1
|
King TL, Bugam SH, Khoo SY, Tan SH, Yeo JJP, Tan SSN, Chan PY, Liew NK, Chunggat J, Tiong LL, Ibrahim AS, Tan FHS, Tiong XT. Effects of 12 Weeks consumption of sago starch on glycaemic and lipid control in patients with type 2 diabetes: A double-blind, randomised, parallel controlled trial. Clin Nutr ESPEN 2025; 68:300-308. [PMID: 40345654 DOI: 10.1016/j.clnesp.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Sago starch, rich in type 2 resistant starch, may offer benefits in glucose and lipid regulations in patients with type 2 diabetes. METHODS We conducted a double-blind, randomised, controlled trial with type 2 diabetes patients, randomised 1:1 into intervention (50g/day sago starch) and control (50g/day corn starch) arms. All participants received standardised diet counselling for diabetes management given at baseline. The primary outcomes were changes in glycaemic endpoints, insulin sensitivity, and lipid levels. Secondary outcomes included changes in calorie intake, body fat, anthropometric, and blood pressure measurements. RESULTS Of 100 patients enrolled (61 % males; mean [standard deviation or SD] age, 56.8 [8.1] years; mean [SD] haemoglobin A1C, 7.21 % [0.52]), 82 completed the 12-week follow-up and were included in primary analyses. There were no significant between-group change in haemoglobin A1C level (mean difference -0.03 % [95 % confidence interval or CI, -0.24 to 0.19]; p = 0.809), fasting glucose (-0.44 mmol/L [95 % CI, -1.08 to 0.19]; p = 0.170), fasting insulin (-0.31mIU/L [95 % CI, -2.74 to 2.11]; p = 0.797), and Homeostasis Model Assessment for Insulin Resistance (-0.22 [95 % CI, -1.04 to 0.60]; p = 0.590). No significant changes were observed in total cholesterol (0.11 mmol/L [95 % CI, -0.19 to 0.42]; p = 0.472, low-density lipoprotein-cholesterol (0.05 mmol/L [95%CI, -0.24 to 0.35], p = 0.731), high-density lipoprotein-cholesterol (0.13 mmol/L [95%CI, -0.04 to 0.29], p = 0.141), or triglycerides (0.13 mmol/L [95%CI,-0.32 to 0.57], p = 0.574). The intervention group showed a greater reduction in daily calorie intake (-166.1 kcal [95 % CI, -324.4 to -7.8]; p = 0.040) at 12-week; however, this was not statistically significant after correction for multiple comparison. No significant between-group differences were found in other secondary outcomes. CONCLUSION In this randomised controlled trial, 12-week of sago starch supplementation did not result in significant improvement in glycaemic and lipid parameters in patients with type 2 diabetes. TRIAL REGISTRATIONS 1. http://nmrr.gov.my - NMRR-21-1905-58409, 2. https://clinicaltrials.gov - NCT05599633.
Collapse
Affiliation(s)
- Teck Long King
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia.
| | - Sal Hazreen Bugam
- Downstream Technology Division, CRAUN Research Sdn. Bhd, Sarawak, Malaysia.
| | - Sing Yee Khoo
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia
| | - Shirin Hui Tan
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia
| | - John Jui Ping Yeo
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia
| | - Shirley Siang Ning Tan
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia; Department of Pharmacy, Sarawak General Hospital, Ministry of Health, Malaysia
| | - Pei Yong Chan
- Department of Dietetics and Food Services, Sarawak General Hospital, Ministry of Health, Malaysia
| | - Ngan Kai Liew
- Department of Dietetics and Food Services, Sarawak General Hospital, Ministry of Health, Malaysia
| | - Jawing Chunggat
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia
| | - Lee Len Tiong
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia
| | | | | | - Xun Ting Tiong
- Clinical Research Centre, Sarawak General Hospital, Institute for Clinical Research, National Institutes of Health, Ministry of Health, Malaysia
| |
Collapse
|
2
|
Li M, Liu X, Chen W, Xu H, Huang F, Yao Q, Jia X, Huang Y. Alleviating Effect of Lactiplantibacillus plantarum HYY-S10 on Colitis in Mice Based on an Analysis of the Immune Axis in the Intestine. Microorganisms 2025; 13:840. [PMID: 40284675 PMCID: PMC12029376 DOI: 10.3390/microorganisms13040840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
The pathogenesis of ulcerative colitis (UC) has been fundamentally associated with intestinal microbiota dysbiosis and disruption of immune homeostasis. This study systematically investigates the therapeutic potential of Lactiplantibacillus plantarum HYY-S10 (HYY-S10), a novel strain isolated from De'ang sour tea in Yun an, China, with a focus on its mechanisms for alleviating colitis through the modulation of gut microbiota. Using a dextran sulfate sodium (DSS)-induced colitis model in C57BL/6J mice, our findings demonstrated that seven days of oral supplementation with HYY-S10 (1 × 108 CFU/mL, 0.2 mL/10 g body weight) significantly improved Disease Activity Index (DAI) scores and attenuated characteristic colitis symptoms, including progressive weight loss, rectal bleeding, and abnormal stool consistency. Administration of HYY-S10 exhibited significant immunomodulatory effects characterized by the downregulation of pro-inflammatory mediators (such as IL-1β, IL-6, IFN-γ, and LPS) while concomitantly upregulating anti-inflammatory IL-10 expression. Additionally, the strain enhanced intestinal antioxidant capacity by increasing GSH-Px activity, which collectively contributed to the reduction in intestinal inflammation. Furthermore, HYY-S10 demonstrated multifaceted protective effects by ameliorating oxidative stress through the restoration of redox homeostasis and modulation of gut microbial ecology. Probiotic intervention significantly increased short-chain fatty acids (SCFAs) production and notably enhanced the relative abundance of beneficial taxa, including Akkermansia and Ruminococcus_B, while restoring microbial diversity and ecological stability. Collectively, our results demonstrate that HYY-S10 alleviates experimental colitis by modulating the intestinal immune axis and microbiota composition, providing mechanistic insights to support its potential as a probiotic-based therapeutic strategy for UC.
Collapse
Affiliation(s)
- Mengna Li
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, College of Food Science and Engineering, Foshan University, Foshan 528225, China; (M.L.)
| | - Xintong Liu
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, College of Food Science and Engineering, Foshan University, Foshan 528225, China; (M.L.)
| | - Weijian Chen
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, College of Food Science and Engineering, Foshan University, Foshan 528225, China; (M.L.)
| | - Haoyue Xu
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, College of Food Science and Engineering, Foshan University, Foshan 528225, China; (M.L.)
| | - Fang Huang
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, College of Food Science and Engineering, Foshan University, Foshan 528225, China; (M.L.)
| | - Qingbo Yao
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, College of Food Science and Engineering, Foshan University, Foshan 528225, China; (M.L.)
| | - Xiangze Jia
- School of Food Sciences and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yanyan Huang
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, College of Food Science and Engineering, Foshan University, Foshan 528225, China; (M.L.)
| |
Collapse
|
3
|
Yu W, Sun S, Yan Y, Zhou H, Liu Z, Fu Q. The role of short-chain fatty acid in metabolic syndrome and its complications: focusing on immunity and inflammation. Front Immunol 2025; 16:1519925. [PMID: 39991152 PMCID: PMC11842938 DOI: 10.3389/fimmu.2025.1519925] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Metabolic syndrome (Mets) is an important contributor to morbidity and mortality in cardiovascular, liver, neurological, and reproductive diseases. Short-chain fatty acid (SCFA), an organismal energy donor, has recently been demonstrated in an increasing number of studies to be an important molecule in ameliorating immuno-inflammation, an important causative factor of Mets, and to improve lipid distribution, blood glucose, and body weight levels in animal models of Mets. This study reviews recent research advances on SCFA in Mets from an immune-inflammatory perspective, including complications dominated by chronic inflammation, as well as the fact that these findings also contribute to the understanding of the specific mechanisms by which gut flora metabolites contribute to metabolic processes in humans. This review proposes an emerging role for SCFA in the inflammatory Mets, followed by the identification of major ambiguities to further understand the anti-inflammatory potential of this substance in Mets. In addition, this study proposes novel strategies to modulate SCFA for the treatment of Mets that may help to mitigate the prognosis of Mets and its complications.
Collapse
Affiliation(s)
- Wenqian Yu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Siyuan Sun
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yutong Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Hong Zhou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Ziyi Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Qiang Fu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Chen M, Wang R, Wang T. Gut microbiota and skin pathologies: Mechanism of the gut-skin axis in atopic dermatitis and psoriasis. Int Immunopharmacol 2024; 141:112658. [PMID: 39137625 DOI: 10.1016/j.intimp.2024.112658] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 08/15/2024]
Abstract
Atopic dermatitis (AD) and psoriasis are chronic skin diseases with a global impact, posing significant challenges to public health systems and severely affecting patients' quality of life. This review delves into the key role of the gut microbiota in these diseases, emphasizing the importance of the gut-skin axis in inflammatory mediators and immune regulation and revealing a complex bidirectional communication system. We comprehensively assessed the pathogenesis, clinical manifestations, and treatment strategies for AD and psoriasis, with a particular focus on how the gut microbiota and their metabolites influence disease progression via the gut-skin axis. In addition, personalized treatment plans based on individual patient microbiome characteristics have been proposed, offering new perspectives for future treatment approaches. We call for enhanced interdisciplinary cooperation to further explore the interactions between gut microbiota and skin diseases and to assess the potential of drugs and natural products in modulating the gut-skin axis, aiming to advance the treatment of skin diseases.
Collapse
Affiliation(s)
- Meng Chen
- Department of Dermatology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Rui Wang
- Department of Dermatology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China.
| | - Ting Wang
- Department of Dermatology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China.
| |
Collapse
|
5
|
Zhang Y, Shen W, Chen Z, He J, Feng L, Wang L, Chen S. Resistant starch reduces glycolysis by HK2 and suppresses high-fructose corn syrup-induced colon tumorigenesis. J Gastroenterol 2024; 59:905-920. [PMID: 39141107 PMCID: PMC11415400 DOI: 10.1007/s00535-024-02138-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 07/25/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND The intake of high-fructose corn syrup (HFCS) may increase the risk of colorectal cancer (CRC). This study aimed to explore the potential effects and mechanisms of resistant starch (RS) in HFCS-induced colon tumorigenesis. METHODS The azoxymethane/dextran sodium sulfate (AOM/DSS) and ApcMin/+ mice models were used to investigate the roles of HFCS and RS in CRC in vivo. An immunohistochemistry (IHC) staining analysis was used to detect the expression of proliferation-related proteins in tissues. 16S rRNA sequencing for microbial community, gas chromatography for short-chain fatty acids (SCFAs), and mass spectrometry analysis for glycolysis products in the intestines were performed. Furthermore, lactic acid assay kit was used to detect the glycolysis levels in vitro. RESULTS RS suppressed HFCS-induced colon tumorigenesis through reshaping the microbial community. Mechanistically, the alteration of the microbial community after RS supplement increased the levels of intestinal SCFAs, especially butyrate, leading to the suppression of glycolysis and CRC cell proliferation by downregulating HK2. CONCLUSIONS Our study identified RS as a candidate of protective factors in CRC and may provide a potential target for HFCS-related CRC treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiyi Shen
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhehang Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiamin He
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lijun Feng
- Department of Nutriology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lan Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
Mann ER, Lam YK, Uhlig HH. Short-chain fatty acids: linking diet, the microbiome and immunity. Nat Rev Immunol 2024; 24:577-595. [PMID: 38565643 DOI: 10.1038/s41577-024-01014-8] [Citation(s) in RCA: 231] [Impact Index Per Article: 231.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
The short-chain fatty acids (SCFAs) butyrate, propionate and acetate are microbial metabolites and their availability in the gut and other organs is determined by environmental factors, such as diet and use of antibiotics, that shape the diversity and metabolism of the microbiota. SCFAs regulate epithelial barrier function as well as mucosal and systemic immunity via evolutionary conserved processes that involve G protein-coupled receptor signalling or histone deacetylase activity. Indicatively, the anti-inflammatory role of butyrate is mediated through direct effects on the differentiation of intestinal epithelial cells, phagocytes, B cells and plasma cells, and regulatory and effector T cells. Intestinally derived SCFAs also directly and indirectly affect immunity at extra-intestinal sites, such as the liver, the lungs, the reproductive tract and the brain, and have been implicated in a range of disorders, including infections, intestinal inflammation, autoimmunity, food allergies, asthma and responses to cancer therapies. An ecological understanding of microbial communities and their interrelated metabolic states, as well as the engineering of butyrogenic bacteria may support SCFA-focused interventions for the prevention and treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Elizabeth R Mann
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ying Ka Lam
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Tang L, Wu J, Aga L, Wang N, Li Y, Li H, Wang X. Structural analysis of type 3 resistant starch from Canna edulis during in vitro simulated digestion and its post-digested residue impact on human gut microbiota. Front Nutr 2024; 11:1403497. [PMID: 38966420 PMCID: PMC11223600 DOI: 10.3389/fnut.2024.1403497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction Resistant starch (RS) has garnered attention for its health benefits, including modulating the gut microbiota and promoting the production of short-chain fatty acids (SCFAs). Methods This study investigates structural changes of type 3 resistant starch from Canna edulis (CE) during in vitro simulated digestion and explores its health-relevant properties using healthy individuals' fecal microbiota. Results CE, prepared with a RS content of 59.38%, underwent a comprehensive analysis employing X-ray diffraction (XRD), fourier-transform infrared spectroscopy (FTIR), and scanning electron microscopy (SEM). During simulated digestion, XRD analysis demonstrated a significant rise in CE's relative crystallinity from 38.92 to 49.34%. SEM illustrated the transition of CE from a smooth to a rough surface, a notable morphological shift. Post-digestion, CE was introduced into microbial fermentation. Notably, propionic acid and valeric acid levels significantly increased compared to the control group. Furthere more, beneficial Bifidobacterium proliferated while pathogenic Escherichia-Shigella was suppressed. When comparing CE to the well-known functional food fructo-oligosaccharide (FOS), CE showed a specific ability to support the growth of Bifidobacterium and stimulate the production of short-chain fatty acids (SCFAs) without causing lactic acid accumulation. Discussion CE demonstrates potential as a functional health food, with implications for gut health enhancement and SCFAs production.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xueyong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Li Y, Yang F, Liu J, Jiang M, Yu Y, Zhou Q, Sun L, Zhang Z, Zhou L. Protective effects of sodium butyrate on fluorosis in rats by regulating bone homeostasis and serum metabolism. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116284. [PMID: 38581912 DOI: 10.1016/j.ecoenv.2024.116284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
Fluorosis due to high fluoride levels in drinking water profoundly affects the development of human skeletal and dental structures. Sodium butyrate (NaB) has been found to regulate overall bone mass and prevent pathological bone loss. However, the mechanism of NaB action on fluorosis remains unclear. In this study, a rat model of fluorosis induced by 100 mg/L sodium fluoride was used to investigate the impact of NaB on bone homeostasis and serum metabolomics. It was found that NaB significantly reduced the levels of bone resorption markers CTX-Ⅰ and TRACP-5B in fluorosis rats. Moreover, NaB increased calcium and magnesium levels in bone, while decreasing phosphorus levels. In addition, NaB improved various bone microstructure parameters, including bone mineral density (BMD), trabecular thickness (Tb. Th), trabecular bone separation (Tb. SP), and structural model index (SMI) in the femur. Notably, NaB intervention also enhanced the antioxidant capacity of plasma in fluorosis rats. Furthermore, a comprehensive analysis of serum metabolomics by LC-MS revealed a significant reversal trend of seven biomarkers after the intervention of NaB. Finally, pathway enrichment analysis based on differential metabolites indicated that NaB exerted protective effects on fluorosis by modulating arginine and proline metabolic pathways. These findings suggest that NaB has a beneficial effect on fluorosis and can regulate bone homeostasis by ameliorating metabolic disorders.
Collapse
Affiliation(s)
- Ying Li
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Fengmei Yang
- School of Public Health, Shenyang Medical College, Shenyang 110034, China; Yulin Center for Disease Control and Prevention, Yulin Municipal Health Committee, Yulin 719100, China
| | - Jie Liu
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Mengqi Jiang
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Ye Yu
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Qingyi Zhou
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Lu Sun
- Radiation Health Center, Liaoning Provincial Center for Disease Control and Prevention, Shenyang 110015, China.
| | - Zhuo Zhang
- School of Public Health, Shenyang Medical College, Shenyang 110034, China.
| | - Lin Zhou
- School of Public Health, Shenyang Medical College, Shenyang 110034, China.
| |
Collapse
|
9
|
Wang N, Zhang C, Li H, Wu J, Zhang D, Li Y, Yang L, Zhang N, Wang X. Structure properties of Canna edulis RS3 (double enzyme hydrolysis) and RS4 (OS-starch and cross-linked starch): Influence on fermentation products and human gut microbiota. Int J Biol Macromol 2024; 265:130700. [PMID: 38458281 DOI: 10.1016/j.ijbiomac.2024.130700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/05/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
This study investigated the in vitro fermentation characteristics of different structural types of Canna edulis resistant starch (RS). RS3 was prepared through a double enzyme hydrolysis method, and RS4 (OS-starch and cross-linked starch) was prepared using octenyl succinic anhydride and sodium trimetaphosphate/sodium tripolyphosphate, respectively. The RS3 and RS4 samples were structurally analyzed using scanning electron microscopy, Fourier-transform infrared spectroscopy, differential scanning calorimetry, and X-ray diffraction analysis. This was followed by in vitro fermentation experiments. The results revealed microstructure differences in the two groups of starch samples. Compared to native starch, RS3 and RS4 exhibited a lower degree of order and endothermic energy, with lower crystallinity (RS3: 29.59 ± 1.11 %; RS4 [OS-starch]: 28.01 ± 1.32 %; RS4 [cross-linked starch]: 30.44 ± 1.73 %) than that in native starch (36.29 ± 0.89 %). The RS content was higher in RS3 (63.40 ± 2.85 %) and RS4 (OS-starch: 71.21 ± 1.28 %; cross-linked starch: 74.33 ± 0.643 %) than in native starch (57.71 ± 2.95 %). RS3 and RS4 exhibited slow fermentation rates, promoting the production of short-chain fatty acids. RS3 and cross-linked starch significantly increased the production of acetate and butyrate. Moreover, RS3 significantly promoted the abundance of Lactobacillus, while OS-starch and cross-linked starch significantly enhanced the abundance of Dorea and Coprococcus, respectively. Hence, the morphological structure and RS content of the samples greatly influenced the fermentation rate. Moreover, the different varieties of RS induced specific gut microbial regulation. Hence, they show potential applications in functional foods for tailored gut microbiota management.
Collapse
Affiliation(s)
- Nan Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Chi Zhang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China; School of Medicine, Linyi University, Linyi 276000, Shandong, China
| | - Houxier Li
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Jiahui Wu
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Dachuan Zhang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Yan Li
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Li Yang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Nan Zhang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Xueyong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China.
| |
Collapse
|
10
|
Cheng J, Zhou J. Unraveling the gut health puzzle: exploring the mechanisms of butyrate and the potential of High-Amylose Maize Starch Butyrate (HAMSB) in alleviating colorectal disturbances. Front Nutr 2024; 11:1285169. [PMID: 38304546 PMCID: PMC10830644 DOI: 10.3389/fnut.2024.1285169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Colorectal disturbances encompass a variety of disorders that impact the colon and rectum, such as colitis and colon cancer. Butyrate, a short-chain fatty acid, plays a pivotal role in supporting gut health by nourishing colonocytes, promoting barrier function, modulating inflammation, and fostering a balanced microbiome. Increasing colorectal butyrate concentration may serve as a critical strategy to improve colon function and reduce the risk of colorectal disturbances. Butyrylated high-amylose maize starch (HAMSB) is an edible ingredient that efficiently delivers butyrate to the colon. HAMSB is developed by esterifying a high-amylose starch backbone with butyric anhydride. With a degree of substitution of 0.25, each hydroxy group of HAMSB is substituted by a butyryl group in every four D-glucopyranosyl units. In humans, the digestibility of HAMSB is 68% (w/w), and 60% butyrate molecules attached to the starch backbone is absorbed by the colon. One clinical trial yielded two publications, which showed that HAMSB significantly reduced rectal O6-methyl-guanine adducts and epithelial proliferation induced by the high protein diet. Fecal microbial profiles were assessed in three clinical trials, showing that HAMSB supplementation was consistently linked to increased abundance of Parabacteroides distasonis. In animal studies, HAMSB was effective in reducing the risk of diet- or AOM-induced colon cancer by reducing genetic damage, but the mechanisms differed. HAMSB functioned through affecting cecal ammonia levels by modulating colon pH in diet-induced cancer, while it ameliorated chemical-induced colon cancer through downregulating miR19b and miR92a expressions and subsequently activating the caspase-dependent apoptosis. Furthermore, animal studies showed that HAMSB improved colitis via regulating the gut immune modulation by inhibiting histone deacetylase and activating G protein-coupled receptors, but its role in bacteria-induced colon colitis requires further investigation. In conclusion, HAMSB is a food ingredient that may deliver butyrate to the colon to support colon health. Further clinical trials are warranted to validate earlier findings and determine the minimum effective dose of HAMSB.
Collapse
Affiliation(s)
- Junrui Cheng
- Global Scientific and Regulatory Department, Ingredion Incorporated, Bridgewater, NJ, United States
| | - Jing Zhou
- Global Scientific and Regulatory Department, Ingredion Incorporated, Bridgewater, NJ, United States
| |
Collapse
|
11
|
López-Enríquez S, Múnera-Rodríguez AM, Leiva-Castro C, Sobrino F, Palomares F. Modulation of the Immune Response to Allergies Using Alternative Functional Foods. Int J Mol Sci 2023; 25:467. [PMID: 38203638 PMCID: PMC10779275 DOI: 10.3390/ijms25010467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/14/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Modulation of the allergic immune response through alternative therapies is a field of study that aims to address allergic reactions differently from traditional approaches. These therapies encompass the utilization of natural functional foods, which have been observed to exert an influence on the immune response, thus mitigating the severity of allergies. Indeed, some studies suggest that the incorporation of these nutraceuticals can regulate immune function, leading to a reduction in histamine release and subsequent alleviation of allergic symptoms. Moreover, certain herbs and dietary supplements, such as curcumin, are believed to possess anti-inflammatory properties, which may serve to moderate allergic responses. Although the results remain somewhat mixed and require further research, these alternative therapies exhibit the potential to impact the allergic immune response, thereby providing complementary options to conventional treatments. Therefore, in this review, we aim to provide an updated account of functional foods capable of modulating the immune response to allergies. In that sense, the review delves into functional foods sourced from plants (phytochemicals), animals, and marine algae. Emphasis is placed on their potential application in the treatment of allergic disorders. It also provides an overview of how these foods can be effectively utilized as functional foods. Additionally, it explores the molecular mechanisms and scientific validity of various bioactive natural compounds in the management of allergies.
Collapse
Affiliation(s)
- Soledad López-Enríquez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Avenue Sanchez Pizjuan s/n, 41009 Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, Virgen Macarena University Hospital, University of Seville, CSIC, 41013 Seville, Spain
| | - Ana M Múnera-Rodríguez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Avenue Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Camila Leiva-Castro
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Avenue Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Francisco Sobrino
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Avenue Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Francisca Palomares
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Avenue Sanchez Pizjuan s/n, 41009 Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, Virgen Macarena University Hospital, University of Seville, CSIC, 41013 Seville, Spain
| |
Collapse
|
12
|
Wen JJ, Li MZ, Nie SP. Dietary supplementation with resistant starch contributes to intestinal health. Curr Opin Clin Nutr Metab Care 2023; 26:334-340. [PMID: 37057658 DOI: 10.1097/mco.0000000000000939] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
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.
Collapse
Affiliation(s)
- Jia-Jia Wen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | | | | |
Collapse
|
13
|
Mirzaei R, Kavyani B, Nabizadeh E, Kadkhoda H, Asghari Ozma M, Abdi M. Microbiota metabolites in the female reproductive system: Focused on the short-chain fatty acids. Heliyon 2023; 9:e14562. [PMID: 36967966 PMCID: PMC10031489 DOI: 10.1016/j.heliyon.2023.e14562] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
Several disorders have been linked to modifications in the gut microbial imbalance, intestinal epithelium, and host immune system. In this regard, microbiota derived short-chain fatty acids (SCFAs) play a key function in the regulation of histone deacetylases (HDACs), which affect modulation of immunity and regulation of inflammatory responses in the intestine and other organs. Studies examining the metabolites produced by polymicrobial bacterial vaginosis (BV) states and Lactobacillus-dominated microbiota have noted a dramatic reduction of lactic acid and a shift toward SCFA synthesis. Along with higher levels of SCFAs, acetate is typically the main metabolite in the cervicovaginal fluid of women with symptomatic bacterial vaginosis. The fact that SCFAs made by the vaginal microbiota have been shown to exhibit antibacterial and immune-modulating properties suggests that they may have promise as indicators of disease and/or disease susceptibility. In this review, we overview and summarize the current findings on the detrimental or protective roles of microbiota metabolites especially SCFAs in the health and disease of the female reproductive system.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Batoul Kavyani
- Department of Medical Microbiology (Bacteriology & Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Edris Nabizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hiva Kadkhoda
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Asghari Ozma
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Abdi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
14
|
Haskey N, Gold SL, Faith JJ, Raman M. To Fiber or Not to Fiber: The Swinging Pendulum of Fiber Supplementation in Patients with Inflammatory Bowel Disease. Nutrients 2023; 15:nu15051080. [PMID: 36904081 PMCID: PMC10005525 DOI: 10.3390/nu15051080] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Evidence-based dietary guidance around dietary fiber in inflammatory bowel disease (IBD) has been limited owing to insufficient reproducibility in intervention trials. However, the pendulum has swung because of our increased understanding of the importance of fibers in maintaining a health-associated microbiome. Preliminary evidence suggests that dietary fiber can alter the gut microbiome, improve IBD symptoms, balance inflammation, and enhance health-related quality of life. Therefore, it is now more vital than ever to examine how fiber could be used as a therapeutic strategy to manage and prevent disease relapse. At present, there is limited knowledge about which fibers are optimal and in what form and quantity they should be consumed to benefit patients with IBD. Additionally, individual microbiomes play a strong role in determining the outcomes and necessitate a more personalized nutritional approach to implementing dietary changes, as dietary fiber may not be as benign as once thought in a dysbiotic microbiome. This review describes dietary fibers and their mechanism of action within the microbiome, details novel fiber sources, including resistant starches and polyphenols, and concludes with potential future directions in fiber research, including the move toward precision nutrition.
Collapse
Affiliation(s)
- Natasha Haskey
- Department of Biology, The Irving K. Barber Faculty of Science, University of British Columbia—Okanagan, 3187 University Way, Kelowna, BC V1V 1V7, Canada
- Division of Gastroenterology, Cumming School of Medicine, University of Calgary, 6D33 TRW Building, 3280 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Stephanie L. Gold
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Jeremiah J. Faith
- Precision Immunology Institute and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Maitreyi Raman
- Division of Gastroenterology, Cumming School of Medicine, University of Calgary, 6D33 TRW Building, 3280 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- Correspondence:
| |
Collapse
|
15
|
Stec A, Sikora M, Maciejewska M, Paralusz-Stec K, Michalska M, Sikorska E, Rudnicka L. Bacterial Metabolites: A Link between Gut Microbiota and Dermatological Diseases. Int J Mol Sci 2023; 24:ijms24043494. [PMID: 36834904 PMCID: PMC9961773 DOI: 10.3390/ijms24043494] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Dysbiosis has been identified in many dermatological conditions (e.g., psoriasis, atopic dermatitis, systemic lupus erythematosus). One of the ways by which the microbiota affect homeostasis is through microbiota-derived molecules (metabolites). There are three main groups of metabolites: short-chain fatty acids (SCFAs), tryptophan metabolites, and amine derivatives including trimethylamine N-oxide (TMAO). Each group has its own uptake and specific receptors through which these metabolites can exert their systemic function. This review provides up-to-date knowledge about the impact that these groups of gut microbiota metabolites may have in dermatological conditions. Special attention is paid to the effect of microbial metabolites on the immune system, including changes in the profile of the immune cells and cytokine disbalance, which are characteristic of several dermatological diseases, especially psoriasis and atopic dermatitis. Targeting the production of microbiota metabolites may serve as a novel therapeutic approach in several immune-mediated dermatological diseases.
Collapse
Affiliation(s)
- Albert Stec
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland
| | - Mariusz Sikora
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland
- Correspondence:
| | - Magdalena Maciejewska
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland
| | - Karolina Paralusz-Stec
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland
| | - Milena Michalska
- Department of General, Vascular and Transplant Surgery, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Ewa Sikorska
- Department of Experimental and Clinical Physiology Center for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82A, 02-008 Warsaw, Poland
| |
Collapse
|
16
|
Hodgkinson K, El Abbar F, Dobranowski P, Manoogian J, Butcher J, Figeys D, Mack D, Stintzi A. Butyrate's role in human health and the current progress towards its clinical application to treat gastrointestinal disease. Clin Nutr 2023; 42:61-75. [PMID: 36502573 DOI: 10.1016/j.clnu.2022.10.024] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/17/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Butyrate is a key energy source for colonocytes and is produced by the gut microbiota through fermentation of dietary fiber. Butyrate is a histone deacetylase inhibitor and also signals through three G-protein coupled receptors. It is clear that butyrate has an important role in gastrointestinal health and that butyrate levels can impact both host and microbial functions that are intimately coupled with each other. Maintaining optimal butyrate levels improves gastrointestinal health in animal models by supporting colonocyte function, decreasing inflammation, maintaining the gut barrier, and promoting a healthy microbiome. Butyrate has also shown protective actions in the context of intestinal diseases such as inflammatory bowel disease, graft-versus-host disease of the gastrointestinal tract, and colon cancer, whereas lower levels of butyrate and/or the microbes which are responsible for producing this metabolite are associated with disease and poorer health outcomes. However, clinical efforts to increase butyrate levels in humans and reverse these negative outcomes have generated mixed results. This article discusses our current understanding of the molecular mechanisms of butyrate action with a focus on the gastrointestinal system, the links between host and microbial factors, and the efforts that are currently underway to apply the knowledge gained from the bench to bedside.
Collapse
Affiliation(s)
- Kendra Hodgkinson
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Faiha El Abbar
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Peter Dobranowski
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Juliana Manoogian
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - James Butcher
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daniel Figeys
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - David Mack
- Department of Paediatrics, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L1, Canada; Children's Hospital of Eastern Ontario Inflammatory Bowel Disease Centre and Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Alain Stintzi
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
17
|
Unlocking the Potential of High-Amylose Starch for Gut Health: Not All Function the Same. FERMENTATION 2023. [DOI: 10.3390/fermentation9020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
High-amylose starch has unique functional properties and nutritional values in food applications. This type of starch is generally resistant to enzymatic digestion in the gastrointestinal tract, and contains an increased fraction of resistant starch (RS), which is a type of dietary fiber. The digestion and fermentation of high-amylose starch in the gut are of current research interest, as the processes are related to its nutritional functionality. This review summarizes recent in vitro and in vivo studies on the digestion and fermentation of high-amylose starches from different botanical sources and those that have been obtained by modifications. The RS content and fermentation properties are compared among high-amylose starches. This review aims to provide a current understanding of the relationship between high-amylose starch structures and fermentation-related nutritional properties. The results of these studies suggest that both modifications and food processing of high-amylose starch result in distinct fermentation products and nutritional properties. The review provides insight into the potential future applications of diverse high-amylose starches as bioactive compounds to modulate colonic fermentation.
Collapse
|
18
|
Thompson MS, Hui Yan T, Saari N, Sarbini SR. A review: Resistant starch, a promising prebiotic for obesity and weight management. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
19
|
Drake AM, Coughlan MT, Christophersen CT, Snelson M. Resistant Starch as a Dietary Intervention to Limit the Progression of Diabetic Kidney Disease. Nutrients 2022; 14:4547. [PMID: 36364808 PMCID: PMC9656781 DOI: 10.3390/nu14214547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 08/15/2023] Open
Abstract
Diabetes is the leading cause of kidney disease, and as the number of individuals with diabetes increases there is a concomitant increase in the prevalence of diabetic kidney disease (DKD). Diabetes contributes to the development of DKD through a number of pathways, including inflammation, oxidative stress, and the gut-kidney axis, which may be amenable to dietary therapy. Resistant starch (RS) is a dietary fibre that alters the gut microbial consortium, leading to an increase in the microbial production of short chain fatty acids. Evidence from animal and human studies indicate that short chain fatty acids are able to attenuate inflammatory and oxidative stress pathways, which may mitigate the progression of DKD. In this review, we evaluate and summarise the evidence from both preclinical models of DKD and clinical trials that have utilised RS as a dietary therapy to limit the progression of DKD.
Collapse
Affiliation(s)
- Anna M. Drake
- Glycation, Nutrition and Metabolism Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne 3004, Australia
| | - Melinda T. Coughlan
- Glycation, Nutrition and Metabolism Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne 3004, Australia
- Baker Heart & Diabetes Institute, Melbourne 3004, Australia
| | - Claus T. Christophersen
- School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
- WA Human Microbiome Collaboration Centre, School of Molecular Life Sciences, Curtin University, Bentley 6102, Australia
| | - Matthew Snelson
- Glycation, Nutrition and Metabolism Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne 3004, Australia
| |
Collapse
|
20
|
Rojas-Velázquez L, Morán P, Serrano-Vázquez A, Portillo-Bobadilla T, González E, Pérez-Juárez H, Hernández E, Partida-Rodríguez O, Nieves-Ramírez M, Padilla A, Zaragoza M, Ximénez C. The regulatory function of Blastocystis spp. on the immune inflammatory response in the gut microbiome. Front Cell Infect Microbiol 2022; 12:967724. [PMID: 36118018 PMCID: PMC9470931 DOI: 10.3389/fcimb.2022.967724] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/04/2022] [Indexed: 11/22/2022] Open
Abstract
Blastocystis spp. is a unicellular organism that resides in digestive tract of various vertebrates, with a worldwide distribution and a variable prevalence. For many years, Blastocystis spp. was considered a cyst of a flagellate, a fungus, or a saprophyte yeast of the digestive tract; in 1996, it is placed in the group of stramenopiles (heterokonts). Since its new classification, many questions have arisen around this protist about its role as a pathogen or non-pathogen organism. Recent evidence indicates that Blastocystis spp. participates in the immune inflammatory response in the intestinal microbiome generating an anti-inflammatory response, showing a lower concentration of fecal inflammatory markers in infected human hosts. Here, we review recent findings on the regulatory function of Blastocystis spp. in the immune inflammatory response to comprehend the purpose of Blastocystis spp. in health and disease, defining if Blastocystis spp. is really a pathogen, a commensal or even a mutualist in the human gut microbiome.
Collapse
Affiliation(s)
- Liliana Rojas-Velázquez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- *Correspondence: Liliana Rojas-Velázquez, ; Cecilia Ximénez,
| | - Patricia Morán
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Angélica Serrano-Vázquez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Tobías Portillo-Bobadilla
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México (UNAM) e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Enrique González
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Horacio Pérez-Juárez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Eric Hernández
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Oswaldo Partida-Rodríguez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Miriam Nieves-Ramírez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Angeles Padilla
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Martha Zaragoza
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Cecilia Ximénez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- *Correspondence: Liliana Rojas-Velázquez, ; Cecilia Ximénez,
| |
Collapse
|
21
|
|