Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Food Funct ; 15(10): 5596-5612, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38722000

ABSTRACT

In the presented study, natural rice containing high resistant starch content was used as a raw material to produce rice resistant starch (RRS) through enzymatic hydrolysis with heat-stable α-amylase and glucoamylase. The chemical composition, structural characteristics and in vitro glycemic index (GI) of RRS were evaluated. The effects of RRS at different doses on the body weight, serum biochemical levels, pathological indexes, production of short-chain fatty acids (SCFAs) in the gut and the intestinal microbial composition in T2DM mice were investigated. The results of physiochemical characterization indicated that, relative to rice flour, RRS mainly comprising resistant starch had higher crystallinity (25.85%) and a more stable structure, which contributed to its lower digestibility and decreased GI in vitro. Compared with the model control group, 1 g per kg BW and 2 g per kg BW oral gavage dosages of RRS effectively enhanced the SCFA productivity in the T2DM mouse gut, as well as alleviating T2DM symptoms, involving an increase in body weight, reduction in fasting blood glucose, total cholesterol, triglyceride, low-density lipoprotein cholesterol, alanine transaminase and aspartate aminotransferase, and an increase in serum insulin and high-density lipoprotein cholesterol. Besides, 1 g per kg BW and 2 g per kg BW dosages of RRS mitigated T2DM-induced pancreas damage. Furthermore, up-regulation in the abundance of probiotics (Lactobacillus, Ruminococcus, etc.) and down-regulation in the number of harmful bacteria (Desulfovibrio, Prevotella, etc.) were observed in all RRS-treated groups. In summary, this work suggested that RRS prepared using heat-stable α-amylase and glucoamylase could be a potential functional component for amelioration of T2DM applied in the fields of food and pharmaceutics.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Glucan 1,4-alpha-Glucosidase , Oryza , Starch , alpha-Amylases , Animals , Oryza/chemistry , Mice , Gastrointestinal Microbiome/drug effects , Glucan 1,4-alpha-Glucosidase/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , alpha-Amylases/metabolism , Male , Starch/chemistry , Starch/metabolism , Starch/pharmacology , Blood Glucose/metabolism , Fatty Acids, Volatile/metabolism , Resistant Starch/pharmacology , Hot Temperature , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Humans
2.
Int J Biol Macromol ; 261(Pt 2): 129812, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38302033

ABSTRACT

The hypoglycemic effects of two recrystallized resistant starches, A-type (ARS) and B-type (BRS), were investigated in type 2 diabetic mice. Mice were treated with low-, medium-, or high-dose ARS, high-dose BRS, or high-dose ARS combined with BRS (ABRS). After 10 weeks of continuous intervention, the medium-dose ARS group showed a significant reduction in fasting blood glucose, area under the curve of glucose, triglyceride (P < 0.01), and low-density lipoprotein (P < 0.05) levels compared to the model group and an increase in high-density lipoprotein levels (P < 0.01). The peptide YY and glucagon-like peptide-1 levels in the high-dose ARS, BRS, and ABRS groups and the butyric acid yield in the medium-dose ARS and BRS groups were significantly increased (P < 0.01) compared to those in the model group. Medium- and high-dose ARS intervention efficiently increased the relative abundance of beneficial Bacteroidetes, Lactobacillus, Lachnospiraceae_NK4A136_group, and Faecalibaculum, and lowered the ratio of Firmicutes to Bacteroidetes. Overall, ARS exhibited greater advantages than BRS in lowering blood sugar levels.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Mice , Animals , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Resistant Starch/pharmacology , Streptozocin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diet, High-Fat/adverse effects , Diabetes Mellitus, Type 2/drug therapy
3.
Int J Biol Macromol ; 262(Pt 1): 129992, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38331070

ABSTRACT

Ionic strength condition is a crucial parameter for food processing, but it remains unclear how ionic strength alters the structure and digestibility of binary complexes containing starch and protein/protein hydrolysates. Here, the binary complex with varied ionic strength (0-0.40 M) was built by native corn starch (NS) and soy protein isolate (SPI)/hydrolysates (SPIH) through NaCl. The inclusion of SPI and SPIH allowed a compact network structure, especially the SPIH with reduced molecule size, which enriched the resistant starch (RS) of NS-SPIH. Particularly, the higher ionic strength caused the larger nonperiodic structures and induced loosener network structures, largely increasing the possibility of amylase for starch digestion and resulting in a decreased RS content from 19.07 % to 15.52 %. In other words, the SPIH hindered starch digestion while increasing ionic strength had the opposite effect, which should be considered in staple food production.


Subject(s)
Resistant Starch , Starch , Starch/chemistry , Resistant Starch/pharmacology , Protein Hydrolysates/pharmacology , Amylases , Osmolar Concentration , Digestion
4.
Int J Biol Macromol ; 253(Pt 3): 126930, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37717867

ABSTRACT

Carboxymethyl chitosan and resistant starch exhibit good performance in diabetes regulation. We prepared carboxymethyl chitosan - resistant starch complex. Test the properties of composite resistant starch by using X-ray diffraction, water contact angle, infrared spectroscopy, and scanning electron microscopy, interactions with intestinal microbiota and mouse experiments were also conducted. The results indicated that the composite resistant starch had a good effect on promoting the proliferation of probiotics on Bifidobacterium and a significant inhibitory effect on Escherichia coli than resistant starch (P < 0.05). After administration, the water intake and weight of diabetic mice were significantly reduced. The blood glucose of diabetic mice was also reduced, and oral glucose tolerance showed that the glucose degradation rates of composite resistant starch were significantly improved compared to model mice. Cholesterol, triglycerides, high-density lipoprotein and low-density lipoprotein were significantly lower than those in the diabetes group (P < 0.05). The diversity of the gut microbiota was also proven.


Subject(s)
Chitosan , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Animals , Mice , Resistant Starch/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Starch/chemistry , Chitosan/pharmacology , Chitosan/chemistry , Diabetes Mellitus, Experimental/drug therapy
5.
Eur J Nutr ; 62(5): 2269-2278, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37083722

ABSTRACT

PURPOSE: Resistant starch (RS) content has exhibited beneficial effects on glycemic control; however, few studies have investigated the effects of this substance on postprandial responses and appetite in subjects with type 2 diabetes (T2D). Here, we aimed to examine the effects of RS from two sources on glycemic response (GR), postprandial lipemia, and appetite in subjects with T2D. METHODS: In a randomized and crossover study, 17 subjects with T2D consumed native banana starch (NBS), high-amylose maize starch (HMS) or digestible maize starch (DMS) for 4 days. On day 5, a 6-h oral meal tolerance test (MTT) was performed to evaluate glycemic and insulinemic responses as well as postprandial lipemia. Besides, subjective appetite assessment was measured using a visual analogue scale. RESULTS: NBS induced a reduction on fasting glycemia, glycemia peak and insulinemic response during MTT. However, no modifications on postprandial lipemia were observed after RS treatments. Both NBS and HMS reduced hunger and increased satiety. CONCLUSION: NBS supplementation induced more beneficial effects on glycemic metabolism than HMS even when all interventions were matched for digestible starch content. RS intake did not modify postprandial lipemia, however, positively affected subjective appetite rates. TRIAL REGISTRATION: This trial was retrospectively registered at www.anzctr.org.au (ACTRN12621001382864) on October 11, 2021.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperlipidemias , Humans , Appetite , Resistant Starch/pharmacology , Cross-Over Studies , Blood Glucose/metabolism , Insulin , Starch/metabolism , Postprandial Period
6.
Curr Opin Clin Nutr Metab Care ; 26(4): 334-340, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37057658

ABSTRACT

PURPOSE OF REVIEW: Resistant starch has received much attention recently as a healthy carbohydrate component of the diet. Resistant starch is not digested in the small intestine and can thus affect the gut microbiota of the host because of its fermentability. This review summarizes the interactions along the resistant starch-gut microbiota-host axis to help understand the health effects of resistant starch. RECENT FINDINGS: Recent studies indicate that resistant starch can be a helpful dietary component for special disease states like diabetes, metabolic syndrome, chronic kidney disease, constipation, and colitis. Its health effects are associated with modulation of the gut microbiota, and with gut microbes converting resistant starch into active and bioavailable metabolites that promote intestinal health. SUMMARY: The results from human clinical trials and studies in animal models indicate that supplementation of the diet with resistant starch in different metabolic diseases help remodel gut microbiota, especially increasing short-chain fatty acid (SCFA)-producing bacteria, and produce bioactive metabolites like SCFA, bile acids, and amino acids responsible for a variety of health effects. The gut microbiota and microbial metabolites probably mediate the effects of resistant starch on intestinal health.


Subject(s)
Resistant Starch , Starch , Animals , Humans , Resistant Starch/pharmacology , Starch/chemistry , Starch/metabolism , Starch/pharmacology , Diet , Bacteria , Fatty Acids, Volatile/metabolism , Dietary Supplements
7.
Food Chem ; 419: 136008, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37004367

ABSTRACT

The effect of eight different l-amino acids (L-AA) on type-3 resistant starch particles (rSPs) derived from short chain glucan (SCG) was investigated. The L-AA were categorized based on their charge and polarity. The results reveal that positively charged L-AA, such as lysine and arginine, decreased the nucleation and growth rate of rSPs, while non-charged L-AA have negligible effects. Negatively charged L-AA, such as glutamic acid and aspartic acid, had a significant impact on the morphology and crystallinity of the rSPs, resulting in particle size of around 3 µm and crystallinity of around 35%. This implies that charged L-AA influence the arrangement of SCG double helices in the particles. Furthermore, the complexation of SCG with charged L-AA reduced the level of RS in rSPs, indicating that L-AA could be useful in modulating the physical properties and digestibility of rSPs.


Subject(s)
Amino Acids , Resistant Starch , Amino Acids/metabolism , Resistant Starch/pharmacology , Crystallization , Glutamic Acid/chemistry , Arginine/chemistry , Glucans/pharmacology , Starch/chemistry , Digestion
8.
Asia Pac J Clin Nutr ; 32(1): 48-56, 2023.
Article in English | MEDLINE | ID: mdl-36997485

ABSTRACT

BACKGROUND AND OBJECTIVES: Few studies exist on resistant starch in rice grains. The Okinawa Institute of Science and Technology Graduate University (OIST) has developed a new rice (OIST rice, OR) rich in resistant starch. This study aimed to clarify the effect of OR on postprandial glucose concentrations. METHODS AND STUDY DESIGN: This single-center, open, randomized, crossover comparative study included 17 patients with type 2 diabetes. All participants completed two meal tolerance tests using OR and white rice (WR). RESULTS: The median age of the participants was 70.0 [59.0-73.0] years, and the mean body mass index was 25.9±3.1 kg/m2. The difference in total area under the curve (AUC) of plasma glucose was -8223 (95% confidence interval [CI]: -10100 to -6346, p<0.001) mg·min/dL. The postprandial plasma glucose was significantly lower with OR than with WR. The difference in the AUC of insulin was -1139 (95% CI: -1839 to -438, p=0.004) µU·min/mL. The difference in the AUC of total gastric inhibitory peptide (GIP) and total glucagon-like peptide-1 (GLP-1) was -4886 (95% CI: -8456 to -1317, p=0.011) and -171 (95% CI: -1034 to 691, p=0.673) pmol·min/L, respectively. CONCLUSIONS: OR can be ingested as rice grains and significantly reduced postprandial plasma glucose compared to WR independent of insulin secretion in patients with type 2 diabetes. OR could have escaped absorption not only from the upper small intestine but also from the lower small intestine.


Subject(s)
Diabetes Mellitus, Type 2 , Oryza , Humans , Middle Aged , Aged , Incretins/pharmacology , Insulin , Blood Glucose , Resistant Starch/pharmacology , Postprandial Period , Cross-Over Studies
9.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-36638279

ABSTRACT

An experiment was conducted to determine how feeding calcium (Ca)-deficient diet would affect gastrointestinal pH and volatile fatty acids (VFAs), Ca digestibility, bone mineral density (BMD), and performance in nursery pigs; and if supplementation of nondigestible oligosaccharides would affect these same parameters. In total, 240 weaned pigs (BW = 7.1 kg) were placed into 80 pens with 3 pigs/pen. The eight dietary treatments consisted of: 1) positive control (PC, 0.83% total Ca), 2) negative control (NC, 0.50% total Ca), 3 and 4) NC + 5% or 7.5% soluble corn fiber (SCF), 5 and 6) NC + 5% or 7.5% resistant corn starch (rCS), 7 and 8) NC + 0.25% or 0.50% fat-protected butyrate (pBRT). Pigs were ad libitum fed the dietary treatments for 21 d to determine average daily gain (ADG), average daily feed intake (ADFI) and gain:feed ratio (GF) with a fecal sample collected from each pen to determine Ca digestibility using acid insoluble ash as the dietary marker, with 1 pig/pen euthanized on d 21 for collection of ileal and colon contents and the left humerus. Pigs fed the NC diet had a lower colonic pH compared with pigs fed the PC (P = 0.06) but no effect on total VFA was observed (P > 0.10). Pigs fed diets containing SCF and rCS had lower colonic pH and total VFA compared to pigs fed the NC diet (P ≤ 0.05). Pigs fed diets containing pBRT had greater colonic total VFA compared to pigs fed the NC diet (P ≤ 0.07), but no difference in colonic pH was observed (P > 0.10). Pigs fed the NC diet had a greater Ca digestibility compared to pigs fed the PC (P ≤ 0.01), with no treatment to the NC having any effect on Ca digestibility compared to pigs fed the NC (P > 0.10). There was no effect of dietary Ca level on BMD and no overall addition of feeding SCF, rCS, or pBRT on BMD compared to pigs fed the NC (P > 0.10). There was no impact on pig ADG, ADFI, or GF by reducing dietary Ca by 40% (i.e., pigs fed the NC) compared to pigs fed the PC (P > 0.10). Relative to pigs fed the NC, there was no overall effect of SCF, rCS, or pBRT on ADG, ADFI, or GF (P > 0.10). In conclusion, feeding young pigs a Ca-deficient diet reduced colonic pH, increased digestibility of Ca, but had no impact on bone mineralization or overall pig performance. Supplementation of nondigestible oligosaccharides pr protected butyrate had either no effect or an inconsistent effect on colonic pH, Ca, or PHOS digestibility, bone mineralization, or overall pig performance.


Calcium (Ca) is a major component of the skeleton in addition to being essential for growth and is imperative for bone mass development. Improvement in Ca absorption in Ca-deficient diets has been shown in human and rodent studies when nondigestible oligosaccharides have been consumed due to a modification of gastrointestinal conditions which increase mineral solubility. Because swine have been shown to be an excellent model for human nutrition research, an experiment was conducted to determine how a moderately Ca-deficient diet would affect gastrointestinal fermentation conditions, Ca and phosphorus (PHOS) digestibility, bone mineralization, and growth performance in nursery pigs; and if supplementation of nondigestible oligosaccharides would affect these same parameters. Results indicate that feeding young pigs a diet below recommended levels of Ca reduced colonic pH, increased apparent total-tract digestibility of Ca and PHOS, but had no impact on bone mineralization or overall pig performance. Supplementation of nondigestible oligosaccharides had inconsistent effects on colonic pH, and did not affect Ca or PHOS digestibility, bone mineralization, or overall pig performance.


Subject(s)
Calcium , Phosphorus , Swine , Animals , Calcium/pharmacology , Zea mays , Resistant Starch/pharmacology , Butyrates/pharmacology , Digestion , Calcium, Dietary/pharmacology , Diet/veterinary , Fatty Acids, Volatile/pharmacology , Animal Feed/analysis
10.
Neurobiol Dis ; 177: 105993, 2023 02.
Article in English | MEDLINE | ID: mdl-36627028

ABSTRACT

Clarifying the risk factors and mechanisms that contribute to the onset of cognitive impairment following estrogen depletion is essential for improving the quality of life of older females. In the current study, using behavioral tests, 16S rDNA sequencing, in vivo and in vitro electrophysiology, optogenetics and chemogenetics, we found that high-fat diet (HFD)-accelerated impairment of hippocampus-dependent memory, gut microbiota, and hippocampal theta rhythmogenesis in ovariectomized (OVX) mice and fecal microbiota transplantation rescued these phenomena. The identification of fasting-activated medial septal neurons showed that PV+ GABAergic neurons in the medial septal area (MSA) respond to gut sensory signals. Optogenetic activation of septohippocampal PV+ GABAergic fibers (but not cholinergic fibers) significantly rescued hippocampal theta rhythmogenesis and spatial memory in HFD-fed OVX mice. Resistant starch supplementation (RSHFD) rectified the gut Prevotellaceae and considerably alleviated reduced septal gut-responsive neurons, decreased hippocampal theta rhythm, and impaired hippocampus-dependent memory in HFD-fed OVX mice. Furthermore, chemogenetic inhibition of septal PV+ GABAergic neurons reversed the neuroprotective effects of resistant starch supplementation. These findings highlight the notable gut-sensory nature of medial septal PV+ GABAergic neurons. A HFD accelerates estrogen deficiency-induced cognitive impairment by disrupting the gut Prevotellaceae-septo-hippocampal pathway. This study contributes to a better understanding of the precise gut-brain control of cognition and cognitive impairment in postmenopausal females.


Subject(s)
Diet, High-Fat , Spatial Memory , Female , Mice , Animals , Diet, High-Fat/adverse effects , Quality of Life , Resistant Starch/metabolism , Resistant Starch/pharmacology , Hippocampus/metabolism , GABAergic Neurons/metabolism , Theta Rhythm/physiology
11.
J Dairy Sci ; 106(2): 1453-1463, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36526457

ABSTRACT

Intestinal hyperpermeability and subsequent immune activation alters nutrient partitioning and thus, decreases productivity. Developing experimental models of intestinal barrier dysfunction in heathy cows is a prerequisite in identifying nutritional strategies to mitigate it. Six cannulated Holstein cows (mean ± standard deviation, 37 ± 10 kg/d milk yield; 219 ± 97 d in milk; 691 ± 70 kg body weight) were used in a replicated 3 × 3 Latin square design experiment with 21-d periods (16-d wash-out and 5-d challenge) to evaluate either feed restriction or hindgut acidosis as potential models for inducing intestinal hyperpermeability. Cows were randomly assigned to treatment sequence within square and treatment sequences were balanced for carryover effects. Treatments during the challenge were (1) control (CTR; ad libitum feeding); (2) feed restriction (FR; total mixed ration fed at 50% of ad libitum feed intake); and (3) resistant starch (RS; 500 g of resistant starch infused in abomasum once a day as a pulse-dose 30 min before morning feeding). The RS (ActiStar RT 75330, Cargill Inc.) was tapioca starch that was expected to be resistant to enzymatic digestion in the small intestine and highly fermentable in the hindgut. Blood samples were collected 4 h after feeding on d 13 and 14 of the wash-out periods (baseline data used as covariate), and on d 1, 3, and 5 of the challenge periods. Fecal samples were collected 4 and 8 h after the morning feeding on d 14 of the wash-out periods and d 5 of the challenge periods. By design, FR decreased dry matter intake (48%) relative to CTR and RS, and this resulted in marked reductions in milk and 3.5% FCM yields over time, with the most pronounced decrease occurring on d 5 of the challenge (34 and 27%, respectively). Further, FR increased somatic cell count by 115% on d 5 of the challenge relative to CTR and RS. Overall, FR increased nonesterified fatty acids (159 vs. 79 mEq/L) and decreased BHB (8.5 vs. 11.2 mg/dL), but did not change circulating glucose relative to CTR. However, RS had no effect on production or metabolism metrics. Resistant starch decreased fecal pH 8 h after the morning feeding (6.26 vs. 6.81) relative to CTR and FR. Further, RS increased circulating lipopolysaccharide binding protein (4.26 vs. 2.74 µg/mL) compared with FR only on d 1 of the challenge. Resistant starch also increased Hp (1.52 vs. 0.48 µg/mL) compared with CTR, but only on d 5 of the challenge. However, neither RS or FR affected concentrations of serum amyloid A, IL1ß, or circulating endotoxin compared with CTR. The lack of consistent responses in inflammatory biomarkers suggests that FR and RS did not meaningfully affect intestinal barrier function. Thus, future research evaluating the effects of hindgut acidosis and FR using more intense insults and direct metrics of intestinal barrier function is warranted.


Subject(s)
Lactation , Resistant Starch , Female , Cattle , Animals , Resistant Starch/metabolism , Resistant Starch/pharmacology , Diet/veterinary , Abomasum/metabolism , Milk/metabolism , Animal Feed/analysis , Rumen/metabolism , Starch/metabolism
12.
Food Chem ; 404(Pt A): 134599, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36444019

ABSTRACT

Ordinary and hyperlipidemic rats were gavaged with lotus seed resistant starch (LRS), and the structure of the small intestinal flora and bile acids composition were determined for four groups of rats to construct a relationship network diagram between different bacterial genera, bile acids and blood lipid profiles, revealing a microbial mechanism for the lipid-lowering effect of LRS in hyperlipidemic rats. LRS inhibited the growth of Romboutsia, Bacillus, Blautia, norank_f__Muribaculaceae and norank_f__Eubacterium_coprostanoligenes_group in hyperlipidemic rats. Meanwhile LRS promoted the production of primary bile acids (CA, CDCA, ß-MCA) and secondary bile acids (LCA, UDCA), and reduced the contents of TCA, Dehydro-LCA, isoLCA, LCA-3-S and THDCA in hyperlipidemic rats. Furthermore, Blautia, norank_f__Muribaculaceae and norank_f__Eubacterium_coprostanoligenes_group were positively correlated with Dehydro-LCA, isoLCA, TCA, LCA-3-S, TCHO, TG and LDL-C. In summary, LRS improves blood lipid levels by regulating small intestinal flora and accelerating the breakdown of cholesterol into bile acids in the liver.


Subject(s)
Hyperlipidemias , Lotus , Resistant Starch , Seeds , Animals , Rats , Bacteroidetes , Bile Acids and Salts , Clostridiales , Gastrointestinal Microbiome/drug effects , Resistant Starch/pharmacology , Seeds/chemistry , Hyperlipidemias/microbiology , Hyperlipidemias/therapy
13.
Nutrients ; 14(2)2022 Jan 09.
Article in English | MEDLINE | ID: mdl-35057449

ABSTRACT

Individual glycemic responses following dietary intake result from complex physiological processes, and can be influenced by physical properties of foods, such as increased resistant starch (RS) from starch retrogradation. Predictive equations are needed to provide personalized dietary recommendations to reduce chronic disease development. Therefore, a precision nutrition model predicting the postprandial glucose response (PPGR) in overweight women following the consumption of potatoes was formulated. Thirty overweight women participated in this randomized crossover trial. Participants consumed 250 g of hot (9.2 g RS) or cold (13.7 g RS) potatoes on two separate occasions. Baseline characteristics included demographics, 10-day dietary records, body composition, and the relative abundance (RA) and α-diversity of gut microbiota. Elastic net regression using 5-fold cross-validation predicted PPGR after potato intake. Most participants (70%) had a favorable PPGR to the cold potato. The model explained 32.2% of the variance in PPGR with the equation: 547.65 × (0 [if cold, high-RS potato], ×1, if hot, low-RS potato]) + (BMI [kg/m2] × 40.66)-(insoluble fiber [g] × 49.35) + (Bacteroides [RA] × 8.69)-(Faecalibacterium [RA] × 73.49)-(Parabacteroides [RA] × 42.08) + (α-diversity × 110.87) + 292.52. This model improves the understanding of baseline characteristics that explain interpersonal variation in PPGR following potato intake and offers a tool to optimize dietary recommendations for a commonly consumed food.


Subject(s)
Blood Glucose/metabolism , Gastrointestinal Microbiome , Models, Biological , Obesity/blood , Postprandial Period , Resistant Starch/pharmacology , Solanum tuberosum/chemistry , Adult , Area Under Curve , Body Mass Index , Cross-Over Studies , Diet , Faecalibacterium , Female , Glycemic Index , Humans , Nutritional Status , Obesity/microbiology , Overweight/blood , Overweight/microbiology , Vegetables/chemistry , Young Adult
14.
Nutrients ; 13(11)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34836304

ABSTRACT

We previously observed beneficial effects of native banana starch (NBS) with a high resistant starch (RS) content on glycemic response in lean and obese participants. Here, we aimed to determine the effects of NBS and high-amylose maize starch (HMS) on glycemic control (GC) and glycemic variability (GV) in patients with type 2 diabetes (T2D) when treatments were matched for digestible starch content. In a randomized, crossover study, continuous glucose monitoring (CGM) was performed in 17 participants (aged 28-65 years, BMI ≥ 25 kg/m2, both genders) consuming HMS, NBS, or digestible maize starch (DMS) for 4 days. HMS and NBS induced an increase in 24 h mean blood glucose during days 2 to 4 (p < 0.05). CONGA, GRADE, and J-index values were higher in HMS compared with DMS only at day 4 (p < 0.05). Yet, NBS intake provoked a reduction in fasting glycemia changes from baseline compared with DMS (p = 0.0074). In conclusion, under the experimental conditions, RS from two sources did not improve GC or GV. Future longer studies are needed to determine whether these findings were affected by a different baseline microbiota or other environmental factors.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/complications , Glycemic Control/methods , Resistant Starch/pharmacology , Adult , Amylose , Blood Glucose Self-Monitoring , Cross-Over Studies , Female , Humans , Male , Middle Aged , Obesity , Starch/administration & dosage , Zea mays/chemistry
15.
Food Funct ; 12(23): 12098-12114, 2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34784410

ABSTRACT

Obesity is a most prevalent human health problem. Several studies showed that appropriate modulation of gut microbiota could help reshape the metabolic profile of obese individuals, thereby altering the development of obesity. A nutritional strategy for treating obesity includes prebiotics. Type 3 Resistant Starch from Canna edulis (Ce-RS3) is a dietary fiber that exerts potential effects on the intestinal microbial community; however, the metabolic landscape and anti-obesity mechanism remain unclear. In the present study, obese mice were treated with Ce-RS3, and 16S rRNA gene sequencing and metabolomics were used to measure changes in gut microbiota and fecal metabolic profiles, respectively. At the end of the treatment (13 weeks), we observed slow weight gain in the mice, and pathological damage and inflammation were substantially reduced. Ce-RS3 constructs a healthy gut microbiota structure and can enhance intestinal immunity and reduce metabolic inflammation. Ce-RS3 increased the diversity of gut microbiota with enrichment of Bifidobacterium and Roseburia. Ce-RS3 regulated the systemic metabolic dysbiosis in obese mice and adjusted 26 abnormal metabolites in amino acids and lipids metabolism, many of which are related to the microbiome. More importantly, we found that the anti-obesity effect of Ce-RS3 can be transferred by fecal transplantation. The beneficial effects of Ce-RS3 might derive from gut microbiota changes, which might improve obesity and metabolic inflammation by altering host-microbiota interactions with impacts on the metabolome. In conclusion, Ce-RS3 can be used as a prebiotic with potential value for the treatment of obesity.


Subject(s)
Gastrointestinal Microbiome/drug effects , Inflammation/metabolism , Obesity/metabolism , Resistant Starch/pharmacology , Zingiberales/chemistry , Animals , Dysbiosis/metabolism , Gastrointestinal Microbiome/genetics , Male , Mice , Mice, Inbred C57BL
16.
Food Funct ; 12(22): 11154-11164, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34694313

ABSTRACT

Liver glycogen α particles in diabetic patients are fragile relative to those in healthy individuals, and restoring these fragile glycogen particles to a normal state shows potential to contribute to the remission of diabetes. Resistant starch (RS) is beneficial for diabetes management through its interactions with the gut microbiota. However, its effects on glycogen fragility are not fully understood. This review aims to summarize the recent understanding of the interactions between RS and the human gut microbiota and the possible connections to liver glycogen biosynthesis to elucidate its role in the development of glycogen fragility. RS might regulate glycogen fragility in diabetes by modulating the postprandial glycemic response and glycogen biosynthesis pathways. Before RS can be applied to repair fragile glycogen, more work should be done to better understand in vivo RS structures and identify the factor binding glycogen ß particles together. This review contains important information on the connections between glycogen fragility and RS-gut microbiota interactions, which could help to better understand the health benefits of RS consumption.


Subject(s)
Gastrointestinal Microbiome , Liver Glycogen , Resistant Starch , Animals , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Liver Glycogen/metabolism , Liver Glycogen/physiology , Mice , Rats , Resistant Starch/metabolism , Resistant Starch/pharmacology
17.
Int J Biol Macromol ; 186: 227-236, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34245735

ABSTRACT

We investigated the ability of lotus seed resistant starch (LRS) to affect the conversion of sodium taurocholate (STCA) by regulating the intestinal flora, using glucose (GLU) and high amylose corn starch (HAMS) as controls. The dominant microbiota in LRS group were mainly Lactobacillus and Escherichia_Shigella, with a small proportion of Bifidobacterium. Meanwhile, Lactobacillus, Bifidobacterium and Enterococcus were dominant microbiota in the HAMS group. Lactobacillus, Burkholderia-Caballeronia-Paraburkholderia and Sphingomonas were found in the GLU group. Furthermore, Bifidobacterium, Enterococcus and Escherichia_Shigella were negatively correlated with STCA and sodium taurodeoxycholate (STDCA), while these bacteria were positively correlated with bile salt hydrolase (BSH) and hydroxysteroid dehydrogenase (HSDH) content. Meanwhile Burkholderia-Caballeronia-Paraburkholderia and Sphingomonas were positively correlated with STCA and STDCA, while these bacteria were negatively correlated with BSH and HSDH content. LRS promoted the proliferation of Bifidobacterium and Escherichia_Shigella to secret more BSH and HSDH, accelerating the hydrolysis of STCA and reducing the conversion of STDCA.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Lotus/chemistry , Prebiotics , Resistant Starch/pharmacology , Seeds/chemistry , Taurocholic Acid/metabolism , Amidohydrolases/metabolism , Animals , Bacteria/growth & development , Feces/microbiology , Fermentation , Hydrolysis , Hydroxysteroid Dehydrogenases/metabolism , Male , Rats, Sprague-Dawley , Taurodeoxycholic Acid/metabolism
18.
Int J Biol Macromol ; 183: 2272-2281, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34097970

ABSTRACT

This study aimed to investigate the synergistic effect of lotus seed resistant starch (LRS) and short-chain fatty acids (SCFAs) on mice fecal bacterial flora and the contents of SCFAs in vitro. Following 24 h of fermentation, 16S rRNA analysis revealed several differences in the fecal microbiota community structure among primal bacteria (PB), LRS and different SCFAs combined with LRS groups (SCFAs-LRS). The LRS group increased the relative abundance of Lactobacillus, Allobaculum, Clostridium, Bacteroides and Prevotella. Among the SCFAs-LRS group, AA-LRS increased the relative abundance of Prevotella, and Bacillus. PA-LRS increased abundance of Sphingomonas and the BA-LRS group significantly increased the relative abundance of Rhizobiales, Brucellaceae and Ochrobactrum. Meanwhile, propionic acid and BA productions significantly increased in the BA-LRS group. The SCFAs-LRS group elicited a beneficial effect on the fecal microbiota by increasing production of SCFAs. We highlight the fact that the combination of LRS and SCFA can increase the contents of SCFAs produced by mice fecal microbiota. In short, the combination of LRS and SCFA can influence intestinal flora by promoting the growth of beneficial bacteria and can serve as new prebiotics for promoting health and disease management.


Subject(s)
Bacteria/drug effects , Fatty Acids, Volatile/pharmacology , Gastrointestinal Microbiome/drug effects , Lotus , Prebiotics , Resistant Starch/pharmacology , Animals , Bacteria/genetics , Bacteria/growth & development , Bacteria/metabolism , Drug Synergism , Fatty Acids, Volatile/isolation & purification , Feces/microbiology , Fermentation/drug effects , Lotus/chemistry , Male , Mice, Inbred ICR , Ribotyping , Seeds/chemistry
19.
Nutrients ; 13(2)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671147

ABSTRACT

The majority of research on the physiological effects of dietary resistant starch type 2 (RS2) has focused on sources derived from high-amylose maize. In this study, we conduct a double-blind, randomized, placebo-controlled, crossover trial investigating the effects of RS2 from wheat on glycemic response, an important indicator of metabolic health, and the gut microbiota. Overall, consumption of RS2-enriched wheat rolls for one week resulted in reduced postprandial glucose and insulin responses relative to conventional wheat when participants were provided with a standard breakfast meal containing the respective treatment rolls (RS2-enriched or conventional wheat). This was accompanied by an increase in the proportions of bacterial taxa Ruminococcus and Gemmiger in the fecal contents, reflecting the composition in the distal intestine. Additionally, fasting breath hydrogen and methane were increased during RS2-enriched wheat consumption. However, although changes in fecal short-chain fatty acid (SCFA) concentrations were not significant between control and RS-enriched wheat roll consumption, butyrate and total SCFAs were positively correlated with relative abundance of Faecalibacterium, Ruminoccocus, Roseburia, and Barnesiellaceae. These effects show that RS2-enriched wheat consumption results in a reduction in postprandial glycemia, altered gut microbial composition, and increased fermentation activity relative to wild-type wheat.


Subject(s)
Blood Glucose/drug effects , Gastrointestinal Microbiome/drug effects , Resistant Starch/classification , Triticum/chemistry , Adult , Bacteria/classification , Bacteria/genetics , Cross-Over Studies , Double-Blind Method , Fatty Acids, Volatile/chemistry , Feces/chemistry , Feces/microbiology , Female , Humans , Male , Middle Aged , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Resistant Starch/pharmacology
20.
Int J Biol Macromol ; 180: 458-469, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33711371

ABSTRACT

Potato resistant starch type 3 (PRS) is helpful for weight-loss. To investigate the regulatory effects of PRS on high-fat diet (HFD)-induced obesity, different doses of PRS (5%, 15% and 25%) were fed to mice for 12 weeks. Metabolic syndrome related to obesity, intestinal microbiota composition and its metabolites as well as the relationship among them were studied. Results showed that PRS could regulate HFD-induced metabolic syndrome in a dose dependent manner; promote the proliferation of intestinal cells and expression of tight junction proteins, such as Occludin and zonula occludens (ZO)-1; reduce the Firmicutes/Bacteroidetes (F/B) rate; regulate the relative abundance of intestinal microbiota, such as Bifidobacterium, Ruminococcus, Bacteroides and Coprococcus; and promote the production of microbial metabolites, such as propionic acid and acetic acid. Besides, the alteration in the intestinal microbiota composition and metabolites were significantly correlated. It could be concluded that propionic acid and acetic acid were the two dominant metabolites of Bifidobacterium, Ruminococcus, Bacteroides, and Coprococcus, which contributed to the anti-obesity potential of PRS, metabolic syndrome alleviation, and intestinal barrier dysfunction.


Subject(s)
Bacteroides/metabolism , Bifidobacterium/metabolism , Gastrointestinal Microbiome/drug effects , Obesity/prevention & control , Resistant Starch/pharmacology , Solanum tuberosum/chemistry , Acetic Acid/metabolism , Animals , Bacteroides/drug effects , Bifidobacterium/drug effects , Body Weight/drug effects , Diet, High-Fat/adverse effects , Lipids/blood , Male , Metabolomics/methods , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Propionates/metabolism , Resistant Starch/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...