Luo, W.; Tan, B.; Wang, X. Insights into Roseburia intestinalis which alleviates experimental colitis pathology by inducing anti-inflammatory responses. J. Gastroenterol. Hepatol. 2018, 33, 1751–1760. [Google Scholar] [CrossRef]
  41. Alam, M.T.; Amos, G.C.A.; Murphy, A.R.J.; Murch, S.; Wellington, E.M.H.; Arasaradnam, R.P. Microbial imbalance in inflammatory bowel disease patients at different taxonomic levels. Gut Pathog. 2020, 12, 1. [Google Scholar] [CrossRef]
  42. Imhann, F.; Vich Vila, A.; Bonder, M.J.; Fu, J.; Gevers, D.; Visschedijk, M.C.; Spekhorst, L.M.; Alberts, R.; Franke, L.; van Dullemen, H.M.; et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018, 67, 108–119. [Google Scholar] [CrossRef]
  43. Pascal, V.; Pozuelo, M.; Borruel, N.; Casellas, F.; Campos, D.; Santiago, A.; Martinez, X.; Varela, E.; Sarrabayrouse, G.; Machiels, K.; et al. A microbial signature for Crohn’s disease. Gut 2017, 66, 813–822. [Google Scholar] [CrossRef]
  44. Waters, J.L.; Ley, R.E. The human gut bacteria Christensenellaceae are widespread, heritable, and associated with health. BMC Biol. 2019, 17, 83. [Google Scholar] [CrossRef] [PubMed]
  45. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T.; et al. Human genetics shape the gut microbiome. Cell 2014, 159, 789–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Sassone-Corsi, M.; Nuccio, S.P.; Liu, H.; Hernandez, D.; Vu, C.T.; Takahashi, A.A.; Edwards, R.A.; Raffatellu, M. Microcins mediate competition among Enterobacteriaceae in the inflamed gut. Nature 2016, 540, 280–283. [Google Scholar] [CrossRef] [PubMed]
  47. Salameh, A.; Klotz, S.A.; Zangeneh, T.T. Disseminated Infection Caused by Eggerthella lenta in a Previously Healthy Young Man: A Case Report. Case Rep. Infect. Dis. 2012, 2012, 517637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Abdulamir, A.S.; Hafidh, R.R.; Bakar, F.A. Molecular detection, quantification, and isolation of Streptococcus gallolyticus bacteria colonizing colorectal tumors: Inflammation-driven potential of carcinogenesis via IL-1, COX-2, and IL-8. Mol. Cancer 2010, 9, 249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Top, J.; Willems, R.; Bonten, M. Emergence of CC17 Enterococcus faecium: From commensal to hospital-adapted pathogen. FEMS Immunol. Med. Microbiol. 2008, 52, 297–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Trompette, A.; Gollwitzer, E.S.; Pattaroni, C.; Lopez-Mejia, I.C.; Riva, E.; Pernot, J.; Ubags, N.; Fajas, L.; Nicod, L.P.; Marsland, B.J. Dietary Fiber Confers Protection against Flu by Shaping Ly6c(-) Patrolling Monocyte Hematopoiesis and CD8(+) T Cell Metabolism. Immunity 2018, 48, 992–1005e1008. [Google Scholar] [CrossRef] [Green Version]
  51. Kim, M.; Qie, Y.; Park, J.; Kim, C.H. Gut Microbial Metabolites Fuel Host Antibody Responses. Cell Host Microbe 2016, 20, 202–214. [Google Scholar] [CrossRef] [Green Version]
  52. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef] [Green Version]
  53. Hosseini, E.; Grootaert, C.; Verstraete, W.; Van de Wiele, T. Propionate as a health-promoting microbial metabolite in the human gut. Nutr. Rev. 2011, 69, 245–258. [Google Scholar] [CrossRef] [PubMed]
  54. Magrone, T.; Jirillo, E. The interaction between gut microbiota and age-related changes in immune function and inflammation. Immun. Ageing 2013, 10, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. O’Toole, P.W.; Jeffery, I.B. Gut microbiota and aging. Science 2015, 350, 1214–1215. [Google Scholar] [CrossRef] [PubMed]
  56. He, F.; Ouwehand, A.C.; Isolauri, E.; Hosoda, M.; Benno, Y.; Salminen, S. Differences in composition and mucosal adhesion of bifidobacteria isolated from healthy adults and healthy seniors. Curr. Microbiol. 2001, 43, 351–354. [Google Scholar] [CrossRef] [PubMed]
  57. Biagi, E.; Franceschi, C.; Rampelli, S.; Severgnini, M.; Ostan, R.; Turroni, S.; Consolandi, C.; Quercia, S.; Scurti, M.; Monti, D.; et al. Gut Microbiota and Extreme Longevity. Curr. Biol. 2016, 26, 1480–1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Hansen, S.; Baptiste, K.E.; Fjeldborg, J.; Horohov, D.W. A review of the equine age-related changes in the immune system: Comparisons between human and equine aging, with focus on lung-specific immune-aging. Ageing Res. Rev. 2015, 20, 11–23. [Google Scholar] [CrossRef]
  59. Puhlmann, L.M.C.; Engert, V.; Apostolakou, F.; Papassotiriou, I.; Chrousos, G.P.; Vrticka, P.; Singer, T. Only vulnerable adults show change in chronic low-grade inflammation after contemplative mental training: Evidence from a randomized clinical trial. Sci. Rep. 2019, 9, 19323. [Google Scholar] [CrossRef]
  60. van den Munckhof, I.C.L.; Kurilshikov, A.; Ter Horst, R.; Riksen, N.P.; Joosten, L.A.B.; Zhernakova, A.; Fu, J.; Keating, S.T.; Netea, M.G.; de Graaf, J.; et al. Role of gut microbiota in chronic low-grade inflammation as potential driver for atherosclerotic cardiovascular disease: A systematic review of human studies. Obes. Rev. 2018, 19, 1719–1734. [Google Scholar] [CrossRef] [Green Version]
  61. Zhai, Y.; Shi, X.M.; Fitzgerald, S.M.; Qian, H.Z.; Kraus, V.B.; Sereny, M.; Hu, P.; Yin, Z.X.; Xu, J.W.; Zeng, Y. High sensitivity C-reactive protein associated with different health predictors in middle-aged and oldest old Chinese. Biomed. Environ. Sci. 2012, 25, 257–266. [Google Scholar] [CrossRef]
  62. Straub, R.H.; Schradin, C. Chronic inflammatory systemic diseases: An evolutionary trade-off between acutely beneficial but chronically harmful programs. Evol. Med. Public Health 2016, 2016, 37–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Singh, T.; Newman, A.B. Inflammatory markers in population studies of aging. Ageing Res. Rev. 2011, 10, 319–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhang, J.; Rane, G.; Dai, X.; Shanmugam, M.K.; Arfuso, F.; Samy, R.P.; Lai, M.K.; Kappei, D.; Kumar, A.P.; Sethi, G. Ageing and the telomere connection: An intimate relationship with inflammation. Ageing Res. Rev. 2016, 25, 55–69. [Google Scholar] [CrossRef] [PubMed]
  65. Helenius, M.; Hanninen, M.; Lehtinen, S.K.; Salminen, A. Aging-induced up-regulation of nuclear binding activities of oxidative stress responsive NF-kB transcription factor in mouse cardiac muscle. J. Mol. Cell Cardiol. 1996, 28, 487–498. [Google Scholar] [CrossRef]
  66. Kim, H.J.; Yu, B.P.; Chung, H.Y. Molecular exploration of age-related NF-kappaB/IKK downregulation by calorie restriction in rat kidney. Free Radic. Biol. Med. 2002, 32, 991–1005. [Google Scholar] [CrossRef]
  67. Green, D.R.; Galluzzi, L.; Kroemer, G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011, 333, 1109–1112. [Google Scholar] [CrossRef] [Green Version]
  68. Freund, A.; Orjalo, A.V.; Desprez, P.Y.; Campisi, J. Inflammatory networks during cellular senescence: Causes and consequences. Trends Mol. Med. 2010, 16, 238–246. [Google Scholar] [CrossRef] [Green Version]
  69. Michaud, M.; Balardy, L.; Moulis, G.; Gaudin, C.; Peyrot, C.; Vellas, B.; Cesari, M.; Nourhashemi, F. Proinflammatory cytokines, aging, and age-related diseases. J. Am. Med. Dir. Assoc. 2013, 14, 877–882. [Google Scholar] [CrossRef]
  70. Cesari, M.; Penninx, B.W.; Pahor, M.; Lauretani, F.; Corsi, A.M.; Rhys Williams, G.; Guralnik, J.M.; Ferrucci, L. Inflammatory markers and physical performance in older persons: The InCHIANTI study. J. Gerontol. A Biol. Sci. Med. Sci. 2004, 59, 242–248. [Google Scholar] [CrossRef] [Green Version]
  71. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [Green Version]
  72. Kohler, O.; Krogh, J.; Mors, O.; Benros, M.E. Inflammation in Depression and the Potential for Anti-Inflammatory Treatment. Curr. Neuropharmacol. 2016, 14, 732–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Nastasi, C.; Candela, M.; Bonefeld, C.M.; Geisler, C.; Hansen, M.; Krejsgaard, T.; Biagi, E.; Andersen, M.H.; Brigidi, P.; Odum, N.; et al. The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Sci. Rep. 2015, 5, 16148. [Google Scholar] [CrossRef] [PubMed]
  74. Dutta, S.; Sengupta, P. Men and mice: Relating their ages. Life Sci. 2016, 152, 244–248. [Google Scholar] [CrossRef] [PubMed]
  75. Brestoff, J.R.; Artis, D. Commensal bacteria at the interface of host metabolism and the immune system. Nat. Immunol 2013, 14, 676–684. [Google Scholar] [CrossRef] [Green Version]
  76. Matt, S.M.; Allen, J.M.; Lawson, M.A.; Mailing, L.J.; Woods, J.A.; Johnson, R.W. Butyrate and Dietary Soluble Fiber Improve Neuroinflammation Associated with Aging in Mice. Front. Immunol. 2018, 9, 1832. [Google Scholar] [CrossRef]
  77. McLoughlin, R.F.; Berthon, B.S.; Jensen, M.E.; Baines, K.J.; Wood, L.G. Short-chain fatty acids, prebiotics, synbiotics, and systemic inflammation: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2017, 106, 930–945. [Google Scholar] [CrossRef] [Green Version]
  78. Wong, J.M.; de Souza, R.; Kendall, C.W.; Emam, A.; Jenkins, D.J. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  79. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
  80. Hutchinson, A.N.; Bergh, C.; Kruger, K.; Susserova, M.; Allen, J.; Ameen, S.; Tingo, L. The Effect of Probiotics on Health Outcomes in the Elderly: A Systematic Review of Randomized, Placebo-Controlled Studies. Microorganisms 2021, 9, 1344. [Google Scholar] [CrossRef]
  81. Barcena, C.; Valdes-Mas, R.; Mayoral, P.; Garabaya, C.; Durand, S.; Rodriguez, F.; Fernandez-Garcia, M.T.; Salazar, N.; Nogacka, A.M.; Garatachea, N.; et al. Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat. Med. 2019, 25, 1234–1242. [Google Scholar] [CrossRef]
  82. Maneerat, S.; Lehtinen, M.J.; Childs, C.E.; Forssten, S.D.; Alhoniemi, E.; Tiphaine, M.; Yaqoob, P.; Ouwehand, A.C.; Rastall, R.A. Consumption of Bifidobacterium lactis Bi-07 by healthy elderly adults enhances phagocytic activity of monocytes and granulocytes. J. Nutr. Sci. 2013, 2, e44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Macfarlane, S.; Cleary, S.; Bahrami, B.; Reynolds, N.; Macfarlane, G.T. Synbiotic consumption changes the metabolism and composition of the gut microbiota in older people and modifies inflammatory processes: A randomised, double-blind, placebo-controlled crossover study. Aliment. Pharmacol. Ther. 2013, 38, 804–816. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, C.S.; Cha, L.; Sim, M.; Jung, S.; Chun, W.Y.; Baik, H.W.; Shin, D.M. Probiotic Supplementation Improves Cognitive Function and Mood with Changes in Gut Microbiota in Community-Dwelling Older Adults: A Randomized, Double-Blind, Placebo-Controlled, Multicenter Trial. J. Gerontol. A Biol. Sci. Med. Sci. 2021, 76, 32–40. [Google Scholar] [CrossRef] [PubMed]
  85. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [Green Version]
  86. Shin, N.R.; Lee, J.C.; Lee, H.Y.; Kim, M.S.; Whon, T.W.; Lee, M.S.; Bae, J.W. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014, 63, 727–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef] [Green Version]
  88. Zhang, X.; Shen, D.; Fang, Z.; Jie, Z.; Qiu, X.; Zhang, C.; Chen, Y.; Ji, L. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS ONE 2013, 8, e71108. [Google Scholar] [CrossRef]
  89. Derrien, M.; Belzer, C.; de Vos, W.M. Akkermansia muciniphila and its role in regulating host functions. Microb. Pathog. 2017, 106, 171–181. [Google Scholar] [CrossRef] [Green Version]
  90. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermudez-Humaran, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [Green Version]
  91. Martin, R.; Miquel, S.; Chain, F.; Natividad, J.M.; Jury, J.; Lu, J.; Sokol, H.; Theodorou, V.; Bercik, P.; Verdu, E.F.; et al. Faecalibacterium prausnitzii prevents physiological damages in a chronic low-grade inflammation murine model. BMC Microbiol. 2015, 15, 67. [Google Scholar] [CrossRef] [Green Version]
  92. Pedersen, H.K.; Gudmundsdottir, V.; Nielsen, H.B.; Hyotylainen, T.; Nielsen, T.; Jensen, B.A.; Forslund, K.; Hildebrand, F.; Prifti, E.; Falony, G.; et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 2016, 535, 376–381. [Google Scholar] [CrossRef]
  93. Tett, A.; Huang, K.D.; Asnicar, F.; Fehlner-Peach, H.; Pasolli, E.; Karcher, N.; Armanini, F.; Manghi, P.; Bonham, K.; Zolfo, M.; et al. The Prevotella copri Complex Comprises Four Distinct Clades Underrepresented in Westernized Populations. Cell Host Microbe 2019, 26, 666–679.e667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Hills, R.D., Jr.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C.; Theberge, C.R. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019, 11, 1613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kong, F.; Hua, Y.; Zeng, B.; Ning, R.; Li, Y.; Zhao, J. Gut microbiota signatures of longevity. Curr. Biol. 2016, 26, R832–R833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Makki, K.; Deehan, E.C.; Walter, J.; Backhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef] [Green Version]
  97. Menni, C.; Jackson, M.A.; Pallister, T.; Steves, C.J.; Spector, T.D.; Valdes, A.M. Gut microbiome diversity and high-fibre intake are related to lower long-term weight gain. Int J. Obes. (Lond.) 2017, 41, 1099–1105. [Google Scholar] [CrossRef] [Green Version]
  98. Oliver, A.; Chase, A.B.; Weihe, C.; Orchanian, S.B.; Riedel, S.F.; Hendrickson, C.L.; Lay, M.; Sewall, J.M.; Martiny, J.B.H.; Whiteson, K. High-Fiber, Whole-Food Dietary Intervention Alters the Human Gut Microbiome but Not Fecal Short-Chain Fatty Acids. mSystems 2021, 6, e00115-21. [Google Scholar] [CrossRef]
  99. Martinez, I.; Kim, J.; Duffy, P.R.; Schlegel, V.L.; Walter, J. Resistant starches types 2 and 4 have differential effects on the composition of the fecal microbiota in human subjects. PLoS ONE 2010, 5, e15046. [Google Scholar] [CrossRef] [Green Version]
  100. Venkataraman, A.; Sieber, J.R.; Schmidt, A.W.; Waldron, C.; Theis, K.R.; Schmidt, T.M. Variable responses of human microbiomes to dietary supplementation with resistant starch. Microbiome 2016, 4, 33. [Google Scholar] [CrossRef] [Green Version]
  101. Ze, X.; Duncan, S.H.; Louis, P.; Flint, H.J. Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon. ISME J. 2012, 6, 1535–1543. [Google Scholar] [CrossRef]
  102. Keenan, M.J.; Zhou, J.; Hegsted, M.; Pelkman, C.; Durham, H.A.; Coulon, D.B.; Martin, R.J. Role of resistant starch in improving gut health, adiposity, and insulin resistance. Adv. Nutr 2015, 6, 198–205. [Google Scholar] [CrossRef] [Green Version]
  103. Zhang, Y.; Chen, L.; Hu, M.; Kim, J.J.; Lin, R.; Xu, J.; Fan, L.; Qi, Y.; Wang, L.; Liu, W.; et al. Dietary type 2 resistant starch improves systemic inflammation and intestinal permeability by modulating microbiota and metabolites in aged mice on high-fat diet. Aging (Albany NY) 2020, 12, 9173–9187. [Google Scholar] [CrossRef] [PubMed]
  104. Alfa, M.J.; Strang, D.; Tappia, P.S.; Graham, M.; Van Domselaar, G.; Forbes, J.D.; Laminman, V.; Olson, N.; DeGagne, P.; Bray, D.; et al. A randomized trial to determine the impact of a digestion resistant starch composition on the gut microbiome in older and mid-age adults. Clin. Nutr. 2018, 37, 797–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Alfa, M.J.; Strang, D.; Tappia, P.S.; Olson, N.; DeGagne, P.; Bray, D.; Murray, B.L.; Hiebert, B. A Randomized Placebo Controlled Clinical Trial to Determine the Impact of Digestion Resistant Starch MSPrebiotic((R)) on Glucose, Insulin, and Insulin Resistance in Elderly and Mid-Age Adults. Front. Med. (Lausanne) 2017, 4, 260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Zhou, J.; Keenan, M.J.; Keller, J.; Fernandez-Kim, S.O.; Pistell, P.J.; Tulley, R.T.; Raggio, A.M.; Shen, L.; Zhang, H.; Martin, R.J.; et al. Tolerance, fermentation, and cytokine expression in healthy aged male C57BL/6J mice fed resistant starch. Mol. Nutr. Food Res. 2012, 56, 515–518. [Google Scholar] [CrossRef]
  107. Tachon, S.; Zhou, J.; Keenan, M.; Martin, R.; Marco, M.L. The intestinal microbiota in aged mice is modulated by dietary resistant starch and correlated with improvements in host responses. FEMS Microbiol. Ecol. 2013, 83, 299–309. [Google Scholar] [CrossRef] [Green Version]
  108. Peixoto, M.C.; Ribeiro, E.M.; Maria, A.P.J.; Loureiro, B.A.; di Santo, L.G.; Putarov, T.C.; Yoshitoshi, F.N.; Pereira, G.T.; Sa, L.R.M.; Carciofi, A.C. Effect of resistant starch on the intestinal health of old dogs: Fermentation products and histological features of the intestinal mucosa. J. Anim. Physiol. Anim. Nutr. (Berl.) 2018, 102, e111–e121. [Google Scholar] [CrossRef]
  109. Wang, Y.; Perfetti, R.; Greig, N.H.; Holloway, H.W.; DeOre, K.A.; Montrose-Rafizadeh, C.; Elahi, D.; Egan, J.M. Glucagon-like peptide-1 can reverse the age-related decline in glucose tolerance in rats. J. Clin. Invest. 1997, 99, 2883–2889. [Google Scholar] [CrossRef] [Green Version]
  110. Swanson, K.S.; Grieshop, C.M.; Flickinger, E.A.; Bauer, L.L.; Healy, H.P.; Dawson, K.A.; Merchen, N.R.; Fahey, G.C., Jr. Supplemental fructooligosaccharides and mannanoligosaccharides influence immune function, ileal and total tract nutrient digestibilities, microbial populations and concentrations of protein catabolites in the large bowel of dogs. J. Nutr. 2002, 132, 980–989. [Google Scholar] [CrossRef]
  111. Beloshapka, A.N.; Alexander, L.G.; Buff, P.R.; Swanson, K.S. The effects of feeding resistant starch on apparent total tract macronutrient digestibility, faecal characteristics and faecal fermentative end-products in healthy adult dogs. J. Nutr. Sci. 2014, 3, e38. [Google Scholar] [CrossRef] [Green Version]
  112. Windey, K.; De Preter, V.; Verbeke, K. Relevance of protein fermentation to gut health. Mol. Nutr. Food Res. 2012, 56, 184–196. [Google Scholar] [CrossRef] [PubMed]
  113. Azuma, N.; Kanaya, M.; Kanamoto, R.; Iwami, K. Feeding soybean resistant protein to rats raises fecal bile acid excretion but counteracts a deoxycholate-caused decrease in colonic aberrant crypt foci. J. Nutr. Sci. Vitaminol. (Tokyo) 1999, 45, 183–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Das, G.; Shin, H.S.; Campos, E.V.R.; Fraceto, L.F.; Del Pilar Rodriguez-Torres, M.; Mariano, K.C.F.; de Araujo, D.R.; Fernandez-Luqueno, F.; Grillo, R.; Patra, J.K. Sericin based nanoformulations: A comprehensive review on molecular mechanisms of interaction with organisms to biological applications. J. Nanobiotechnol. 2021, 19, 30. [Google Scholar] [CrossRef] [PubMed]
  115. Okazaki, Y.; Tomotake, H.; Tsujimoto, K.; Sasaki, M.; Kato, N. Consumption of a resistant protein, sericin, elevates fecal immunoglobulin A, mucins, and cecal organic acids in rats fed a high-fat diet. J. Nutr. 2011, 141, 1975–1981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Guarderas, F.; Leavell, Y.; Sengupta, T.; Zhukova, M.; Megraw, T.L. Assessment of Chicken-Egg Membrane as a Dressing for Wound Healing. Adv. Skin Wound Care 2016, 29, 131–134. [Google Scholar] [CrossRef]
  117. Kalman, D.S.; Hewlings, S. The effect of oral hydrolyzed eggshell membrane on the appearance of hair, skin, and nails in healthy middle-aged adults: A randomized double-blind placebo-controlled clinical trial. J. Cosmet. Dermatol. 2020, 19, 1463–1472. [Google Scholar] [CrossRef]
  118. Vuong, T.T.; Ronning, S.B.; Suso, H.P.; Schmidt, R.; Prydz, K.; Lundstrom, M.; Moen, A.; Pedersen, M.E. The extracellular matrix of eggshell displays anti-inflammatory activities through NF-kappaB in LPS-triggered human immune cells. J. Inflamm Res. 2017, 10, 83–96. [Google Scholar] [CrossRef] [Green Version]
  119. Wang, Y.; Fei, Y.; Liu, L.; Xiao, Y.; Pang, Y.; Kang, J.; Wang, Z. Polygonatum odoratum Polysaccharides Modulate Gut Microbiota and Mitigate Experimentally Induced Obesity in Rats. Int. J. Mol. Sci. 2018, 19, 3587. [Google Scholar] [CrossRef] [Green Version]
Figure 2. Illustration of the potential mechanisms by which probiotics, resistant starch, and resistant proteins contribute to the mitigation of aging-related chronic low-grade inflammation by producing SCFAs, improving phagocytic activity (B. lactis, B. longum), or directly reducing the production of pro-inflammatory cytokines.
Figure 2. Illustration of the potential mechanisms by which probiotics, resistant starch, and resistant proteins contribute to the mitigation of aging-related chronic low-grade inflammation by producing SCFAs, improving phagocytic activity (B. lactis, B. longum), or directly reducing the production of pro-inflammatory cytokines.
Nutrients 14 00747 g002
Table 1. The importance of several bacteria and their fermentation products to the host.
Table 1. The importance of several bacteria and their fermentation products to the host.
ComponentRole or Function
Microbial Fermentation Metabolites
SCFAs (overall)Stimulating immune system [50]
Providing energy for and supporting the growth of beneficial bacteria [8]
Increasing both intestinal immunoglobulin A (IgA) and systemic immunoglobulin G (IgG) responses to prevent pathogens growth [51]
AcetateSupporting the growth of probiotics [52]
ButyratePrimary energy source for human colonocytes [21]
Preventing dysbiosis in epithelial cells [22]
Reducing pro-inflammatory cytokines and chemokines [23]
PropionateLowering lipogenesis, serum cholesterol levels, and carcinogenesis in other tissues [53]
Probiotics
Christensenellaceae familyAssociated with health conditions, particularly body mass index (BMI) and inflammatory bowel disease (IBD) [41,42,43,44,45]
Akkermansia
muciniphila
Anti-inflammatory activities [28]
Protecting intestinal epithelial integrity [34]
Supporting colonization of SCFA-producing bacteria [35]
Bifidobacterium spp.Producing lactate and acetate that can reduce the population of pathogenic bacteria [36]
Suppressing serum levels of pro-inflammatory cytokines [36]
Clostridium
cluster XIVa
Producing butyrate [38]
Lachnospiraceae
bacterium
Producing butyrate [38]
Faecalibacterium prausnitziiProducing butyrate [27]
Strong anti-inflammatory properties [27]
Coprococcus spp.Producing butyrate [39]
Roseburia spp.Producing butyrate [39]
Strong anti-inflammatory properties [40]
SCFAs, short chain fatty acids.
Table 2. Summary of study investigating the effects of resistant starch intake in either aged human or animal subjects.
Table 2. Summary of study investigating the effects of resistant starch intake in either aged human or animal subjects.
ReferenceSubjectType of
Resistant Starch
MethodsEffects
[104]Elderly (≥70 years old) compared to middle-aged adults (30–50 years old)MSPrebiotic® **Prospective, placebo-controlled, randomized, double-blinded study, 30 g of RS intervention for 3 months.↑ bifidobacteria
↓ Proteobacteria dysbiosis
[105]Elderly
(≥70 years old)
MSPrebiotic® **Prospective, blinded, placebo-controlled study,
30 g of RS consumption for 12 weeks.
↓ blood glucose and insulin resistance
(↓ type-2 diabetes risk in elderly)
[103]Healthy 18-month-old mice *HAMRS2High-fat diet, supplemented with 20% of RS2 for 16 weeks.↓ systemic endotoxemia expression, pro-inflammatory cytokines (LPS, IL-2, IL-4)
↑ gut barrier function
↓ pathogen related to obesity, inflammation, and aging
[106]Healthy 18 to 20-month-old mice *HAMRS20, 18, and 36% of HAMRS2 diet for 10 weeks.↑ gut microbial fermentation
↑ cecal proglucagon (recuperate aging-related decline in glucose tolerance)
[107]18 to 20-month-old mice *HAMRS20, 18, and 36% of HAMRS2 intervention for 10 weeks.↑ Bacteroidetes, Bifidobacterium spp., Akkermansia spp., and Allobaculum spp.
↑ proglucagon level
[108]11.5-year-old dogsRSFeed added with 1.46% RS was given for 51 days.↑ fecal butyrate and total SCFA concentrations
* Mice 18 to 20 months of age are equivalent to 60–66-year-old humans [74]. ** MSPrebiotic® is a commercial resistant starch product containing 70% RS2 from Solanum tuberosum extract [105]. HAMRS2: high amylose maize resistant starch type 2; RS: resistant starch; LPS: lipopolysaccharide; SCFA: short-chain fatty acid. ↑: increase, ↓: reduce or decrease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Warman, D.J.; Jia, H.; Kato, H. The Potential Roles of Probiotics, Resistant Starch, and Resistant Proteins in Ameliorating Inflammation during Aging (Inflammaging). Nutrients 2022, 14, 747. https://doi.org/10.3390/nu14040747

AMA Style

Warman DJ, Jia H, Kato H. The Potential Roles of Probiotics, Resistant Starch, and Resistant Proteins in Ameliorating Inflammation during Aging (Inflammaging). Nutrients. 2022; 14(4):747. https://doi.org/10.3390/nu14040747

Chicago/Turabian Style

Warman, Dwina Juliana, Huijuan Jia, and Hisanori Kato. 2022. "The Potential Roles of Probiotics, Resistant Starch, and Resistant Proteins in Ameliorating Inflammation during Aging (Inflammaging)" Nutrients 14, no. 4: 747. https://doi.org/10.3390/nu14040747

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

Article Metrics

Back to TopTop