Next Article in Journal
Promoting Healthier Drinking Habits: Using Sound to Encourage the Choice for Non-Alcoholic Beers in E-Commerce
Previous Article in Journal
Improving the Sensory, Nutritional and Physicochemical Quality of Fresh Meat
Previous Article in Special Issue
In Vitro Starch Digestibility, Rheological, and Physicochemical Properties of Water Caltrop Starch Modified with Cycled Heat-Moisture Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Resistant Starches and Non-Communicable Disease: A Focus on Mediterranean Diet

Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy
*
Author to whom correspondence should be addressed.
Foods 2021, 10(9), 2062; https://doi.org/10.3390/foods10092062
Submission received: 9 July 2021 / Revised: 25 August 2021 / Accepted: 27 August 2021 / Published: 1 September 2021
(This article belongs to the Special Issue Starch: Properties, Processing, and Functionality in Food Systems)

Abstract

:
Resistant starch (RS) is the starch fraction that eludes digestion in the small intestine. RS is classified into five subtypes (RS1–RS5), some of which occur naturally in plant-derived foods, whereas the others may be produced by several processing conditions. The different RS subtypes are widely found in processed foods, but their physiological effects depend on their structural characteristics. In the present study, foods, nutrition and biochemistry are summarized in order to assess the type and content of RS in foods belonging to the Mediterranean Diet (MeD). Then, the benefits of RS consumption on health are discussed, focusing on their capability to enhance glycemic control. RS enters the large bowel intestine, where it is fermented by the microbiome leading to the synthesis of short-chain fatty acids as major end products, which in turn have systemic health effects besides the in situ one. It is hoped that this review will help to understand the pros of RS consumption as an ingredient of MeD food. Consequently, new future research directions could be explored for developing advanced dietary strategies to prevent non-communicable diseases, including colon cancer.

Graphical Abstract

1. Introduction

Non-communicable diseases (NCDs) are chronic disorders, which tend to be of long duration. NCDs are due to a combination of genetic, physiological, environmental and behavioral factors, including unhealthy diets, physical inactivity, tobacco smoke or alcohol use. NCDs can be preventable by nutrition and the adoption of an active lifestyle [1,2]. Nutritional habits and physical activity represent a winning combination to counteract the rising burden of NCDs. Both unhealthy diets and a lack of physical activity are determinants to develop obesity, which in turn is linked to: (i) rise of blood pressure, (ii) increase of blood glucose and (iii) elevation in blood lipids, i.e., all conditions leading to the development of metabolic diseases. Therefore, obesity is strongly associated with chronic inflammation, which in turn leads to metabolic and cardiovascular diseases and even cancer [3]. On this basis, understanding the mechanisms leading to obesity is essential to develop preventive strategies and new treatments [4,5]. Food intake plays a critical role in the development of metabolic diseases. Recently, an investigation on European children from six different nations has highlighted that the consumption of fiber-rich foods does not meet present recommended daily intake guidelines [6]. The authors have found out a weak but significant correlation between an increase in BMI and a decrease in consumption frequency of wholegrain cereals and wholemeal products (bread, cereals, biscuits, pasta, rice). Furthermore, adopting the consumption of high-fiber foods in early childhood can delay the initiation of impaired condition [6,7]. These results are mostly driven by Italy (lower cereals consumption frequency). Angel Keys was the first to describe the Mediterranean Diet (MeD) as such and to present it to the popular thoughts as it is today. He was a biologist and physiologist who focused his studies on the dietary habits of people living in the South of Italy. Following the joint candidacy of Italy, Spain, Greece and Morocco, succeeded by Cyprus, Croatia and Portugal, the MeD was recognized by UNESCO, WHO and FAO. Such a diet varies by country and region; therefore, it has a range of definitions. It usually includes a low intake of meat and eggs, a moderate intake of dairy products and milk, and a large consumption of vegetables, fruits rich in phenols, fish, olive oil and seeds, rich in unsaturated fats [8,9]. Furthermore, legumes, whole cereals, potato, bread and rice are indicated in the Mediterranean food pie chart (Figure 1).
Taken together, those latter foods positively influence blood glycemia since their soluble fiber contents lower their glycemic index (GI), and gut bacteria ferment them. In this view, recent MeD intervention was proven to alter the gut microbiome in older people, reducing their frailty and improving health status [10]. Among the soluble fibers able to act as prebiotics, resistant starch (RS) present in foods from the Mediterranean area assumes certain importance in the MeD. This review will discuss the MeD with a particular focus on RS as a strategy to prevent NCDs, including cancer.

2. Type of Resistant Starch and Its Content in Mediterranean Food

According to the cause of digesting resistance, Englyst et al. classified resistant starch (RS) into four categories [11]. Later on, a new type of RS was found, which became the fifth kind of RS [12], leading to the new classification indicated in Table 1.
The formation of RS in food processing seems to be related to the amylose content, water availability and starch–lipid interaction, as recently reviewed by Ibrahim O. Mohamed [13]. RS3 refers to starch molecules that have undergone retrogradation or the realignment of starch molecules after gelatinization and chilling. Because retrograded starch molecules have a higher gelatinization temperature, they cannot fit into amylase’s substrate-binding site [13]. Although RS is not digestible, it interacts positively with the human body; in fact, it modulates the absorption of carbohydrates, determining a lowering of GI, and indirectly, a decrease in blood lipid levels. The main foods of the MeD containing RS are: (i) cereals (pasta, rice and bread), (ii) legumes (peas, beans, lentils and chickpeas) and (iii) potatoes. In particular, raw, unground grains contain more than 10% of RS, but they are not usually consumed. Different amounts of starch are present in pasta depending on its nature; for example, fresh pasta contains 49.1 ± 6.8 g/kg (dry weight ± SD) of starch, in which not more than 6% is represented by RS [14,15]. RS was also studied in other diets, such as that of India and in commonly consumed foods in the United States [16]. Legumes naturally contain RS from 4% to 5%, depending on their origin, conservation mode and type of cooking [17]. Legumes boiled and then kept for 24 h in a refrigerator can increase their levels of RS up to 6% of the total weight, permitting some fraction of amylose to recrystallize [18]. A worldwide used tubers in the MED is potatoes (white and yellow). Their processing conditions can affect health benefits. For example, cooling potatoes after cooking can substantially increase their amount of RS, tripling its content [19]. RS food content in Mediterranean foods is present in Table 2 and Table 3. RS can be determined through RS Assay Kit by Megazyme, Bray, Ireland, and recognized as the Official analysis method by the codex alimentarius methods (AOAC Method 2002.02, AACC Method 32-40.01, CODEX Type II Method) [20,21]. Although, RS obtained by food processing needs chemical-physical structure characterization [22,23,24,25,26,27,28,29,30]. To this, several techniques to determine their crystallinity, structural order, chain-length distribution and conformation, helicity, as well as double-helical structures are used [31,32,33,34,35,36,37,38]. These include: (i) scanning electron microscopy (SEM), (ii) differential scanning calorimetry (DSC), (iii) X-ray diffraction (XRD) analysis, (iv) solid-state 13C nuclear magnetic resonance (13C-NMR), (v) permethylation-GC-MS and (vi) Fourier transform infrared spectroscopy (FT-IR).

3. Resistant Starch in Human Nutritional Intervention Studies: GI and Impact on Inflammation and Gut Microbiome

In recent years, the gut microbiota has been widely investigated, and its imbalance, due to antibiotics use [41], has been related to many disorders, including inflammation and oxidative stress, which underlie several chronic diseases, such as obesity, type 2 diabetes and chronic kidney disease [42]. Several reports have evidenced that a prebiotic supplemented diet can healthily regulate the gut microbiota, thus relieving disorders due to its imbalance. Prebiotics comprises non-digestible dietary soluble fiber, which can be used by the gut microbiota for fermentation. In this context, RS can act as a substrate for microbial fermentation in the large intestine [42] by supplying an energy source and fermentative products, such as short-chain fatty acids (SCFAs) in their anionic forms, in such a way RS is able to modulate microbial growth and could influence colonic health (Figure 2). On this basis, RS is believed to be a prebiotic and to influence the GI of foods favoring a lower intestinal absorption of glucose, even in the presence of polyphenols that in turn inhibits enterocytic starch digestion enzymes [43]
It has been reported that the extended lack of dietary fiber can lead to irreversible changes in the gut microbiota composition and elicit gut dysbiosis, even impairing gut inflammatory mediators, thereby inducing several bowel diseases [44].

3.1. Resistant Starch and Enhancement of Glycemic Control

When compared to food containing only readily digestible starch, the rate of digestion of RS-containing foods in the small intestine is substantially slower. As a result, consumption of such food leads to a sustained and lower level of glucose release [45]. This effect is reflected by the GI, a ranking system that organizes different food products based on the glycemic response to food consumption [45]. Researchers discovered a decrease in starch digestibility in treated food compared to untreated food after producing retrogradation in test meals [46]. They also observed a slower rise in blood glucose levels in human subjects upon consuming treated food when compared to those consuming untreated food [46,47]. Several studies have reported that potatoes generally have medium to high GI, which has often negatively impacted their consumption, but such studies have overlooked the many nutritional and health benefits of potatoes [48,49]. Interestingly, the GI varies depending on the potato variety, origin, maturity and processing methods, which can alter the starch digestibility of consumed foods. Hence, the concept of glycemic load (GL) was developed to simultaneously describe the quality (i.e., GI) and quantity of carbohydrates in a meal or diet [50,51]. Potato starch consists of 70–80% amylopectin, which is a highly branched, high molecular weight biopolymer. Amylose represents approximately 20–30% of starch, and it is a relatively long, linear, α-glucan with only a few branches. The relative proportion of amylose and amylopectin is important, given that amylose acts as a restraint to swelling and, upon cooling, it forms retrograded starch more readily [52,53,54]. Other than total starch, structure and moisture content [55], as stated above, other factors can also impact the glycemic response, including the growing conditions, maturity of the potato variety and cooking methods [56]. Potato varieties, maturity level, starch structure, methods of food processing and composition of the meal affect the GI of potatoes. Boiling, baking, microwave or oven cooking, extrusion and frying result in diverse degrees of gelatinization and starch crystallinity in potatoes. Cooling or storage after processing potatoes significantly reduces their GI because of the retrogradation of starch molecules. Although beneficial effects of resistant starch consumption have been observed, the current results of studies on the underlying molecular mechanisms in both animals and humans are not yet conclusive. Additional research efforts are necessary in order to reach a better understanding of the effects of habitual RS consumption on glycemic control.

3.2. Resistant Starch, Gut Microbiome and Inflammation

The mammalian gastrointestinal microbiota makes important contributions to the health of the host, including immune system development, nutrient metabolism and absorption, drug metabolism as well as protection against infection [57]. An altered microbiota (dysbiosis) has been associated with human diseases, such as diabetes, obesity, inflammatory bowel diseases, fecal occult blood and colorectal cancer [57,58,59]. By the way, diet is also considered a key modulator of the composition and function of the gut microbiota. Over the past few decades, the dietary intake of RS has been investigated. RS is a type of fermentable fiber considered a prebiotic since it can reach the large intestine, in which gut bacteria ferment it. RS fermentation leads to SCFAs production and pH reduction in the proximal large intestine. It has long been known that diet influences the microbial communities of the gastrointestinal tract. Although studies to understand how different classes of RS can affect microbiota are limited, it is clear that high-fiber diets greatly modulate the composition of mammalian microbiota [60]. Among the different classes of resistant starches, RS1–RS5 (Table 1), type 3 (RS3) is endowed with the strongest prebiotic properties [61]. Recent studies have explored the comparative physiological effects of diverse types of RS, investigating RS-induced changes in the microbiome that might be substantial in health and disease. Bacteria in the large intestine can be exposed to as much as 20 g of RS per day in humans [62]. RS may exert protective effects through broader mechanisms associated with fermentation. Acetate, propionate and butyrate are the most abundant anionic form of SCFAs generated from colonic microbial metabolism. SCFAs play a key role in the regulation of the inflammation process; contextually, they induce protective effects by stimulating or lessening inflammatory cytokines production, as well as by inhibiting or facilitating immune cells recruitment [63]. Furthermore, the fermentation of RS in the colon results in the production of gases (methane, hydrogen and carbon dioxide), small amounts of organic acid in its anionic form (lactate, succinate and formate), branched SCFAs either in anionic form such as valerate and butyrate. This latter is mainly obtained from the fermentation of RS2 than that of other RSs; notably, it has displayed interesting anti-inflammatory properties [62,64,65]. In particular, RS2 can promote a greater growth of bacteria belonging to the families of Bifidobacteriaceae and Lactobacillaceae, which are known to reduce inflammation [64]. Additionally, RS2 supplementation led to an increase in the gut level of Faecalibacterium; this prompted the authors to hypothesize a bacterial involvement in the anti-inflammatory effect exerted by the prebiotic fiber [65]. A future research direction to better assess gut/fecal microbial composition and serum concentration of anionic form SCFAs before and after RS2 intake could be represented by rodent models [66,67].
In any case, thanks to the healthy properties of RS, several nutraceuticals containing it are commercially available. A recent, randomized, placebo-controlled clinical trial evaluated the effects of resistant potato starch (RPS; MSPrebiotic®) containing 28% of fiber, administrated for 12 weeks (30 g/d), in healthy adult subjects focusing on the microbiome, reporting a reduction in the abundance of Proteobacteria. RPS consumers had a gut microbiome containing higher Parasutterella (phylum Proteobacteria) levels than subjects consuming placebo, andsuch increases were correlated with reductions in the blood levels of low-density lipoproteins. On this basis, it is feasible that the effect of Parasutterella on the host’s metabolism might depend upon several partly unknown factors, including prebiotic consumption, and they could play a critical role in cholesterol homeostasis [68,69].

3.3. RS, Blood Lipid Profile and Cytokines Levels

The beneficial effect of a long-term (12 months) dietary intervention with increased fiber intake, including RS on humans, was assessed [69,70]. In the study, two dietary groups were investigated, RS group subjects ingested higher amounts of food rich in RS (especially cereals and legumes) in order to consume about 15 g/day of RS, while subjects in the fiber group received general advice to ingest vegetables rich in fiber, without specific advice on the intake of RS-rich foods. At the end of the study, in the RS group, anthropometric parameters, such as body weight, body mass index (BMI) and waist circumference, were slightly more decreased, and a negative correlation was found between RS intake and adiponectin level, along with a negative correlation between RS intake and blood level of resistin, a possible pro-inflammatory mediator of insulin resistance. Furthermore, leptin and apelin levels were significantly decreased only in the RS group [71,72]. As already discussed above, RS exists in different types, having different chemical structures. In a double-blind controlled, crossover intervention study, a diet enriched with RS4, a chemically modified starch, was found to lower blood cholesterol and improve body composition measured by dual-energy, X-ray absorptiometry (DXA) [73]. The RS4 enriched diet was used for 26 weeks in the management of metabolic syndrome (MetS) using RS4 flour at 30% (v/v). Regular flour was used as a control (CF). Similar results on human lipid profiles were highlighted with RS3 (40 g/d for 21 days) in overweight and obese females [73] and RS2 (>25 g/d for 12 months) in overweight and obese subjects [73,74]. High–RS potato starch (with low protein) was used as a source of RS in patients with early type 2 diabetic nephropathy (DN) for 12 weeks [75]. In the study, the control group subjects consumed each day protein restriction diet with an everyday staple, whereas subjects of the intervention group ingested 50 g of high-RS, low-protein flour instead of an everyday staple of equal quality at lunch and dinner every day. A significant reduction in the levels of hemoglobin A1c (HbA1c) and lipid profile was found in the intervention group, along with a marked decrease of serum uric acid and urinary β2-microglobulin levels [74], thus delaying the progression of early type 2 DN. Several short-term intervention studies employing whole grain (WG) foods, including WG barley or rye derivatives rich in intact kernels, dietary fiber and RS, exhibited anti-obesogenic and anti-diabetic effects in healthy subjects [39,40,75]. More recently, a short-term, crossover, randomized study investigated the effect of rye-based bread preparation on healthy middle-aged subjects by employing white wheat flour bread (WWB) as a reference in order to evaluate possible effects on cardiometabolic risk markers, cognitive functions and mood [40]. Rye-based bread was prepared using a WG rye kernel/flour mixture (1:1 ratio) supplemented with RS2 (RB + RS2). Such a dietary treatment significantly increased insulin sensitivity, fasting concentrations of plasma butyrate, acetate and total SCFAs anionic form, which in turn have beneficial systemic effects [40]. Moreover, the dietary treatment significantly increased the fasting levels of plasma gut hormones, such as the peptide YY (PYY) and the glucagon-like peptide (GLP)-2. Furthermore, fasting levels of the inflammatory marker interleukin (IL)-1β were significantly decreased. Remarkably, the fasting concentrations of butyrate and acetate and the breath hydrogen excretion were significantly increased when preceded by the RB + RS2 intervention, suggesting that increased gut microbial fermentation of dietary fiber could mediate the observed good effects. On the other hand, blood levels of lipids, and of other inflammatory markers, including C-reactive protein, brain-derived neurotrophic factor, IL-6 and IL-18, did not significantly differ between the RB + RS2 and WWB group, as well as no significant differences in appetite sensations were observed (satiety, desire to eat or hunger), and in cognitive performance. However, insulin sensitivity was found to be positively correlated with working memory test performance. Furthermore, subjects receiving the RB + RS2 intervention felt glad more, pleased, happy, active, awake and peppy when compared to those of the WWB group. Overall, studies in humans have highlighted that the dietary intake of RS seems to not directly and significantly affect body weight and composition. In the same way, the effects of RS on reducing energy intake, increasing satiety and improving lipid profiles are controversial. On the other hand, RS supplementation can healthily affect glucose homeostasis by decreasing fasting or postprandial glucose levels and improving insulin sensitivity. Additionally, RS exerts beneficial effects on the gut microbiota, and it positively modulates gut hormones, such as GLP-1 and PYY. Nevertheless, such statements are still not definitive since further research is required, especially regarding the need for a larger sample size and longer intervention times [76]. Additionally, Chang et al. showed that the anti-inflammatory properties, as well as the inhibition of pro-inflammatory responses by intestinal macrophages, performed by butyrate, were dependent on its histone deacetylase (HDAC) inhibitory activity. Such inhibition, in turn, increased histone H3 acetylation within genetic loci required for regulatory T cells (also called Tregs) induction [77]. Moreover, it was evidenced that butyrate increased the release of the anti-inflammatory cytokine IL-10 and decreased the production of pro-inflammatory cytokines, such as TNF-α, IL-6 and nitric oxide (NO) [78]. Recent studies highlighted that a diet supplemented with buckwheat RS induced a significant decrease in TNF-α and interleukin-6 (IL-6) levels in high-fat diet (HFD)-fed mice; therefore, buckwheat RS supplementation could improve inflammatory response in plasma [79]. Instead, controversial results emerged from human nutritional clinical studies regarding the RS2 effect on inflammatory mediators. Indeed, a study performed on hemodialysis patients, who had taken RS2 for 4 weeks, evidenced a decreased IL-6 concentration [80]. In agreement, in patients with systemic inflammation associated with chronic kidney disease, a supplemented diet containing high-amylose maize RS2 elicited a significant decrease in serum levels of IL-6 and TNFα with respect to the placebo [81]. On the contrary, in women with type 2 diabetes, RS2 dietary intake did not induce any significant change in IL-6 concentration compared to the placebo [82]. RS2 significantly decreased TNF-α level and ameliorated both glycemic and lipid profiles in women with T2DM after 8 weeks of treatment [83]. Conversely, after 12 weeks of supplementation with RS, prediabetes adults displayed reduced concentrations of plasmatic TNF-α but no significant improvement in insulin resistance [84]. These controversial results were recently reported in a systematic review with meta-analysis. The authors concluded that RS2 could not reduce inflammatory mediators, but they also recognized the need for more randomized controlled trials with longer intervention in terms of duration, use of higher dose and studies in different countries [85].

4. Potential Mechanisms of Resistant Starch in Prevention of Colon Cancer

As discussed above, fermentation products, especially butyrate, benefit colonic health by regulating colonic enterocyte proliferation, differentiation and apoptosis. This regulation leads to a less proliferative and more differentiated phenotype and results in fewer pre-neoplastic lesions induced by colon carcinogenesis. RS diets have a significant impact on the composition of the colonic bacterial community. A recent study showed that the three major phyla predominating in the mammalian gut, Firmicutes, Bacteroidetes and Actinobacteria, showed significant changes in their relative abundance in animals fed RS [86]. Thus, the changes in the gut microbiota evoked by RS could favor microorganisms producing butyrate, which in turn serve as an energy source for the colonocytes, and they have been proposed to protect against colon cancer [87]. Colon cancer is one of the most common gastrointestinal tumors, second in women and third in men worldwide. The incidence of colorectal cancer rises with age, and it is well recognized that environmental factors, such as adiposity, poor physical activity and junk-food diet, play a major role in carcinogenesis [86,87,88]. A systematic review of the epidemiological literature has reported convincing evidence that higher intakes of red meat, processed meat, increases in body fat and alcoholic drinks increase the risk of colorectal cancer. In contrast, increased physical activity, foods containing dietary fiber and garlic, as well as calcium, may reduce the risk [88]. Although studies of RS and human colonic health are still needed, RS may protect the human colon against possibly damaging aspects of dietary red meat and have important biological effects, including colon cancer reduction [89,90]. Several potential mechanisms have been proposed by which dietary RSs are believed to alter the development or progression of colon cancer. The most common hypotheses have focused on the increase of fecal wet weight, fecal pH, defecation frequency and modification of the microbiota, with increased production of important metabolites, SCFAs, such as acetate, propionate and butyrate, which appear to have important biological effects. As already mentioned, RSs cannot be digested by amylases in the small intestine and arrive into the colon to be fermented by microbiota. One protective mechanism for RS is indeed the production of fermentation products, in particular, butyrate has raised the most interest since it may be protective against colorectal cancer [91,92]. Butyrate is a histone deacetylase inhibitor and is considered an important factor in the maintenance of the healthy function in colorectal mucosa. Normal colonocytes gain 70–80% of their energy from butyrate; therefore epithelial proliferation is supported [93]. On the other hand, on intestinal tumor cell lines, butyrate showed anti-tumorigenic effects, including reduction of cell proliferation and induction of differentiation and apoptosis [94,95]. RS led to modifications in the morphologically normal colonic mucosa, similar to those observed in cell cultures after treatment with butyrate. Recently, a rodent study highlighted that RS supplementation to a high red meat diet increased colonic butyrate levels, thus decreasing inflammation, attenuating red meat-induced DNA damage, and reducing adenocarcinoma formation in response to carcinogens [96]. In addition, animal studies identified that high butyrate levels induced by RS supplementation led to an increased expression of genes involved in DNA repair, which is expected to result in fewer mutations and reduced carcinogenesis in rapidly dividing populations of the colonic mucosa [97]. Hence, enhanced removal of damaged cells and an increased repair efficiency owing to lower proliferation could be involved in tumor prevention by RS. Butyrate’s capability to regulate gene expression results from epigenetic mechanisms, including its role as a histone deacetylase inhibitor able to modulate DNA methylation and the expression of microRNA (miRNA). Butyrate has also been shown to influence gene expression in the colon by modulating RNA splicing [98] and exerting an influence on cell proliferation and differentiation through the modulation of several signal transduction pathways, among which the most important seems to be the modulation of the Wnt signaling pathway [99]. In some colon cancer cell lines, constitutive expression of the canonical Wnt pathway, an initiating event in most colorectal cancers, is upregulated by butyrate treatment, resulting in a strong apoptotic response [100]. Gene expression is also regulated by epigenetic mechanisms, such as histone modifications or DNA methylation and the expression of miRNA. The randomized, controlled, crossover trial by Humphreys et al. [101] was the first reported human study investigating the effects of RS on miRNA expression. The study highlighted how a diet rich in red meat could increase by approximately 30% miRNA expression from the miR-17-92 cluster, an oncogenic cluster overexpressed in colorectal cancer, while expression of five miRNAs from this cluster, namely miR-17, miR-19a, miR-19b, miR-20a and miR-92a were significantly reduced in subjects fed with RS (Figure 3).
In addition to inhibiting tumor cell proliferation, butyrate might reduce colorectal cancer risk by enhancing the apoptotic response to DNA damage induced by genotoxic colorectal carcinogen [102,103]. Moreover, butyrate has been shown to affect the in vivo production and composition of mucus, which plays important protective and immunological roles, and provides the environment for colonic microbiota [104,105]. All these data may indicate a preventive role for butyrate-producing RS in the development of colorectal neoplasia. As mentioned above, RS may also exert its protective effect through broader mechanisms associated with fiber, such as by reshaping gut microbiota versus a more beneficial state, reducing bile acid metabolism, increasing fecal bulk, decreasing transit time and reducing pH levels in the colonic lumen [106,107]. Hence, altogether these effects might contribute to the prevention of and protection from colorectal cancer.

5. Food Claims Regarding Resistant Starch

Resistant starch is naturally found in processed and whole starchy foods belonging to the Mediterranean diet, including bread, cereals, grains, pasta, potatoes, rice and legumes. Raw foods have the highest RS content; cooked and then chilled potatoes and cereals have a higher RS content than the boiled or heated ones, as the refrigeration process favors retrogradation of starch granules to make them less digestible. Shelf life also increases RS content in some foods, such as durum wheat pasta. RS food claims were based on clinical trials [12,16,39,40,68,69,70,71,72,73,74,75].
The European Food Safety Authority (EFSA) authorized a health claim concerning the benefits of RS consumption on postprandial blood glucose concentrations [108,109]. In addition, the US Food and Drug Administration (FDA) authorized a qualified health claim for high-amylose maize RS and reduction of type 2 diabetes risk [108,109,110].

6. Conclusions and Future Direction

The Mediterranean diet lifestyle pattern is important to delay and fight non-communicable diseases, the assessment of resistance starches content in foods from the Mediterranean diet could improve Mediterranean diet adherence. As future research directions, the development of advanced dietary strategies highlighting the percentage of resistant starches in the nutritional label of food products would be useful. In this view, the regulatory institutions and/or governments should include the resistant starches ingredient, labelling it in the food manufacturers. This could be a strategy to make consumers safe to choose foods able to prevent non-communicable diseases, including colon cancer.

Author Contributions

Conceptualization, E.C., P.T. and A.R.C.; data curation, A.F., A.R.C., G.L, P.T. and R.C.; writing—original draft preparation, A.F., A.R.C., G.L., P.T. and R.C.; writing—review and editing, E.C., R.C. and P.T.; supervision, V.D. 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.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Budreviciute, A.; Damiati, S.; Sabir, D.K.; Onder, K.; Schuller-Goetzburg, P.; Plakys, G.; Katileviciute, A.; Khoja, S.; Kodzius, R. Management and Prevention Strategies for Non-communicable Diseases (NCDs) and Their Risk Factors. Front. Public Health 2020, 8. [Google Scholar] [CrossRef] [PubMed]
  2. Caprara, G. Mediterranean-Type Dietary Pattern and Physical Activity: The Winning Combination to Counteract the Rising Burden of Non-Communicable Diseases (NCDs). Nutrients 2021, 13, 429. [Google Scholar] [CrossRef] [PubMed]
  3. Bhattacharya, S.; Juyal, R.; Hossain, M.M.; Singh, A. Non-communicable diseases viewed as “collateral damage” of our decisions: Fixing accountabilities and finding sloutions in primary care settings. J. Fam. Med. Prim. Care 2020, 9, 2176–2179. [Google Scholar] [CrossRef] [PubMed]
  4. Gortmaker, S.L.; Swinburn, B.A.; Levy, D.; Carter, R.; Mabry, P.L.; Finegood, D.; Huang, T.; Moodie, M. Changing the future of obesity: Science, policy, and action. Lancet 2011, 378, 838–847. [Google Scholar] [CrossRef] [Green Version]
  5. González-Muniesa, P.; Mártinez-González, M.A.; Hu, F.; Depres, J.P.; Matsuzawa, Y.; Loos, R.J.F.; Moreno, L.A.; Bray, G.A.; Martinez, J.A. Obesity. Nat. Rev. Dis. Primers 2017, 3, 17034. [Google Scholar] [CrossRef]
  6. Hörmann-Wallner, M.; Krause, R.; Alfaro, B.; Jilani, H.; Laureati, M.; Almi, V.L.; Sandell, M.; Sandvik, P.; Zeinstra, G.Z.; Methven, L. Intake of Fibre-Associated Foods and Texture Preferences in Relation to Weight Status Among 9-12 Years Old Children in 6 European Countries. Front Nutr. 2021, 8, 633807. [Google Scholar] [CrossRef]
  7. Cannataro, R.; Fazio, A.; La Torre, C.; Caroleo, M.C.; Cione, E. Polyphenols in the Mediterranean Diet: From Dietary Sources to microRNA Modulation. Antioxidants 2021, 10, 328. [Google Scholar] [CrossRef] [PubMed]
  8. Trichopoulou, A.; Martínez-González, M.A.; Tong, T.Y.; Forouhi, N.G.; Khandelwal, S.; Prabhakaran, D.; Mozaffarian, D.; de Longeril, M. Definitions and potential health benefits of the Mediterranean diet: Views from experts around the world. BMC Med. 2014, 12, 112. [Google Scholar] [CrossRef] [Green Version]
  9. Ghosh, T.S.; Rampelli, S.; Jeffery, I.B.; Santoro, A.; Neto, M.; Capri, M.; Giampieri, E.; Jennings, A.; Candela, M.; Turroni, S.; et al. Mediterranean diet intervention alters the gut microbiome in older people reducing frailty and improving health status: The NU-AGE 1-year dietary intervention across five European countries. Gut 2020, 69, 1218–1228. [Google Scholar] [CrossRef] [Green Version]
  10. Englyst, H.N.; Kingman, S.M.; Cummings, J.H. Classification and measurement of nutritionally important starch fractions. Eur. J. Clin. Nutr. 1992, 46, 33–50. [Google Scholar]
  11. Woo, K.; Seib, P. Cross-linked resistant starch: Preparation and properties 1. Cereal Chem. 2002, 79, 819–825. [Google Scholar] [CrossRef]
  12. Hasjim, J.; Lee, S.-O.; Hendrich, S.; Setiawan, S.; Setiawan, A.; Ai, Y.; Jane, J.L. Characterization of a novel resistant-starch and its effects on postprandial plasma-glucose and insulin responses. Cereal Chem. 2010, 87, 257–262. [Google Scholar] [CrossRef]
  13. Ibrahim, O.M. Effects of processing and additives on starch physicochemical and digestibility properties. Carbohydr. Polym. Technol. Appl. 2021, 2, 100039. [Google Scholar]
  14. Brighenti, F.; Casiraghi, M.C.; Baggio, C. Resistant starch in the Italian diet. Br. J. Nutr. 1998, 80, 333–341. [Google Scholar]
  15. Platel, K.; Shurpalekar, K.S. Resistant starch content of Indian foods. Plant. Foods Hum. Nutr. 1994, 45, 91–95. [Google Scholar] [CrossRef]
  16. Patterson, M.A.; Maiya, M.; Stewart, M.L. Resistant Starch Content in Foods Commonly Consumed in the United States: A Narrative Review. J. Acad. Nutr. Diet 2020, 120, 230–244. [Google Scholar] [CrossRef]
  17. Fabbri, A.D.T.; Schacht, R.W.; Crosby, G.A. Evaluation of Resistant Starch Content of Cooked Black Beans, Pinto Beans and Chickpeas. NFS J. 2016, 3, 8–12. [Google Scholar] [CrossRef] [Green Version]
  18. Narwojsz, A.; Borowska, E.J.; Polak-Śliwińska, M.; Danowska-Oziewicz, M. Effect of Different Methods of Thermal Treatment on Starch and Bioactive Compounds of Potato. Plant Foods Hum. Nutr. 2020, 75, 298–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Jaiturong, P.; Laosirisathian, N.; Sirithunyalug, B.; Eitssayeam, S.; Sirilun, S.; Chaiyana, W.; Sirithunyalug, J. Physicochemical and prebiotic properties of resistant starch from Musa sapientum Linn., ABB group, cv. Kluai Namwa Luang. Heliyon 2020, 6, e05789. [Google Scholar] [CrossRef]
  20. Okumus, B.N.; Tacer-Caba, Z.; Kahraman, K.; Nilufer-Erdil, D. Resistant starch type V formation in brown lentil (Lens culinaris Medikus) starch with different lipids/fatty acids. Food Chem. 2018, 240, 550–558. [Google Scholar] [CrossRef] [PubMed]
  21. Ma, Z.; Boye, J.I. Research advances on structural characterization of resistant starch and its structure-physiological function relationship: A review. Crit. Rev. Food Sci. Nutr. 2018, 58, 1059–1083. [Google Scholar] [CrossRef]
  22. Polesi, L.F.; Sarmento, S.B.S. Structural and physicochemical characterization of resistant starch prepared using hydrolysis and heat treatments of chickpea starch. Starch-Starke 2011, 63, 226–235. [Google Scholar] [CrossRef]
  23. Dingting, Z.; Zhen, M.; Xinzhong, H. Isolated Pea Resistant Starch Substrates with Different Structural Features Modulate the Production of Short-Chain Fatty Acids and Metabolism of Microbiota in Anaerobic Fermentation In Vitro. J. Agric. Food Chem. 2021, 69, 5392–5404. [Google Scholar]
  24. Zhang, Y.; Zeng, H.; Wang, Y.; Zeng, S.; Zheng, B. Structural characteristics and crystalline properties of lotus seed resistant starch and its prebiotic effects. Food Chem. 2014, 155, 311–318. [Google Scholar] [CrossRef]
  25. Guo, J.; Liu, L.; Lian, X.; Li, L.; Wu, H. The properties of different cultivars of Jinhai sweet potato starches in China. Int. J. Biol. Macromol. 2014, 67, 1–6. [Google Scholar] [CrossRef]
  26. Barreto, F.F.V.; Bello-Pérez, L.A. Chemical, Structural, Technological Properties and Applications of Andean Tuber Starches: A Review. Food Rev. 2021, 1–16. [Google Scholar] [CrossRef]
  27. Buléon, A.; Colonna, P.; Planchot, V.; Ball, S. Starch granules: Structure and biosynthesis. Int. J. Biol. Macromol. 1998, 23, 85–112. [Google Scholar] [CrossRef] [Green Version]
  28. Wong, K.T.; Poh, G.Y.Y.; Kelvin, K.; Goh, T.; Wee, M.S.W.; Henry, C.J. Comparison of physicochemical properties of jackfruit seed starch with potato and rice starches. Int. J. Food Prop. 2021, 24, 364–379. [Google Scholar] [CrossRef]
  29. Eerlingen, R.C.; Crombez, M.; Delcour, J.A. Enzyme-Resistant starch. I. Quantitative and qualitative influence of incubation-time and temperature of autoclaved starch on resistant starch formation. Cereal Chem. 1993, 70, 339–344. [Google Scholar]
  30. Shamai, K.; Shimoni, E.; Bianco-Peled, H. Small-angle X-ray scattering of resistant starch type III. Biomacromolecules 2004, 5, 219–223. [Google Scholar] [CrossRef]
  31. Mutungi, C.; Onyango, C.; Doert, T.; Paasch, S.; Thiele, S.; Machill, S.; Jaros, D.; Rohm, H. Long- and short-range structural changes of recrystallised cassava starch subjected to in vitro digestion. Food Hydrocoll. 2011, 25, 477–485. [Google Scholar] [CrossRef]
  32. Gidley, M.J.; Bociek, S.M. Molecular-Organization in Starches—A C-13 Cp Mas Nmr-Study. J. Am. Chem. Soc. 1985, 107, 7040–7044. [Google Scholar] [CrossRef]
  33. Thèrien, H. Study of hydration of cross-linked high amylose starch by solid-state 13C NMR spectroscopy. Carbohydr. Res. 2007, 342, 1525–1529. [Google Scholar] [CrossRef]
  34. Thompson, D.B. Strategies for the manufacture of resistant starch. Trends Food Sci. Technol. 2000, 11, 245–253. [Google Scholar] [CrossRef]
  35. Katoh, E.; Murata, K.; Fujita, N. 13C CP/MAS NMR Can Discriminate Genetic Backgrounds of Rice Starch. ACS Omega 2020, 5, 24592–24600. [Google Scholar] [CrossRef]
  36. Ruiz-Matute, A.I.; Hernandez-Hernandez, O.; Rodrıguez-Sanchez, S.; Sanz, M.L.; Martınez-Castro, I. Derivatization of carbohydrates for GC and GC-MS analyses. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2010, 879, 1226–1240. [Google Scholar] [CrossRef] [PubMed]
  37. Mahadevamma, S.; Prashanth, K.H.; Tharanathan, R. Resistant starch derived from processed legumes–Purification and structural characterization. Carbohydr. Polym. 2003, 54, 215–219. [Google Scholar] [CrossRef]
  38. Zhang, Z.; Wang, Y.; Zhang, Y.; Chen, K.; Chang, H.; Ma, C.; Jiang, S.; Huo, D.; Liu, W.; Jha, R.; et al. Synergistic Effects of the Jackfruit Seed Sourced Resistant Starch and Bifidobacterium pseudolongum subsp. globosum on Suppression of Hyperlipidemia in Mice. Foods. 2021, 10, 1431. [Google Scholar] [CrossRef]
  39. Johansson, E.V.; Nilsson, A.C.; Östman, E.M.; Björck, I.M.E. Effects of indigestible carbohydrates in barley on glucose metabolism, appetite and voluntary food intake over 16 h in healthy adults. Nutr. J. 2013, 12, 46. [Google Scholar] [CrossRef] [Green Version]
  40. Nilsson, A.C.; Johansson-Boll, E.V.; Bjorck, I.M. Increased gut hormones and insulin sensitivity index following a 3-d intervention with a barley kernelbased product: A randomised cross-over study in healthy middle-aged subjects. Br. J. Nutr. 2015, 114, 899–907. [Google Scholar] [CrossRef]
  41. Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as Major Disruptors of Gut Microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef]
  42. Dolan, K.T.; Chang, E.B. Diet, gut microbes, and the pathogenesis of inflammatory bowel diseases. Mol. Nutr. Food Res. 2017, 61, 1600129. [Google Scholar] [CrossRef] [PubMed]
  43. Ayua, E.O.; Nkhata, S.G.; Namaumbo, S.J.; Kamau, E.H.; Ngoma, T.N.; Aduol, K.O. Polyphenolic inhibition of enterocytic starch digestion enzymes and glucose transporters for managing type 2 diabetes may be reduced in food systems. Heliyon 2021, 12, e06245. [Google Scholar] [CrossRef]
  44. Fuentes-Zaragoza, E.; Riquelme-Navarrete, M.; Sánchez-Zapata, E.; Pérez-Álvarez, J. Resistant starch as functional ingredient: A review. Food Res. Int. 2010, 43, 931–942. [Google Scholar] [CrossRef]
  45. Burton, P.; Lightowler, H. The impact of freezing and toasting on the glycaemic response of white bread. Eur. J. Clin. Nutr. 2008, 62, 594–599. [Google Scholar] [CrossRef] [PubMed]
  46. Frei, M.; Siddhuraju, P.; Becker, K. Studies on the in vitro starch digestibility and the glycemic index of six different indigenous rice cultivars from the Philippines. Food Chem. 2003, 83, 395–402. [Google Scholar] [CrossRef]
  47. Foster-Powell, K.; Holt, S.H.; Brand-Miller, J.C. International Tables of Glycemic Index and Glycemic Load Values: 2002. Am. J. Clin. Nutr. 2002, 76, 5–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Atkinson, F.S.; Foster-Powell, K.; Brand-Miller, J.C. International Tables of Glycemic Index and Glycemic Load Values. Diabet. Care 2008, 3, 2281–2283. [Google Scholar] [CrossRef] [Green Version]
  49. Salmeron, J.; Monson, J.E.; Stampfer, M.S.; Colditz, G.A.; Wing, A.L.; Willett, W.C. Dietary Fiber, Glycemic Load and Risk of Non-Insulin Dependent Diabetes Mellitus on Woman. JAMA 1997, 277, 472–477. [Google Scholar] [CrossRef]
  50. Brennan, C.S. Dietary Fiber, Glycemic Response and Diabetes. Mol. Nutr. Food Res. 2005, 49, 560–570. [Google Scholar] [CrossRef]
  51. Hoover, R. Composition, Molecular Structure, and Physico-Chemical Properties of Tuber and Root Starches: A Review. Carbohydr. Polym. 2001, 45, 253–267. [Google Scholar] [CrossRef]
  52. Bertoft, E.; Blennow, A. Structure of Potato Starch. In Advances in Potato Chemistry and Technology, 2nd ed.; Singh, J., Kaur, L., Eds.; Elsevier: Copenaghen, Denmark, 2008; pp. 83–98. [Google Scholar]
  53. Pérez, S.; Bertoft, E. The Molecular Structures of Starch Components and Their Contribution to the Architecture of Starch Granules: A Comprehensive Review. Starch 2010, 62, 389–420. [Google Scholar] [CrossRef]
  54. Lynch, D.R.; Liu, Q.; Tarn, T.R.; Bizimungu, B.; Chen, Q.; Harris, P.; Chik, C.L.; Skjodt, N.M. Glycemic Index—A Review and Implications for the Potato Industry. Am. J. Potato Res. 2007, 84, 179–190. [Google Scholar] [CrossRef]
  55. Fernandes, G.; Velangi, A.; Wolever, T.M.S. Glycemic Index of Potatoes Commonly Consumed in North America. J. Acad. Nutr. Diet. 2005, 105, 557–562. [Google Scholar] [CrossRef] [PubMed]
  56. Dodevska, M.S.; Sobajic, S.S.; Djordjevic, P.B.; Dimitrijevic-Sreckovic, V.S.; Spasojevic-Kalimanovska, V.S.; Djordjevic, B.I. Effects of total fibre or resistant starch-rich diets within lifestyle intervention in obese prediabetic adults. Eur. J. Nutr. 2016, 55, 127–137. [Google Scholar] [CrossRef] [PubMed]
  57. Cafiero, C.; Re, A.; Pisconti, S.; Trombetti, M.; Perri, M.; Colosimo, M.; D’Amato, G.; Gallelli, L.; Cannataro, R.; Molinario, C.; et al. Dysbiosis in intestinal microbiome linked to fecal blood determined by direct hybridization. Biotech 2020, 10, 358. [Google Scholar] [CrossRef]
  58. Wong, S.H.; Yu, J. Gut microbiota in colorectal cancer: Mechanisms of action and clinical applications. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 690–704. [Google Scholar] [CrossRef]
  59. Wu, G.D.; Chen, Y.Y.; Hoffmann, C.; Bittinger, K.; Chen, Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [Green Version]
  60. Asp, N.G.; van Amelsvoort, J.M.M.; Hautvast, J.G.A.J. Nutritional implications of resistant starch. Nutr. Res. Rev. 1996, 9, 1–31. [Google Scholar] [CrossRef] [Green Version]
  61. Topping, D.L.; Clifton, P.M. Short-chain fatty acids and human colonic function: Roles of resistant starch and non-starch polysaccharides. Physiol Rev. 2001, 81, 1031–1064. [Google Scholar] [CrossRef]
  62. Corrêa-Oliveira, R.; Fachi, J.L.; Vieira, A.; Sato, F.T.; Vinolo, M.A. Regulation of immune cell function by short-chain fatty acids. Clin. Transl. Immunol. 2016, 5, e73. [Google Scholar] [CrossRef] [PubMed]
  63. Le Leu, R.K.; Hu, Y.; Brown, I.L.; Young, G.P. Effect of high amylose maize starches on colonic fermentation and apoptotic response to DNA-damage in the colon of rats. J. Nutr. Metab. 2009, 6, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Arpaia, N.; Rudensky, A.Y. Microbial metabolites control gut inflammatory responses. Proc. Natl. Acad. Sci. USA 2014, 111, 2058–2059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef] [Green Version]
  66. Birt, D.F.; Phillips, G.J. Diet, Genes, and Microbes: Complexities of Colon Cancer Prevention. Toxicol Pathol. 2014, 42, 182–188. [Google Scholar] [CrossRef] [Green Version]
  67. Sekirov, I.; Russell, S.L.; Antunes, C.M.; Finlay, B.B. Gut microbiota in health and disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [Green Version]
  68. Laffin, M.R.; Tayebi Khosroshahi, H.; Park, H.; Laffin, L.J.; Madsen, K.; Kafil, H.S.; Abedi, B.; Shiralizadeh, S.; Vaziri, N.D. Amylose resistant starch (HAM-RS2) supplementation increases the proportion of Faecalibacterium bacteria in end-stage renal disease patients: Microbial analysis from a randomized placebo-controlled trial. Hemodial. Int. 2019, 23, 343–347. [Google Scholar] [CrossRef] [Green Version]
  69. Bush, J.R.; Alfa, M.J. Increasing levels of Parasutterella in the gut microbiome correlate with improving low-density lipoprotein levels in healthy adults consuming resistant potato starch during a randomised trial. BMC Nutr. 2020, 11, 72. [Google Scholar]
  70. 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® on Glucose, Insulin, and Insulin Resistance in Elderly and Mid-Age Adults. Front. Med. 2018, 4, 260. [Google Scholar] [CrossRef] [Green Version]
  71. 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–e00121. [Google Scholar]
  72. Luo, R.; Li, X.; Jiang, R.; Gao, X.; Lu, Z.; Hua, W. Serum Concentrations of Resistin and Adiponectin and Their Relation-ship to Insulin Resistance in Subjects with Impaired Glucose Tolerance. J. Int. Med. Res. 2012, 40, 621–630. [Google Scholar] [CrossRef]
  73. Nichenametla, S.N.; Lee, A.; Weidauer, L.A.; Wey, H.E.; Beare, T.M.; Specker, B.L.; Dey, M. Resistant starch type 4-enriched diet lowered blood cholesterols and improved body composition in a double-blind controlled crossover intervention. Mol. Nutr. Food Res. 2014, 58, 1365–1369. [Google Scholar] [CrossRef]
  74. Park, O.J.; Ekang, N.; Chang, M.J.; Kim, W.K. Resistant starch supplementation influences blood lipid concentrations and glucose control in overweight subjects. J. Nutr. Sci. Vitaminol. 2004, 50, 93–99. [Google Scholar] [CrossRef] [Green Version]
  75. Meng, Y.; Bai, H.; Yu, Q.; Yan, J.; Zhao, L.; Wang, S.; Zhaoping, L.; Wang, Q.; Chen, L. High-resistant starch, low-protein flour intervention on patients with early type 2 diabetic nephropathy: A randomized trial. J. Ren Nutr. 2019, 29, 386–393. [Google Scholar] [CrossRef] [PubMed]
  76. Sandberg, J.C.; Björck, I.M.E.; Nilsson, A.C. Impact of rye-based evening meals on cognitive functions, mood and cardiometabolic risk factors: A randomized controlled study in healthy middle-aged subjects. Nutr. J. 2018, 17, 102. [Google Scholar] [CrossRef] [PubMed]
  77. Guo, J.; Tan, L.; Kong, L. Impact of dietary intake of resistant starch on obesity and associated metabolic profiles in human: A systematic review of the literature. Crit. Rev. Food 2021, 61, 889–905. [Google Scholar] [CrossRef]
  78. Vinolo, M.A.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Zhou, Y.; Zhao, S.; Jiang, Y.; Wei, Y.; Zhou, X. Regulatory function of buckwheat-resistant starch supplementation on lipid profile and gut microbiota in mice fed with a high-fFat diet. J. Food. Sci 2019, 84, 2674–2681. [Google Scholar] [CrossRef] [PubMed]
  80. Gargari, B.P.; Namazi, N.; Khalili, M.; Sarmadi, B.; Jafarabadi, M.A.; Dehghan, P. Is there any place for resistant starch, as alimentary prebiotic, for patients with type 2 diabetes? Complement. Ther. Med. 2015, 23, 810–815. [Google Scholar] [CrossRef]
  81. Dobranowski, P.A.; Stintzi, A. Resistant starch, microbiome, and precision modulation. Gut Microbes. 2021, 13, 1926842. [Google Scholar] [CrossRef]
  82. Esgalhado, M.; Kemp, J.A.; Azevedo, R.; Laffin, L.J.; Madsen, K.; Kafil, H.S.; Abedi, B.; Shiralizadeh, S.; Vaziri, N.D. Could resistant starch supplementation improve inflammatory and oxidative stress biomarkers and uremic toxins levels in hemodialysis patients? A pilot randomized controlled trial. Food Funct. 2018, 9, 6508–6516. [Google Scholar] [CrossRef]
  83. Deehan, E.C.; Yang, C.; Perez-Muñoz, M.E.; Nguyen, N.K.; Cheng, C.C.; Triador, L.; Zhang, Z.; Bakal, J.A.; Walter, J. Precision Microbiome Modulation with Discrete Dietary Fiber Structures Directs Short-Chain Fatty Acid Production. Cell Host Microbe 2020, 27, 389–404.e6. [Google Scholar] [CrossRef]
  84. Peterson, C.M.; Beyl, R.A.; Marlatt, K.L.; Martin, C.K.; Aryana, K.J.; Marco, M.L.; Martin, R.J.; Keenan, M.J.; Ravussin, E. Effect of 12 wk of resistant starch supplementation on cardiometabolic risk factors in adults with prediabetes: A randomized controlled trial. Am. J. Clin. Nutr. 2018, 108, 492–501. [Google Scholar] [CrossRef]
  85. Haghighatdoost, F.; Gholami, A.; Hariri, M. Effect of resistant starch type 2 on inflammatory mediators: A systematic review and meta-analysis of randomized controlled trials. Complement. Ther. Med. 2021, 56, 102597. [Google Scholar] [CrossRef] [PubMed]
  86. Chapkin, R.S.B.; Navarro, S.L.; Hullar, M.A.J.; Lampem, J.W. Diet and Gut Microbes Act Coordinately to Enhance Programmed Cell Death and Reduce Colorectal Cancer Risk. Dig. Dis. Sci. 2020, 65, 840–851. [Google Scholar] [CrossRef] [Green Version]
  87. Keenan, M.J.; Zhou, J.; Hegsted, M.; Pelkman, C.; Durham, H.A.; Coulon, D.; Martin, R.J. Role of Resistant Starch in Improving Gut Health, Adiposity, and Insulin Resistance. Adv. Nutr. 2015, 6, 198–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Marmot, M.; Atinmo, T.; Byers, T.; Chen, J.; Hirohata, T.; Jackson, A.; James, W.; Kolonel, L.; Kumanyika, S.; Leitzmann, C.; et al. Food, Nutrition, Physical Activity, and the Prevention of Cancer: A Global Perspective; World Cancer Research Fund/American Institute for Cancer Research: Washington, DC, USA, 2007. [Google Scholar]
  89. Humphreys, K.J.; Conlon, M.A.; Young, G.P.; Topping, D.L.; Hu, Y.; Winter, J.M.; Bird, A.R.; Cobiac, L.; Kennedy, N.A.; Michael, M.Z.; et al. Dietary manipulation of oncogenic microRNA expression in human rectal mucosa: A randomized trial. Cancer Prev. Res. 2014, 7, 786–795. [Google Scholar] [CrossRef] [Green Version]
  90. Cassidy, A.; Bingham, S.A.; Cummings, J.H. Starch intake and colorectal cancer risk: An international comparison. Br. J. Cancer 1994, 69, 937–942. [Google Scholar] [CrossRef] [Green Version]
  91. Williams, E.A.; Coxhead, J.M.; Mathers, J.C. Anti-cancer effects of butyrate: Use of micro-array technology to investigate mechanisms. Proc. Nutr. Soc. 2003, 62, 107–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Mathers, J.C.; Smith, H.; Carter, S. Dose-response effects of raw potato starch on small-intestinal escape, large-bowel fermentation and gut transit time in the rat. Br. J. Nutr. 1997, 78, 1015–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Blottiere, H.M.; Buecher, B.; Galmiche, J.P.; Cherut, C. Molecular analysis of the effect of short-chain fatty acids on intestinal cell proliferation. Proc. Nutr. Soc. 2003, 62, 101–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Daly, K.; Shirazi-Beechey, S.P. Microarray analysis of butyrate regulated genes in colonic epithelial cells. DNA Cell Biol. 2006, 25, 49–62. [Google Scholar] [CrossRef] [PubMed]
  95. Fung, K.Y.; Cosgrove, L.; Lockett, T.; Head, R.; Topping, D.L. A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate. Br. J. Nutr. 2012, 108, 820–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Toden, S.; Bird, A.R.; Topping, D.L.; Conlon, M.A. Resistant starch prevents colonic DNA damage induced by high dietary cooked red meat or casein in rats. Cancer Biol. Ther. 2006, 5, 267–272. [Google Scholar] [CrossRef] [Green Version]
  97. Conlon, M.A.; Kerr, C.A.; McSweeney, C.S.; Dunne, R.A.; Shaw, J.M.; Kang, S.; Bird, A.R.; Morell, M.K.; Lockett, T.J.; Molloy, P.L.; et al. Resistant starches protect against colonic DNA damage and alter microbiota and gene expression in rats fed a Western diet. J. Nutr. 2012, 142, 832–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Bordonaro, M. Crosstalk between Wnt signaling and RNA processing in colorectal cancer. J. Cancer 2013, 4, 96–103. [Google Scholar] [CrossRef]
  99. Bordonaro, M.; Mariadason, J.M.; Aslam, F.; Heerdt, B.G.; Augenlicht, L.H. Butyrate-induced apoptotic cascade in colonic carcinoma cells: Modulation of the beta-catenin-Tcf pathway and concordance with effects of sulindac and trichostatin A but not curcumin. Cell Growth Differ. 1999, 10, 713–720. [Google Scholar]
  100. Lazarova, D.L.; Bordonaro, M.; Carbone, R.; Sartorelli, A.C. Linear relationship between Wnt activity levels and apoptosis in colorectal carcinoma cells exposed to butyrate. Int. J. Cancer 2004, 110, 523–531. [Google Scholar] [CrossRef]
  101. Del Cornò, M.; Donninelli, G.; Conti, L.; Gessani, S. Linking Diet to Colorectal Cancer: The Emerging Role of MicroRNA in the Communication between Plant and Animal Kingdoms. Front. Microbiol. 2017, 8, 597. [Google Scholar] [CrossRef] [Green Version]
  102. Bauer-Marinovic, M.; Florian, S.; Müller-Schmehl, K.; Glatt, H.; Jacobasch, G. Dietary resistant starch type 3 prevents tumor induction by 1,2-dimethylhydrazine and alters proliferation, apoptosis and dedifferentiation in rat colon. Carcinogenesis 2006, 27, 1849–1859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chen, J.; Zhao, K.N.; Vitetta, L. Effects of Intestinal Microbial-Elaborated Butyrate on Oncogenic Signaling Pathways. Nutrients. 2019, 11, 1026. [Google Scholar] [CrossRef] [Green Version]
  104. Sharma, R.; Schumacher, U.; Ronaasen, V.; Coates, M. Rat intestinal mucosal responses to a microbial flora and different diets. Gut 1995, 36, 209–214. [Google Scholar] [CrossRef] [Green Version]
  105. Deplancke, B.; Gaskins, H.R. Microbial modulation of innate defense: Goblet cells and the intestinal mucus layer. Am. J. Clin. Nutr. 2001, 73, 1131S–1141S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. McGarr, S.E.; Ridlon, J.M.; Hylemon, P.B. Diet, anaerobic bacterial metabolism, and colon cancer: A review of the literature. J. Clin. Gastroenterol. 2005, 39, 98–109. [Google Scholar] [PubMed]
  107. Jacobasch, G.; Dongowski, G.; Schmiedl, D.; Muller-Schmehl, K. Hydrothermal treatment of Novelose 330 results in high yield of resitant starch type 3 with beneficial prebiotic properties and decreased secondary bile acid formation in rats. Br. J. Nutr. 2006, 95, 1063–1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014, 7, 17–44. [Google Scholar] [CrossRef] [PubMed]
  109. Scientific opinion on the substantiation of health claims related to resistant starch and reduction of post-prandial glycaemic responses (ID 681), “digestive health benefits” (ID 682) and “favours a normal colon metabolism” (ID 783) pursuant to Article 13(1) of Regulation (EC) No 1924/2006. EFSA J. 2011, 9, 2024.
  110. Food and Drug Administration, Department of Health and Human Services. High-Amylose Starch and Diabetes. Docket Number FDA2015-Q-2352; Food and Drug Administration: College Park, MD, USA, 2015.
Figure 1. Mediterranean food pie chart.
Figure 1. Mediterranean food pie chart.
Foods 10 02062 g001
Figure 2. Resistant starch effect on the GI and microbiome SCFAs synthesis.
Figure 2. Resistant starch effect on the GI and microbiome SCFAs synthesis.
Foods 10 02062 g002
Figure 3. Poor resistant starch in diet as a promoting factor of colon cancer.
Figure 3. Poor resistant starch in diet as a promoting factor of colon cancer.
Foods 10 02062 g003
Table 1. Classification of RS and example of foods rich in RS.
Table 1. Classification of RS and example of foods rich in RS.
ClassificationDescriptionExample
RS1Physically inaccessible starchWhole grains
RS2Starch with B- or C-polymorphUncooked potato, high-amylose maize starch
RS3Retrograded starchCooked and cooled potato starch
RS4Chemically modified starchCross-linked starch in thickeners
RS5Amylose–lipid complexPalmitic acid-amylose complex
Table 2. Resistant starch in grains and legumes.
Table 2. Resistant starch in grains and legumes.
SampleTTTS (%)RDS (%)SDS (%)RS (%)HIpGIReference
Bean flourRAW43.14 ± 0.14 c82.11 ± 0.43 b10.25 ± 0.45 i7.64 ± 0.58 f87.71 ± 0.44 b87.86 ± 0.60 a
ANN43.06 ± 0.33 c77.93 ± 0.68 d11.11 ± 0.54 h10.96 ± 0.19 c78.11 ± 0.59 e82.59 ± 0.49 d[17,18,19]
HMT43.65 ± 0.31 c59.63 ± 0.65 g25.11 ± 0.96 c15.26 ± 0.43 a64.67 ± 0.27 j75.21 ± 0.11 e
Broad bean flourRAW43.43 ± 0.57 c80.26 ± 0.22 c11.14 ± 0.44 h8.60 ± 0.32 e84.66 ± 0.42 c86.18 ± 0.58 b
ANN42.26 ± 0.67 d73.75 ± 0.33 e16.61 ± 0.55 e9.64 ± 0.26 d76.42 ± 0.03 f81.66 ± 0.04 d[17,18,19]
HMT42.11 ± 0.76 d60.52 ± 0.68 g26.19 ± 0.76 c13.29 ± 0.43 b68.42 ± 0.80 g77.27 ± 0.08 e
Chickpea flourRAW45.32 ± 0.29 b85.26 ± 0.77 a9.05 ± 0.76 j5.69 ± 0.46 g90.15 ± 0.39 a89.20 ± 0.37 a
ANN44.95 ± 0.87 b77.10 ± 0.19 d14.26 ± 0.20 f8.64 ± 0.09 e76.16 ± 0.85 f81.52 ± 0.27 d[22]
HMT44.99 ± 0.55 b61.10 ± 0.37 g27.11 ± 0.89 b11.79 ± 0.78 b67.21 ± 0.16 h76.60 ± 0.49 e
Lentil flourRAW47.25 ± 0.11 a80.06 ± 0.34 c12.68 ± 0.65 g7.26 ± 0.61 f82.16 ± 0.49 d84.81 ± 0.83 c
ANN47.61 ± 0.98 a70.60 ± 0.44 f19.26 ± 0.39 d10.14 ± 0.65 c75.33 ± 0.55 f81.06 ± 0.65 d[20]
HMT47.65 ± 0.54 a59.60 ± 0.97 g30.14 ± 0.65 a10.26 ± 0.17 c66.36 ± 0.47 i76.14 ± 0.69 e
PeaH159.9 ± 1.78 c 3.7 ± 0.12 [23]
H2 3.2 ± 0.11
WheatH169.8 ± 1.10 d 1.9 ± 0.21 [14,15,16]
H2 1.8 ± 0.14
RiceH181.4 ± 1.10 f 1.4 ± 0.16 [28]
H2 1.2 ± 0.08
BarleyH165.6 ± 0.76 c 2.8 ± 0.23 [39,40]
H2 2.6 ± 0.09
PotatoH185.51 ± 1.64 g 1.8 ± 0.15 [18,28]
H2 1.7 ± 0.08
Different letters in the same column indicate significant differences (p < 0.05). TT: Thermal Treatment; TS: total starch, RDS: rapidly digestible starch, SDS: slowly digestible starch, RS: resistant starch, HI: hydrolysis index, pGI: predicted glycemic index; ANN: annealing, HMT: heat moisture treatment, H1: conventional boiling, H2: pressure cooking.
Table 3. Starch of whole legume seeds.
Table 3. Starch of whole legume seeds.
SampleTS (%)Thermal TreatmentRS (%)Reference
Bean38.34 + 0.7Boiled4.96 + 0.9 a (12.9) *
Cooked8.45 + 1.1 b (22.0) *[17,19]
Reheated8.24 + 0.3 b (21.5) *
Chickpea 41.36 + 1.0Boiled4.35 + 0.4 a (10.5) *
Cooked5.48 + 0.2 b (13.2) *[17,19]
Reheated5.58 + 0.1 b (13.5) *
Lentil46.72 + 2.1Boiled7.56 + 0.6 a (16.2) *
Cooked8.60 + 0.3 bc (18.4) *[20]
Reheated7.62 + 0.3 ab (16.3) *
Results are expressed as a percentage of dry matter (mean standard deviation, n = 4). Different letters in the same column indicate significant differences (p < 0.05) between the data for each legume. * Values in parentheses are RS expressed as a percentage of TS.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cione, E.; Fazio, A.; Curcio, R.; Tucci, P.; Lauria, G.; Cappello, A.R.R.; Dolce, V. Resistant Starches and Non-Communicable Disease: A Focus on Mediterranean Diet. Foods 2021, 10, 2062. https://doi.org/10.3390/foods10092062

AMA Style

Cione E, Fazio A, Curcio R, Tucci P, Lauria G, Cappello ARR, Dolce V. Resistant Starches and Non-Communicable Disease: A Focus on Mediterranean Diet. Foods. 2021; 10(9):2062. https://doi.org/10.3390/foods10092062

Chicago/Turabian Style

Cione, Erika, Alessia Fazio, Rosita Curcio, Paola Tucci, Graziantonio Lauria, Anna Rita Rita Cappello, and Vincenza Dolce. 2021. "Resistant Starches and Non-Communicable Disease: A Focus on Mediterranean Diet" Foods 10, no. 9: 2062. https://doi.org/10.3390/foods10092062

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