1
|
Xia D, Jin L, Wang B, Jin Y, Zheng Q, Xu J, Chen S. Alpha-glucosidase inhibitor decreases the risk of colorectal adenoma in the aged with Type 2 diabetes. Sci Rep 2025; 15:583. [PMID: 39748054 PMCID: PMC11696837 DOI: 10.1038/s41598-024-84294-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
The rapidly aging population is fueling a surge in diabetes, especially Type 2, which heightens colorectal cancer (CRC) risk. Colorectal adenoma, a precursor, compounds this trend. Although alpha-glucosidase inhibitors are effective hypoglycemic drugs working in the GI tract, the link between them and colorectal adenoma formation remains unexplored. A retrospective cross-sectional study was conducted on type 2 diabetes patients aged 60 and above using data from Wenzhou Central Hospital from January 2021 to May 2024. We used multivariable logistic regression and propensity score matching analysis (PSM) to calculate adjusted ORs for colorectal adenoma, controlling for potential confounders. A total of 311 subjects were enrolled in the study, with a mean age of 67.55 years. 138 (44.4%) were diagnosed with colorectal adenoma. Multivariate logistic regression analysis revealed that the AGI (Alpha-glucosidase inhibitor) Group had an adjusted OR of 0.399 (95% CI = 0.22-0.723, p = 0.002) compared to those with AGI free people. A similar trend was also observed in the PSM analysis (OR = 0.362, 95% CI = 0.176-0.744, p = 0.004). Subgroup analysis reveals hypertension as a potential modulator of the inverse relationship between AGI and colorectal adenoma occurrence post-PSM (p = 0.049). And AGI reduces serum iron levels, both before (p = 0.01) and after PSM (p = 0.028). In summary, our findings indicate that AGI significantly mitigates the risk of colorectal adenoma among individuals aged 60 and above, particularly among those afflicted with hypertension. Additionally, it substantially decreases serum iron levels.
Collapse
Affiliation(s)
- Dingchao Xia
- Department of Infectious Diseases, Wenzhou Central Hospital, Wenzhou, 325000, Zhejiang, China
- Department of Infectious Diseases, Wenzhou Sixth People's Hospital, Wenzhou, 325000, Zhejiang, China
| | - Lanling Jin
- Department of Neurology, Pujiang County People's Hospital, Wenzhou, Jinhua, 322200, Zhejiang, China
| | - Binfeng Wang
- Department of Gastroenterology, Affiliated Yueqing Hospital,Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yi Jin
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qun Zheng
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jie Xu
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Senzhong Chen
- Department of Gerontology, Wenzhou Central Hospital, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
2
|
Brown HA, Morris AL, Pudlo NA, Hopkins AE, Martens EC, Golob JL, Koropatkin NM. Acarbose impairs gut Bacteroides growth by targeting intracellular glucosidases. mBio 2024; 15:e0150624. [PMID: 39565129 PMCID: PMC11633381 DOI: 10.1128/mbio.01506-24] [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: 05/16/2024] [Accepted: 10/08/2024] [Indexed: 11/21/2024] Open
Abstract
Acarbose is a type 2 diabetes medicine that prevents dietary starch breakdown into glucose by inhibiting host amylase and glucosidase enzymes. Numerous gut species in the Bacteroides genus enzymatically break down starch and change in relative abundance within the gut microbiome in acarbose-treated individuals. To mechanistically explain this observation, we used two model starch-degrading Bacteroides, Bacteroides ovatus (Bo), and Bacteroides thetaiotaomicron (Bt). Bt growth on starch polysaccharides is severely impaired by acarbose, whereas Bo growth is much less affected by the drug. The Bacteroides use a starch utilization system (Sus) to grow on starch. We hypothesized that Bo and Bt Sus enzymes are differentially inhibited by acarbose. Instead, we discovered that although acarbose primarily targets the Sus periplasmic GH97 enzymes in both organisms, the drug affects starch processing at multiple other points. Acarbose competes for transport through the TonB-dependent SusC proteins and binds to the Sus transcriptional regulators. Furthermore, Bo expresses a non-Sus GH97 (BoGH97D) when grown in starch with acarbose. The Bt homolog, BtGH97H, is not expressed in the same conditions, nor can overexpression of BoGH97D complement the Bt growth inhibition in the presence of acarbose. This work informs us about unexpected complexities of Sus function and regulation in Bacteroides, including variation between related species. Furthermore, this indicates that the gut microbiome may be a source of variable response to acarbose treatment for diabetes. IMPORTANCE Acarbose is a type 2 diabetes medication that works primarily by stopping starch breakdown into glucose in the small intestine. This is accomplished by the inhibition of host enzymes, leading to better blood sugar control via reduced ability to derive glucose from dietary starches. The drug and undigested starch travel to the large intestine where acarbose interferes with the ability of some bacteria to grow on starch. However, little is known about how gut bacteria interact with acarbose, including microbes that can use starch as a carbon source. Here, we show that two gut species, Bacteroides ovatus (Bo) and Bacteroides thetaiotaomicron (Bt), respond differently to acarbose: Bt growth is inhibited by acarbose, while Bo growth is less affected. We reveal a complex set of mechanisms involving differences in starch import and sensing behind the different Bo and Bt responses. This indicates the gut microbiome may be a source of variable response to acarbose treatment for diabetes via complex mechanisms in common gut microbes.
Collapse
Affiliation(s)
- Haley A. Brown
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Adeline L. Morris
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nicholas A. Pudlo
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ashley E. Hopkins
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Eric C. Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jonathan L. Golob
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nicole M. Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Brown HA, Morris AL, Pudlo NA, Hopkins AE, Martens EC, Golob JL, Koropatkin NM. Acarbose Impairs Gut Bacteroides Growth by Targeting Intracellular GH97 Enzymes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595031. [PMID: 38826241 PMCID: PMC11142093 DOI: 10.1101/2024.05.20.595031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Acarbose is a type-2 diabetes medicine that inhibits dietary starch breakdown into glucose by inhibiting host amylase and glucosidase enzymes. Numerous gut species in the Bacteroides genus enzymatically break down starch and change in relative abundance within the gut microbiome in acarbose-treated individuals. To mechanistically explain this observation, we used two model starch-degrading Bacteroides, Bacteroides ovatus (Bo) and Bacteroides thetaiotaomicron (Bt). Bt growth is severely impaired by acarbose whereas Bo growth is not. The Bacteroides use a starch utilization system (Sus) to grow on starch. We hypothesized that Bo and Bt Sus enzymes are differentially inhibited by acarbose. Instead, we discovered that although acarbose primarily targets the Sus periplasmic GH97 enzymes in both organisms, the drug affects starch processing at multiple other points. Acarbose competes for transport through the Sus beta-barrel proteins and binds to the Sus transcriptional regulators. Further, Bo expresses a non-Sus GH97 (BoGH97D) when grown in starch with acarbose. The Bt homolog, BtGH97H, is not expressed in the same conditions, nor can overexpression of BoGH97D complement the Bt growth inhibition in the presence of acarbose. This work informs us about unexpected complexities of Sus function and regulation in Bacteroides, including variation between related species. Further, this indicates that the gut microbiome may be a source of variable response to acarbose treatment for diabetes.
Collapse
Affiliation(s)
- Haley A. Brown
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Adeline L. Morris
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nicholas A. Pudlo
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ashley E. Hopkins
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Eric C. Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jonathan L. Golob
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nicole M. Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
4
|
Lui O, Dridi L, Gonzalez E, Yasmine S, Kubinski R, Billings H, Bohlmann J, Withers SG, Maurice C, Castagner B. Characterizing the Effect of Amylase Inhibitors on Maltodextrin Metabolism by Gut Bacteria Using Fluorescent Glycan Labeling. ACS Chem Biol 2023; 18:356-366. [PMID: 36728836 PMCID: PMC9942685 DOI: 10.1021/acschembio.2c00791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/29/2022] [Indexed: 02/03/2023]
Abstract
Diet-derived polysaccharides are an important carbon source for gut bacteria and shape the human gut microbiome. Acarbose, a compound used clinically to treat type 2 diabetes, is known to inhibit the growth of some bacteria on starches based on its activity as an inhibitor of α-glucosidases and α-amylases. In contrast to acarbose, montbretin A, a new drug candidate for the treatment of type 2 diabetes, has been reported to be more specific for the inhibition of α-amylase, notably human pancreatic α-amylase. However, the effects of both molecules on glycan metabolism across a larger diversity of human gut bacteria remain to be characterized. Here, we used ex vivo metabolic labeling of a human microbiota sample with fluorescent maltodextrin to identify gut bacteria affected by amylase inhibitors. Metabolic labeling was performed in the presence and absence of amylase inhibitors, and the fluorescently labeled bacteria were identified by fluorescence-activated cell sorting coupled with 16S rDNA amplicon sequencing. We validated the labeling results in cultured isolates and identified four gut bacteria species whose metabolism of maltodextrin is inhibited by acarbose. In contrast, montbretin A slowed the growth of only one species, supporting the fact that it is more selective. Metabolic labeling is a valuable tool to characterize glycan metabolism in microbiota samples and could help understand the untargeted impact of drugs on the human gut microbiota.
Collapse
Affiliation(s)
- Olivia Lui
- Department
of Pharmacology & Therapeutics, McGill
University, Montreal, Quebec H3G 1Y6, Canada
| | - Lharbi Dridi
- Department
of Pharmacology & Therapeutics, McGill
University, Montreal, Quebec H3G 1Y6, Canada
| | - Emmanuel Gonzalez
- Canadian
Centre for Computational Genomics, McGill Genome Center, Montreal, Quebec H3A 0G1, Canada
- Department
of Human Genetics, McGill University, Montreal, Quebec H3A 0C7, Canada
- Gerald
Bronfman Department of Oncology, McGill
University, Montreal, Quebec H4A 3T2, Canada
| | - Suraya Yasmine
- Department
of Pharmacology & Therapeutics, McGill
University, Montreal, Quebec H3G 1Y6, Canada
| | - Ryszard Kubinski
- Department
of Pharmacology & Therapeutics, McGill
University, Montreal, Quebec H3G 1Y6, Canada
| | - Hannah Billings
- Department
of Pharmacology & Therapeutics, McGill
University, Montreal, Quebec H3G 1Y6, Canada
| | - Joerg Bohlmann
- Michael
Smith Laboratories, University of British
Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Department
of Botany, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Department
of Forest and Conservation Sciences, University
of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Stephen G Withers
- Michael
Smith Laboratories, University of British
Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Department
of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Department
of Chemistry, Faculty of Science, University
of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Corinne Maurice
- Department
of Microbiology & Immunology, McGill
University, Montreal, Quebec H3A 2B4, Canada
| | - Bastien Castagner
- Department
of Pharmacology & Therapeutics, McGill
University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
5
|
Senftleber NK, Ramne S, Moltke I, Jørgensen ME, Albrechtsen A, Hansen T, Andersen MK. Genetic Loss of Sucrase-Isomaltase Function: Mechanisms, Implications, and Future Perspectives. Appl Clin Genet 2023; 16:31-39. [PMID: 36994449 PMCID: PMC10041990 DOI: 10.2147/tacg.s401712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/10/2023] [Indexed: 03/31/2023] Open
Abstract
Genetic variants causing loss of sucrase-isomaltase (SI) function result in malabsorption of sucrose and starch components and the condition congenital sucrase-isomaltase deficiency (CSID). The identified genetic variants causing CSID are very rare in all surveyed populations around the globe, except the Arctic-specific c.273_274delAG loss-of-function (LoF) variant, which is common in the Greenlandic Inuit and other Arctic populations. In these populations, it is, therefore, possible to study people with loss of SI function in an unbiased way to elucidate the physiological function of SI, and investigate both short-term and long-term health effects of reduced small intestinal digestion of sucrose and starch. Importantly, a recent study of the LoF variant in Greenlanders reported that adult homozygous carriers have a markedly healthier metabolic profile. These findings indicate that SI inhibition could potentially improve metabolic health also in individuals not carrying the LoF variant, which is of great interest considering the massive number of individuals with obesity and type 2 diabetes worldwide. Therefore, the objectives of this review, are 1) to describe the biological role of SI, 2) to describe the metabolic impact of the Arctic SI LoF variant, 3) to reflect on potential mechanisms linking reduced SI function to metabolic health, and 4) to discuss what knowledge is necessary to properly evaluate whether SI inhibition is a potential therapeutic target for improving cardiometabolic health.
Collapse
Affiliation(s)
- Ninna Karsbæk Senftleber
- Clinical Research, Copenhagen University Hospital – Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Stina Ramne
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Moltke
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marit Eika Jørgensen
- Clinical Research, Copenhagen University Hospital – Steno Diabetes Center Copenhagen, Herlev, Denmark
- Centre for Public Health in Greenland, National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- Steno Diabetes Center Greenland, Nuuk, Greenland
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Correspondence: Mette K Andersen, University of Copenhagen, Blegdamsvej 3B, Mærsk Tårnet, 8. sal, 2200 København N., Copenhagen, Denmark, Tel +45 35325282, Email
| |
Collapse
|
6
|
Oku T, Nakamura S, Takeshita T, Hashiguchi M, Tanabe K. Metabolic Fate and Expectation of Health Benefits of [U- 14C]-Sucrose Inhibited from Digestion Using Morus alba Leaf Extract. J Nutr Sci Vitaminol (Tokyo) 2022; 68:294-302. [PMID: 36047101 DOI: 10.3177/jnsv.68.294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Morus alba leaf extract (MLE), a strong inhibitor of sucrase, suppresses blood glucose elevation following sucrose ingestion. To investigate that sucrose inhibited from digestion using MLE is utilized through gut microbiota, [U-14C]-sucrose solutions with or without MLE were administered orally to conventional and antibiotic-treated rats, and the excretion of 14CO2 and H2 produced by gut microbiota was measured for 24 h. After an administration of [U-14C]-sucrose to conventional rats, the unit excreted 14CO2 peaked at 4 h, and the cumulative 14CO2 excreted over 24 h was approximately 60% of the radioactivity administered. No H2 was excreted. Following an administration of [U-14C]-sucrose and MLE in conventional rats, the unit excreted 14CO2 peaked later, at 8 h, and was significantly lower (p<0.05). The cumulative 14CO2 excreted over 24 h was equal in both groups, although there was a time lag of 2-3 h in rats given [U-14C]-sucrose and MLE. The amount of H2 excreted by these rats peaked 8 h after administration. Following the administration of [U-14C]-sucrose and MLE to antibiotic-treated rats, the unit excreted 14CO2 peaked lower, and the cumulative 14CO2 excretion over 24 h was approximately 40%. In this group, H2 was minimally excreted. H2 and 14CO2 produced by gut microbiota were excreted simultaneously. In conclusion, sucrose inhibited from digestion using MLE was fermented spontaneously by gut microbiota and was not excreted into feces. In addition, it confirmed that H2 excretion could be used directly to indicate the degree of fermentation of nondigestible carbohydrates.
Collapse
Affiliation(s)
- Tsuneyuki Oku
- Institute of International Nutrition & Health, Jumonji University.,Department of Nutritional Science, University of Nagasaki Siebold
| | - Sadako Nakamura
- Institute of International Nutrition & Health, Jumonji University
| | - Tomoya Takeshita
- Department of Nutritional Science, University of Nagasaki Siebold
| | | | - Kenichi Tanabe
- Faculty of Nutrition Sciences, Nakamura Gakuen University
| |
Collapse
|
7
|
Andersen MK, Skotte L, Jørsboe E, Polito R, Stæger FF, Aldiss P, Hanghøj K, Waples RK, Santander CG, Grarup N, Dahl-Petersen IK, Diaz LJ, Overvad M, Senftleber NK, Søborg B, Larsen CVL, Lemoine C, Pedersen O, Feenstra B, Bjerregaard P, Melbye M, Jørgensen ME, Færgeman NJ, Koch A, Moritz T, Gillum MP, Moltke I, Hansen T, Albrechtsen A. Loss of Sucrase-Isomaltase Function Increases Acetate Levels and Improves Metabolic Health in Greenlandic Cohorts. Gastroenterology 2022; 162:1171-1182.e3. [PMID: 34914943 DOI: 10.1053/j.gastro.2021.12.236] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The sucrase-isomaltase (SI) c.273_274delAG loss-of-function variant is common in Arctic populations and causes congenital sucrase-isomaltase deficiency, which is an inability to break down and absorb sucrose and isomaltose. Children with this condition experience gastrointestinal symptoms when dietary sucrose is introduced. We aimed to describe the health of adults with sucrase-isomaltase deficiency. METHODS The association between c.273_274delAG and phenotypes related to metabolic health was assessed in 2 cohorts of Greenlandic adults (n = 4922 and n = 1629). A sucrase-isomaltase knockout (Sis-KO) mouse model was used to further elucidate the findings. RESULTS Homozygous carriers of the variant had a markedly healthier metabolic profile than the remaining population, including lower body mass index (β [standard error], -2.0 [0.5] kg/m2; P = 3.1 × 10-5), body weight (-4.8 [1.4] kg; P = 5.1 × 10-4), fat percentage (-3.3% [1.0%]; P = 3.7 × 10-4), fasting triglyceride (-0.27 [0.07] mmol/L; P = 2.3 × 10-6), and remnant cholesterol (-0.11 [0.03] mmol/L; P = 4.2 × 10-5). Further analyses suggested that this was likely mediated partly by higher circulating levels of acetate observed in homozygous carriers (β [standard error], 0.056 [0.002] mmol/L; P = 2.1 × 10-26), and partly by reduced sucrose uptake, but not lower caloric intake. These findings were verified in Sis-KO mice, which, compared with wild-type mice, were leaner on a sucrose-containing diet, despite similar caloric intake, had significantly higher plasma acetate levels in response to a sucrose gavage, and had lower plasma glucose level in response to a sucrose-tolerance test. CONCLUSIONS These results suggest that sucrase-isomaltase constitutes a promising drug target for improvement of metabolic health, and that the health benefits are mediated by reduced dietary sucrose uptake and possibly also by higher levels of circulating acetate.
Collapse
Affiliation(s)
- Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Line Skotte
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Emil Jørsboe
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ryan Polito
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederik F Stæger
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Aldiss
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Hanghøj
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ryan K Waples
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Cindy G Santander
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Inger K Dahl-Petersen
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Lars J Diaz
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | | | - Ninna K Senftleber
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Bolette Søborg
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Christina V L Larsen
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark; Greenland Centre for Health Research, University of Greenland, Nuuk, Greenland
| | - Clara Lemoine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Bjerregaard
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Marit E Jørgensen
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Gentofte, Denmark; Greenland Centre for Health Research, University of Greenland, Nuuk, Greenland
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Anders Koch
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark; Greenland Centre for Health Research, University of Greenland, Nuuk, Greenland; Department of Infectious Diseases, Rigshospitalet University Hospital, Copenhagen, Denmark
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthew P Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Moltke
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Wu B, Yan J, Yang J, Xia Y, Li D, Zhang F, Cao H. Extension of the Life Span by Acarbose: Is It Mediated by the Gut Microbiota? Aging Dis 2022; 13:1005-1014. [PMID: 35855337 PMCID: PMC9286917 DOI: 10.14336/ad.2022.0117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Acarbose can extend the life span of mice through a process involving the gut microbiota. Several factors affect the life span, including mitochondrial function, cellular senescence, telomere length, immune function, and expression of longevity-related genes. In this review, the effects of acarbose-regulated gut microbiota on the life span-influencing factors have been discussed. In addition, a novel theoretical basis for improving our understanding of the mechanisms by which acarbose extends the life span of mice has been suggested.
Collapse
Affiliation(s)
- Baiyun Wu
- Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China.
- School of Medicine, Nantong University, Nantong, China.
| | - Jiai Yan
- Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Ju Yang
- Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Yanping Xia
- Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Dan Li
- Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Feng Zhang
- Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Correspondence should be addressed to: Dr. Hong Cao, () and Dr. Feng Zhang (), Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hong Cao
- Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi, China.
- Correspondence should be addressed to: Dr. Hong Cao, () and Dr. Feng Zhang (), Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
9
|
Muribaculaceae Genomes Assembled from Metagenomes Suggest Genetic Drivers of Differential Response to Acarbose Treatment in Mice. mSphere 2021; 6:e0085121. [PMID: 34851167 PMCID: PMC8636109 DOI: 10.1128/msphere.00851-21] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The drug acarbose is used to treat diabetes and, by inhibiting α-amylase in the small intestine, increases the amount of starch entering the lower digestive tract. This results in changes to the composition of the microbiota and their fermentation products. Acarbose also increases longevity in mice, an effect that has been correlated with increased production of the short-chain fatty acids propionate and butyrate. In experiments replicated across three study sites, two distantly related species in the bacterial family Muribaculaceae were dramatically more abundant in acarbose-treated mice, distinguishing these responders from other members of the family. Bacteria in the family Muribaculaceae are predicted to produce propionate as a fermentation end product and are abundant and diverse in the guts of mice, although few isolates are available. We reconstructed genomes from metagenomes (MAGs) for nine populations of Muribaculaceae to examine factors that distinguish species that respond positively to acarbose. We found two closely related MAGs (B1A and B1B) from one responsive species that both contain a polysaccharide utilization locus with a predicted extracellular α-amylase. These genomes also shared a periplasmic neopullulanase with another, distantly related MAG (B2) representative of the only other responsive species. This gene differentiated these three MAGs from MAGs representative of nonresponding species. Differential gene content in B1A and B1B may be associated with the inconsistent response of this species to acarbose across study sites. This work demonstrates the utility of culture-free genomics for inferring the ecological roles of gut bacteria, including their response to pharmaceutical perturbations. IMPORTANCE The drug acarbose is used to treat diabetes by preventing the breakdown of starch in the small intestine, resulting in dramatic changes in the abundance of some members of the gut microbiome and its fermentation products. In mice, several of the bacteria that respond most positively are classified in the family Muribaculaceae, members of which produce propionate as a primary fermentation product. Propionate has been associated with gut health and increased longevity in mice. We found that genomes of the most responsive Muribaculaceae showed signs of specialization for starch fermentation, presumably providing them a competitive advantage in the large intestine of animals consuming acarbose. Comparisons among genomes enhance existing models for the ecological niches occupied by members of this family. In addition, genes encoding one type of enzyme known to participate in starch breakdown were found in all three genomes from responding species but none of the other genomes.
Collapse
|
10
|
Habitual Dietary Intake Affects the Altered Pattern of Gut Microbiome by Acarbose in Patients with Type 2 Diabetes. Nutrients 2021; 13:nu13062107. [PMID: 34205413 PMCID: PMC8235473 DOI: 10.3390/nu13062107] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
The aim of this research was to reveal the characteristics of gut microbiome altered by acarbose intervention in Japanese patients with type 2 diabetes (T2D) and its possible association with habitual dietary intake. Eighteen patients with T2D were administered acarbose for four weeks. The abundances of two major phyla, namely Actinobacteria and Bacteroidetes, were reciprocally changed accompanied by the acarbose intervention. There were also significant changes in the abundances of ten genera, including the greater abundance of Bifidobacterium, Eubacterium, and Lactobacillus and the lower abundance of Bacteroides in the group after the intervention than that before the intervention. Hierarchical clustering of habitual dietary intake was performed based on the pattern of changes in the gut microbiota and were classified into distinct three clusters. Cluster I consisted of sucrose, cluster II mainly included fat intake, and cluster III mainly included carbohydrate intake. Moreover, the amount of change in Faecalibacterium was positively correlated with the intake of rice, but negatively correlated with the intake of bread. The intake of potato was negatively correlated with the amount of change in Akkermansia and Subdoligranulum. Acarbose altered the composition of gut microbiome in Japanese patients with T2D, which might be linked to the habitual dietary intake.
Collapse
|
11
|
Smith DL, Orlandella RM, Allison DB, Norian LA. Diabetes medications as potential calorie restriction mimetics-a focus on the alpha-glucosidase inhibitor acarbose. GeroScience 2021; 43:1123-1133. [PMID: 33006707 PMCID: PMC8190416 DOI: 10.1007/s11357-020-00278-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
The field of aging research has grown rapidly over the last half-century, with advancement of scientific technologies to interrogate mechanisms underlying the benefit of life-extending interventions like calorie restriction (CR). Coincident with this increase in knowledge has been the rise of obesity and type 2 diabetes (T2D), both associated with increased morbidity and mortality. Given the difficulty in practicing long-term CR, a search for compounds (CR mimetics) which could recapitulate the health and longevity benefits without requiring food intake reductions was proposed. Alpha-glucosidase inhibitors (AGIs) are compounds that function predominantly within the gastrointestinal tract to inhibit α-glucosidase and α-amylase enzymatic digestion of complex carbohydrates, delaying and decreasing monosaccharide uptake from the gut in the treatment of T2D. Acarbose, an AGI, has been shown in pre-clinical models to increase lifespan (greater longevity benefits in males), with decreased body weight gain independent of calorie intake reduction. The CR mimetic benefits of acarbose are further supported by clinical findings beyond T2D including the risk for other age-related diseases (e.g., cancer, cardiovascular). Open questions remain regarding the exclusivity of acarbose relative to other AGIs, potential off-target effects, and combination with other therapies for healthy aging and longevity extension. Given the promising results in pre-clinical models (even in the absence of T2D), a unique mechanism of action and multiple age-related reduced disease risks that have been reported with acarbose, support for clinical trials with acarbose focusing on aging-related outcomes and incorporating biological sex, age at treatment initiation, and T2D-dependence within the design is warranted.
Collapse
Affiliation(s)
- Daniel L Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1720 2nd Avenue S, Webb 423, Birmingham, AL, 35294-3360, USA.
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, USA.
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA.
- Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Rachael M Orlandella
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David B Allison
- School of Public Health, Indiana University - Bloomington, Bloomington, IN, USA
| | - Lyse A Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1720 2nd Avenue S, Webb 423, Birmingham, AL, 35294-3360, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
12
|
Li C, Kuang J, Zhao Y, Sun H, Guan H. Effect of type 2 diabetes and antihyperglycemic drug therapy on signs of tumor invasion in papillary thyroid cancer. Endocrine 2020; 69:92-99. [PMID: 32291737 DOI: 10.1007/s12020-020-02291-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/26/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE This retrospective study investigated the association between worrisome pathological features of papillary thyroid cancer indicative of invasion and type 2 diabetes, as well as antihyperglycemic drug therapy for diabetes. METHODS The records of 14,167 patients who had undergone primary surgery for thyroid cancer were retrospectively reviewed and screened for concomitant diabetes. The diabetic and nondiabetic groups were age and gender matched, and further stratified by treatment, including five single antihyperglycemic drugs. RESULTS The study population comprised 942 patients, including 471 patients each with and without diabetes. The rate of worrisome pathological features was higher in diabetic patients than in nondiabetic patients (49.26% cf. 30.57%, P < 0.001), mainly reflected by extrathyroidal extension (34.82% cf. 5.94%, P < 0.001) and lymph node metastasis (42.68% cf. 33.55%, P < 0.001). The number of lymph node metastases in those treated with acarbose (0.32) was significantly lower compared with groups given any of the other four antihyperglycemic drugs (ranging from 1.51 to 2.15; P = 0.001 to 0.05). CONCLUSIONS Papillary thyroid cancer complicated with type 2 diabetes has a higher risk of invasive tumor growth. Compared with other antihyperglycemic drugs, patients with acarbose had the lowest risk of aggressive tumor growth. These results may evoke pathophysiological hypotheses to be explored in preclinical and clinical studies.
Collapse
Affiliation(s)
- Changlin Li
- Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Laboratory of Surgical Translational Medicine, Jilin Provincial Engineering Laboratory of Thyroid Disease Prevention and Control, Changchun, Jilin Province, PR China
| | - Jian Kuang
- Department of Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, PR China
| | - Yishen Zhao
- Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Laboratory of Surgical Translational Medicine, Jilin Provincial Engineering Laboratory of Thyroid Disease Prevention and Control, Changchun, Jilin Province, PR China
| | - Hui Sun
- Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Laboratory of Surgical Translational Medicine, Jilin Provincial Engineering Laboratory of Thyroid Disease Prevention and Control, Changchun, Jilin Province, PR China.
| | - Haixia Guan
- Department of Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, PR China.
- Department of Endocrinology and Metabolism, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, 110001, Liaoning, PR China.
| |
Collapse
|
13
|
Cerqueira FM, Photenhauer AL, Pollet RM, Brown HA, Koropatkin NM. Starch Digestion by Gut Bacteria: Crowdsourcing for Carbs. Trends Microbiol 2020; 28:95-108. [DOI: 10.1016/j.tim.2019.09.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/29/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
|
14
|
Hamada Y, Goto M, Nishimura G, Nagasaki H, Seino Y, Kamiya H, Nakamura J. The alpha-glucosidase inhibitor miglitol increases hepatic CYP7A1 activity in association with altered short-chain fatty acid production in the gut of obese diabetic mice. Metabol Open 2020; 5:100024. [PMID: 32812937 PMCID: PMC7424806 DOI: 10.1016/j.metop.2020.100024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/29/2019] [Accepted: 01/08/2020] [Indexed: 01/07/2023] Open
Abstract
Purpose Bile acids (BAs) have been shown to contribute to glucose and energy homeostasis. We have recently reported that miglitol, an alpha-glucosidase inhibitor, increases fecal BA excretion and ameliorate insulin resistance and obesity in mice. The aim of this study was to clarify the mechanisms by which miglitol affects BA metabolism. The expression of genes regulating BA metabolism, gut microbiome and short-chain fatty acids (SCFA) were examined. Procedures NSY mice, representing an obese type 2 diabetic model, were fed with a high-fat diet with or without miglitol for 4 weeks. The expression of BA-related genes in the liver and the lower intestine were measured. Alterations in fecal microbiome, fecal SCFA along with plasma lipid levels were also evaluated. Major findings Miglitol significantly increased fecal BA secretion and markedly upregulated the mRNA expression, protein levels and enzyme activity of hepatic cholesterol 7α-hydroxylase, a rate-limiting enzyme of BA synthesis. In the intestine, miglitol treatment significantly suppressed the mRNA expression of apical sodium-dependent bile acid transporter and ATP-binding cassette transporter G5 and G8. In fecal microbiome, the prevalence of prevotella was remarkably reduced and that of clostridium subcluster XIVa was increased by miglitol. Miglitol elevated formic and n-butyric acids along with total SCFA concentration in feces, while succinic acid was decreased. There was no change in plasma total cholesterol levels. Conclusions Collectively, miglitol may affect BA metabolism via enhanced CYP7A1 activity resulting from at least in part the alterations in gut microbiome and SCFA production in obese diabetic mice.
Collapse
Affiliation(s)
- Yoji Hamada
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, 480-1195, Japan
| | - Moritaka Goto
- Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho Co, Ltd, 363 Shiozaki, Hokusei-cho, Inabe, 511-0406, Japan
| | - Go Nishimura
- Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho Co, Ltd, 363 Shiozaki, Hokusei-cho, Inabe, 511-0406, Japan
| | - Hiroshi Nagasaki
- Department of Physiology I, Fujita University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yusuke Seino
- Department of Endocrinology and Diabetes, Fujita University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hideki Kamiya
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, 480-1195, Japan
| | - Jiro Nakamura
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, 480-1195, Japan
| |
Collapse
|
15
|
Smith BJ, Miller RA, Ericsson AC, Harrison DC, Strong R, Schmidt TM. Changes in the gut microbiome and fermentation products concurrent with enhanced longevity in acarbose-treated mice. BMC Microbiol 2019; 19:130. [PMID: 31195972 PMCID: PMC6567620 DOI: 10.1186/s12866-019-1494-7] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/17/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Treatment with the α-glucosidase inhibitor acarbose increases median lifespan by approximately 20% in male mice and 5% in females. This longevity extension differs from dietary restriction based on a number of features, including the relatively small effects on weight and the sex-specificity of the lifespan effect. By inhibiting host digestion, acarbose increases the flux of starch to the lower digestive system, resulting in changes to the gut microbiota and their fermentation products. Given the documented health benefits of short-chain fatty acids (SCFAs), the dominant products of starch fermentation by gut bacteria, this secondary effect of acarbose could contribute to increased longevity in mice. To explore this hypothesis, we compared the fecal microbiome of mice treated with acarbose to control mice at three independent study sites. RESULTS Microbial communities and the concentrations of SCFAs in the feces of mice treated with acarbose were notably different from those of control mice. At all three study sites, the bloom of a single bacterial taxon was the most obvious response to acarbose treatment. The blooming populations were classified to the largely uncultured Bacteroidales family Muribaculaceae and were the same taxonomic unit at two of the three sites. Propionate concentrations in feces were consistently elevated in treated mice, while the concentrations of acetate and butyrate reflected a dependence on study site. Across all samples, Muribaculaceae abundance was strongly correlated with propionate and community composition was an important predictor of SCFA concentrations. Cox proportional hazards regression showed that the fecal concentrations of acetate, butyrate, and propionate were, together, predictive of mouse longevity even while controlling for sex, site, and acarbose. CONCLUSION We observed a correlation between fecal SCFAs and lifespan in mice, suggesting a role of the gut microbiota in the longevity-enhancing properties of acarbose. Treatment modulated the taxonomic composition and fermentation products of the gut microbiome, while the site-dependence of the responses illustrate the challenges facing reproducibility and interpretation in microbiome studies. These results motivate future studies exploring manipulation of the gut microbial community and its fermentation products for increased longevity, testing causal roles of SCFAs in the observed effects of acarbose.
Collapse
Affiliation(s)
- Byron J Smith
- Department of Ecology & Evolutionary Biology, University of Michigan, Ann Arbor, 48109 MI USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, 48109 MI USA
| | - Aaron C Ericsson
- University of Missouri Metagenomics Center, University of Missouri, Columbia, 65201 MO USA
| | | | - Randy Strong
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, 78229 TX USA
- Barshop Institute for Longevity and Aging Studies, San Antonio, 78245 TX USA
- Geriatric Research, Education and Clinical Center and Research Service, South Texas Veterans Health Care System, San Antonio, 78229 TX USA
| | - Thomas M Schmidt
- Department of Ecology & Evolutionary Biology, University of Michigan, Ann Arbor, 48109 MI USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, 48109 MI USA
| |
Collapse
|
16
|
The Glucoamylase Inhibitor Acarbose Has a Diet-Dependent and Reversible Effect on the Murine Gut Microbiome. mSphere 2019; 4:4/1/e00528-18. [PMID: 30728281 PMCID: PMC6365613 DOI: 10.1128/msphere.00528-18] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The gut microbial community has a profound influence on host physiology in both health and disease. In diabetic individuals, the gut microbiota can affect the course of disease, and some medications for diabetes, including metformin, seem to elicit some of their benefits via an interaction with the microbiota. Here, we report that acarbose, a glucoamylase inhibitor for type 2 diabetes, changes the murine gut bacterial community structure in a reversible and diet-dependent manner. In both high-starch and high-fiber diet backgrounds, acarbose treatment results in increased short-chain fatty acids, particularly butyrate, as measured in stool samples. As we learn more about how human disease is affected by the intestinal bacterial community, the interplay between medications such as acarbose and the diet will become increasingly important to evaluate. Acarbose is a safe and effective medication for type 2 diabetes that inhibits host glucoamylases to prevent starch digestion in the small intestines and thus decrease postprandial blood glucose levels. This results in an increase in dietary starch in the distal intestine, where it becomes food for the gut bacterial community. Here, we examined the effect of acarbose therapy on the gut community structure in mice fed either a high-starch (HS) or high-fiber diet rich in plant polysaccharides (PP). The fecal microbiota of animals consuming a low dose of acarbose (25 ppm) was not significantly different from that of control animals that did not receive acarbose. However, a high dose of acarbose (400 ppm) with the HS diet resulted in a substantial change to the microbiota structure. Most notably, the HS diet with a high dose of acarbose lead to an expansion of the Bacteroidaceae and Bifidobacteriaceae and a decrease in the Verrucomicrobiaceae (such as Akkermansia muciniphila) and the Bacteroidales S24-7. Once acarbose treatment ceased, the community composition quickly reverted to mirror that of the control group, suggesting that acarbose does not irreversibly alter the gut community. The high dose of acarbose in the PP diet resulted in a distinct community structure with increased representation of Bifidobacteriaceae and Lachnospiraceae. Short-chain fatty acids (SCFAs) measured from stool samples were increased, especially butyrate, as a result of acarbose treatment in both diets. These data demonstrate the potential of acarbose to change the gut community structure and increase beneficial SCFA output in a diet-dependent manner. IMPORTANCE The gut microbial community has a profound influence on host physiology in both health and disease. In diabetic individuals, the gut microbiota can affect the course of disease, and some medications for diabetes, including metformin, seem to elicit some of their benefits via an interaction with the microbiota. Here, we report that acarbose, a glucoamylase inhibitor for type 2 diabetes, changes the murine gut bacterial community structure in a reversible and diet-dependent manner. In both high-starch and high-fiber diet backgrounds, acarbose treatment results in increased short-chain fatty acids, particularly butyrate, as measured in stool samples. As we learn more about how human disease is affected by the intestinal bacterial community, the interplay between medications such as acarbose and the diet will become increasingly important to evaluate.
Collapse
|
17
|
Fu X, Liu Z, Zhu C, Mou H, Kong Q. Nondigestible carbohydrates, butyrate, and butyrate-producing bacteria. Crit Rev Food Sci Nutr 2018; 59:S130-S152. [PMID: 30580556 DOI: 10.1080/10408398.2018.1542587] [Citation(s) in RCA: 286] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nondigestible carbohydrates (NDCs) are fermentation substrates in the colon after escaping digestion in the upper gastrointestinal tract. Among NDCs, resistant starch is not hydrolyzed by pancreatic amylases but can be degraded by enzymes produced by large intestinal bacteria, including clostridia, bacteroides, and bifidobacteria. Nonstarch polysaccharides, such as pectin, guar gum, alginate, arabinoxylan, and inulin fructans, and nondigestible oligosaccharides and their derivatives, can also be fermented by beneficial bacteria in the large intestine. Butyrate is one of the most important metabolites produced through gastrointestinal microbial fermentation and functions as a major energy source for colonocytes by directly affecting the growth and differentiation of colonocytes. Moreover, butyrate has various physiological effects, including enhancement of intestinal barrier function and mucosal immunity. In this review, several representative NDCs are introduced, and their chemical components, structures, and physiological functions, including promotion of the proliferation of butyrate-producing bacteria and enhancement of butyrate production, are discussed. We also describe the strategies for achieving directional accumulation of colonic butyrate based on endogenous generation mechanisms.
Collapse
Affiliation(s)
- Xiaodan Fu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Zhemin Liu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Changliang Zhu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Haijin Mou
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Qing Kong
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| |
Collapse
|
18
|
Xu GD, Cai L, Ni YS, Tian SY, Lu YQ, Wang LN, Chen LL, Ma WY, Deng SP. Comparisons of Effects on Intestinal Short-Chain Fatty Acid Concentration after Exposure of Two Glycosidase Inhibitors in Mice. Biol Pharm Bull 2018; 41:1024-1033. [PMID: 29962399 DOI: 10.1248/bpb.b17-00978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acarbose and voglibose are the most widely used diabetes drugs as glycosidase inhibitors. In this study, the use of these two inhibitors significantly increased the content of starch in large intestine, and altered the concentration of short-chain fatty acids (SCFAs) by affecting the intestinal microbiota. However, there are some differences in the intestinal microbiome of the two groups of mice, mainly in bacteria such as Bacteroidaceae bacteroides and Desulfovibrionaceae desulfovibrio. The productions of acetate and propionate in caecum in voglibose group were significantly higher than those in acarbose group and two kinds of glycosidase inhibitors were close in the production of butyrate in caecum. The Tax4Fun analysis based on Kyoto Encyclopedia of Genes and Genomes (KEGG) data indicated that different productions of acetate and propionate between acarbose group and voglibose group may be related to 2-oxoisovalerate dehydrogenase and pyruvate oxidase. In addition, in-vitro experiments suggested that voglibose had less effect on epithelial cells than acarbose after direct stimulation. According to the recent researches of SCFAs produced by intestinal microbiota, our comparative study shown higher concentration of these beneficial fatty acids in the lumen of voglibose-treated mice, which implied a lower level of inflammation.
Collapse
Affiliation(s)
- Guo-Dong Xu
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Lei Cai
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Yi-Shu Ni
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Shi-Yi Tian
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Ying-Qi Lu
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Li-Na Wang
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Lian-Lian Chen
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Wen-Ya Ma
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| | - Shao-Ping Deng
- Food Sensory Science Laboratory, School of Food Science and Biotechnology, Zhejiang Gongshang University
| |
Collapse
|
19
|
Alpha-glucosidase inhibitors and risk of cancer in patients with diabetes mellitus: a systematic review and meta-analysis. Oncotarget 2017; 8:81027-81039. [PMID: 29113364 PMCID: PMC5655259 DOI: 10.18632/oncotarget.17515] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/17/2017] [Indexed: 01/25/2023] Open
Abstract
Several studies have shown that anti-diabetic medications may modify the risk of cancer. We performed a systematic review and meta-analysis to evaluate the effect of alpha-glucosidase inhibitors (AGIs) on the risk of cancer in patients with diabetes mellitus. We conducted a systematic search of Medline, EMBASE, and Web of Science databases, up to September 30, 2016. Random-effects model was used to estimate the summary odds ratios (ORs) with 95% CI. Twenty-five studies (14 cohort, 7 case-control, and 4 randomized controlled trials) involving 1,285,433 patients with diabetes were included. Meta-analysis of observational studies showed that the use of AGIs was associated with a lower risk of developing cancer (OR = 0.86, 95% CI 0.78-0.96), especially gastrointestinal cancer (OR = 0.83, 95% CI 0.71-0.97). There was considerable heterogeneity across the studies introduced partly by the quality of included studies and adjustment for potential confounders. Meta-analysis of randomized controlled trials did not reveal any significant association between AGIs and cancer risk. Meta-analysis of observational studies indicated that AGIs may decrease the risk of cancer in individuals with diabetes.
Collapse
|
20
|
Zhang X, Fang Z, Zhang C, Xia H, Jie Z, Han X, Chen Y, Ji L. Effects of Acarbose on the Gut Microbiota of Prediabetic Patients: A Randomized, Double-blind, Controlled Crossover Trial. Diabetes Ther 2017; 8:293-307. [PMID: 28130771 PMCID: PMC5380489 DOI: 10.1007/s13300-017-0226-y] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The α-glucosidase inhibitor acarbose is an efficacious medicine for the treatment and prevention of type 2 diabetes mellitus (T2DM). However, the response of gut microbiota to acarbose is important, as the microbiota may have a critical role in the development of metabolic diseases, and acarbose is metabolized exclusively within the gastrointestinal tract. We explored the changes in the proportion and diversity of gut microbiota before and after treatment with acarbose in patients with prediabetes. METHODS We designed a randomized, double-blind, controlled crossover trial in which 52 Chinese patients with prediabetes by an oral glucose tolerance test (OGTT) with a BMI of 18-35 kg/m2 were randomly allocated to treatment with acarbose or placebo. Gut microbiota characterizations were determined with 16S rDNA-based high-throughput sequencing. RESULTS Of the 52 participants who entered the study, 40 (76.9%) completed the protocol. On the basis of the operational taxonomic unit (OTU) profiles, a total of 107 OTUs were significantly altered after acarbose treatment, with 76 (71%) assigned to the order of Clostridiales. Ruminococcaceae (15 OTUs) and Lachnospiraceae (22 OTUs) decreased in response to acarbose, and 48 OTUs increased by 12.8-fold, including Lactobacillaceae (8 of 9 belonging to Lactobacillus), Ruminococcaceae (6 of 11 belonging to Faecalibacterium), and Veillonellaceae (8 of 15 belonging to Dialister). At genera level, five flourished after treatment with acarbose, including Lactobacillus and Dialister, while Butyricicoccus, Phascolarctobacterium, and Ruminococcus were inhibited. CONCLUSION This study suggests that the benefits of acarbose for T2DM may correlate with the selective modulation of the gut microbiota. TRIAL REGISTRATION Chinese Clinical Trial Register number, ChiCTR-TTRCC-13004112.
Collapse
Affiliation(s)
- Xiuying Zhang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Centre, Beijing, 100044, China
| | - Zhiwei Fang
- Institute for Systems Biology, Jianghan University, Wuhan, 430056, China
| | - Chunfang Zhang
- Department of Clinical Epidemiology, Peking University People's Hospital, Beijing, 100044, China
| | | | - Zhuye Jie
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Centre, Beijing, 100044, China
| | - Yingli Chen
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Centre, Beijing, 100044, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Centre, Beijing, 100044, China.
| |
Collapse
|
21
|
Brewer RA, Gibbs VK, Smith DL. Targeting glucose metabolism for healthy aging. NUTRITION AND HEALTHY AGING 2016; 4:31-46. [PMID: 28035340 PMCID: PMC5166514 DOI: 10.3233/nha-160007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Advancing age is the greatest single risk factor for numerous chronic diseases. Thus, the ability to target the aging process can facilitate improved healthspan and potentially lifespan. Lack of adequate glucoregulatory control remains a recurrent theme accompanying aging and chronic disease, while numerous longevity interventions result in maintenance of glucoregulatory control. In this review, we propose targeting glucose metabolism to enhance regulatory control as a means to ameliorate the aging process. We highlight that calorie restriction improves glucoregulatory control and extends both lifespan and healthspan in model organisms, but we also indicate more practical interventions (i.e., calorie restriction mimetics) are desirable for clinical application in humans. Of the calorie restriction mimetics being investigated, we focus on the type 2 diabetes drug acarbose, an α-glucosidase inhibitor that when taken with a meal, results in reduced enzymatic degradation and absorption of glucose from complex carbohydrates. We discuss alternatives to acarbose that yield similar physiologic effects and describe dietary sources (e.g., sweet potatoes, legumes, and berries) of bioactive compounds with α-glucosidase inhibitory activity. We indicate future research should include exploration of how non-caloric compounds like α-glucosidase inhibitors modify macronutrient metabolism prior to disease onset, which may guide nutritional/lifestyle interventions to support health and reduce age-related disease risk.
Collapse
Affiliation(s)
- Rachel A. Brewer
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victoria K. Gibbs
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, USA
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, USA
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
22
|
Lehmann A, Hornby PJ. Intestinal SGLT1 in metabolic health and disease. Am J Physiol Gastrointest Liver Physiol 2016; 310:G887-98. [PMID: 27012770 DOI: 10.1152/ajpgi.00068.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/21/2016] [Indexed: 01/31/2023]
Abstract
The Na(+)-glucose cotransporter 1 (SGLT1/SLC5A1) is predominantly expressed in the small intestine. It transports glucose and galactose across the apical membrane in a process driven by a Na(+) gradient created by Na(+)-K(+)-ATPase. SGLT2 is the major form found in the kidney, and SGLT2-selective inhibitors are a new class of treatment for type 2 diabetes mellitus (T2DM). Recent data from patients treated with dual SGLT1/2 inhibitors or SGLT2-selective drugs such as canagliflozin (SGLT1 IC50 = 663 nM) warrant evaluation of SGLT1 inhibition for T2DM. SGLT1 activity is highly dynamic, with modulation by multiple mechanisms to ensure maximal uptake of carbohydrates (CHOs). Intestinal SGLT1 inhibition lowers and delays the glucose excursion following CHO ingestion and augments glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) secretion. The latter is likely due to increased glucose exposure of the colonic microbiota and formation of metabolites such as L cell secretagogues. GLP-1 and PYY secretion suppresses food intake, enhances the ileal brake, and has an incretin effect. An increase in colonic microbial production of propionate could contribute to intestinal gluconeogenesis and mediate positive metabolic effects. On the other hand, a threshold of SGLT1 inhibition that could lead to gastrointestinal intolerability is unclear. Altered Na(+) homeostasis and increased colonic CHO may result in diarrhea and adverse gastrointestinal effects. This review considers the potential mechanisms contributing to positive metabolic and negative intestinal effects. Compounds that inhibit SGLT1 must balance the modulation of these mechanisms to achieve therapeutic efficacy for metabolic diseases.
Collapse
Affiliation(s)
- Anders Lehmann
- Division of Endocrinology, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; and
| | - Pamela J Hornby
- Cardiovascular and Metabolic Disease, Janssen Research and Development, LLC, Spring House, Pennsylvania
| |
Collapse
|
23
|
Schnell O, Weng J, Sheu WHH, Watada H, Kalra S, Soegondo S, Yamamoto N, Rathod R, Zhang C, Grzeszczak W. Acarbose reduces body weight irrespective of glycemic control in patients with diabetes: results of a worldwide, non-interventional, observational study data pool. J Diabetes Complications 2016; 30:628-37. [PMID: 26935335 DOI: 10.1016/j.jdiacomp.2016.01.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/27/2016] [Accepted: 01/28/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The objective of this study is to examine the effect of acarbose, an alpha-glucosidase inhibitor, on body weight in a real-life setting by pooling data from post-marketing surveillance. METHODS Data from 10 studies were pooled (n=67,682) and the effect of acarbose on body weight was analysed taking into account baseline body weight, glycemic parameters and other baseline characteristics. RESULTS The mean relative reduction in body weight was 1.45 ± 3.24% at the 3-month visit (n=43,510; mean baseline 73.4 kg) and 1.40 ± 3.28% at the last visit (n=54,760; mean baseline 73.6 kg) (both p<0.0001). These reductions were dependent on baseline body weight (overweight: -1.33 ± 2.98% [n=13,498; mean baseline 71.6 kg]; obese: -1.98 ± 3.40% [n=20,216; mean baseline 81.3 kg]). When analysed by baseline glycemic parameter quartiles, the reduction was independent of fasting plasma glucose (FPG), postprandial plasma glucose (PPG), glycated hemoglobin (HbA1c) and postprandial glucose excursion (PPGE). A bivariate analysis of covariance identified female sex, South East Asian and East Asian ethnicity, younger age, higher body mass index, short duration of diabetes, and no previous treatment as factors likely to impact positively on body weight reduction with acarbose. CONCLUSIONS This post-hoc analysis showed that acarbose treatment reduces body weight independent of glycemic control status but dependent on baseline body weight.
Collapse
Affiliation(s)
- Oliver Schnell
- Forschergruppe Diabetes e.V. at the Helmholtz Center Munich, Munich, Germany.
| | - Jianping Weng
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou; Department of Endocrinology and Metabolic Disease, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.
| | - Hirotaka Watada
- Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Sanjay Kalra
- Bharti Research Institute of Diabetes & Endocrinology, Karnal, India.
| | | | | | | | | | | |
Collapse
|
24
|
Smallbone K, M Corfe B. A mathematical model of the colon crypt capturing compositional dynamic interactions between cell types. Int J Exp Pathol 2014; 95:1-7. [PMID: 24354351 PMCID: PMC3919643 DOI: 10.1111/iep.12062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 09/24/2013] [Indexed: 12/27/2022] Open
Abstract
Models of the development and early progression of colorectal cancer are based upon understanding the cycle of stem cell turnover, proliferation, differentiation and death. Existing crypt compartmental models feature a linear pathway of cell types, with little regulatory mechanism. Previous work has shown that there are perturbations in the enteroendocrine cell population of macroscopically normal crypts, a compartment not included in existing models. We show that existing models do not adequately recapitulate the dynamics of cell fate pathways in the crypt. We report the progressive development, iterative testing and fitting of a developed compartmental model with additional cell types, and which includes feedback mechanisms and cross-regulatory mechanisms between cell types. The fitting of the model to existing data sets suggests a need to invoke cross-talk between cell types as a feature of colon crypt cycle models.
Collapse
Affiliation(s)
- Kieran Smallbone
- Manchester Centre for Integrative Systems Biology, University of Manchester, Manchester, UK
| | | |
Collapse
|
25
|
Yang W, Liu J, Shan Z, Tian H, Zhou Z, Ji Q, Weng J, Jia W, Lu J, Liu J, Xu Y, Yang Z, Chen W. Acarbose compared with metformin as initial therapy in patients with newly diagnosed type 2 diabetes: an open-label, non-inferiority randomised trial. Lancet Diabetes Endocrinol 2014; 2:46-55. [PMID: 24622668 DOI: 10.1016/s2213-8587(13)70021-4] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Metformin is the only first-line oral hypoglycaemic drug for type 2 diabetes recommended by international guidelines with proven efficacy, safety, and cost-effectiveness. However, little information exists about its use in Asian populations. We aimed to ascertain the effectiveness of the α-glucosidase inhibitor acarbose, extensively adopted in China, compared with metformin as the alternative initial therapy for newly diagnosed type 2 diabetes. METHODS In this 48-week, randomised, open-label, non-inferiority trial, patients who were newly diagnosed with type 2 diabetes, with a mean HbA1c of 7·5%, were enrolled from 11 sites in China. After a 4-week lifestyle modification run-in, patients were assigned to 24 weeks of monotherapy with metformin or acarbose as the initial treatment, followed by a 24-week therapy phase during which add-on therapy was used if prespecified glucose targets were not achieved. Primary endpoints were to establish whether acarbose was non-inferior to metformin in HbA1c reduction at week 24 and week 48 timepoints. The non-inferiority margin was 0·3%, with an expected null difference in the change from baseline to week 48 in HbA1c. Analysis was done on a modified intention-to-treat population. This study was registered with Chinese Clinical Trial Registry, number ChiCTR-TRC-08000231. FINDINGS Of the 788 patients randomly assigned to treatment groups, 784 patients started the intended study drug. HbA1c reduction at week 24 was -1·17% in the acarbose group and -1·19% in the metformin group. At week 48, the HbA1c reduction was -1·11% (acarbose) and -1·12% (metformin) with difference 0·01% (95% CI -0·12 to 0·14, p=0·8999). Six (2%) patients in the acarbose group and seven (2%) patients in the metformin group had serious adverse events, and two (1%) and four (1%) had hypoglycaemic episodes. INTERPRETATION This study provides evidence that acarbose is similar to metformin in efficacy, and is therefore a viable choice for initial therapy in Chinese patients newly diagnosed with type 2 diabetes. FUNDING Bayer Healthcare (China) and Double Crane Phama.
Collapse
Affiliation(s)
- Wenying Yang
- China-Japan Friendship Hospital, Beijing, China.
| | - Jie Liu
- Shanxi Province People's Hospital, Taiyuan, China
| | - Zhongyan Shan
- The First Hospital of China Medical University, Shenyang, China
| | - Haoming Tian
- West China Hospital, Sichuan University, Chengdu, China
| | - Zhiguang Zhou
- Xiangya Second Hospital of Central South University, Changsha, China
| | - Qiuhe Ji
- Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianping Weng
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weiping Jia
- Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juming Lu
- Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jing Liu
- Gansu Provincial Hospital, Lanzhou, China
| | - Yuan Xu
- Beijing Chao Yang Hospital, Beijing, China
| | | | - Wei Chen
- Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
26
|
den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013; 54:2325-40. [PMID: 23821742 DOI: 10.1194/jlr.r036012] [Citation(s) in RCA: 3026] [Impact Index Per Article: 252.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Short-chain fatty acids (SCFAs), the end products of fermentation of dietary fibers by the anaerobic intestinal microbiota, have been shown to exert multiple beneficial effects on mammalian energy metabolism. The mechanisms underlying these effects are the subject of intensive research and encompass the complex interplay between diet, gut microbiota, and host energy metabolism. This review summarizes the role of SCFAs in host energy metabolism, starting from the production by the gut microbiota to the uptake by the host and ending with the effects on host metabolism. There are interesting leads on the underlying molecular mechanisms, but there are also many apparently contradictory results. A coherent understanding of the multilevel network in which SCFAs exert their effects is hampered by the lack of quantitative data on actual fluxes of SCFAs and metabolic processes regulated by SCFAs. In this review we address questions that, when answered, will bring us a great step forward in elucidating the role of SCFAs in mammalian energy metabolism.
Collapse
Affiliation(s)
- Gijs den Besten
- Center for Liver, Digestive, and Metabolic Diseases, Department of Pediatrics and University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
27
|
Lee HJ, Lee HS, Choi JW, Ra KS, Kim JM, Suh HJ. Novel tripeptides with α-glucosidase inhibitory activity isolated from silk cocoon hydrolysate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:11522-11525. [PMID: 21895007 DOI: 10.1021/jf202686m] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Active compounds with antidiabetic potential were isolated from silk peptide E5K6 by consecutive ultrafiltration and gel filtration using Biogel P-2 and RS-HPLC using a YMC-Pack Pro C18 column. The highest α-glucosidase inhibitory activity of silk peptide E5K6 resulted from fractions with MW <1 kDa. The activities of gel-filtered fractions from silk peptide E5K6 of <1 kDa were assayed in vitro, demonstrating that the fourth peak (F4) had the highest α-glucosidase inhibitory activity (IC(50) = 37.1 mg/mL). F4 of silk peptide E5K6 was separated by HPLC into two peaks. Moreover, the purified compounds were identified as Gly-Glu-Tyr (GEY, MW = 367 Da) and Gly-Tyr-Gly (GYG, MW = 295 Da) according to amino acid sequences, and their α-glucosidase inhibitory activities (IC(50)) were 2.7 and 1.5 mg/mL, respectively.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Department of Food and Nutrition, Korea University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
28
|
Worthley DL, Le Leu RK, Whitehall VL, Conlon M, Christophersen C, Belobrajdic D, Mallitt KA, Hu Y, Irahara N, Ogino S, Leggett BA, Young GP. A human, double-blind, placebo-controlled, crossover trial of prebiotic, probiotic, and synbiotic supplementation: effects on luminal, inflammatory, epigenetic, and epithelial biomarkers of colorectal cancer. Am J Clin Nutr 2009; 90:578-86. [PMID: 19640954 DOI: 10.3945/ajcn.2009.28106] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Diet is an important factor in colorectal carcinogenesis; thus, dietary supplements may have a role in colorectal cancer prevention. OBJECTIVE The objective was to establish the relative luminal, epithelial, and epigenetic consequences of prebiotic, probiotic, and synbiotic dietary supplementation in humans. DESIGN This was a randomized, double-blind, placebo-controlled, 4-wk crossover trial of resistant starch and Bifidobacterium lactis, either alone or as a combined synbiotic preparation, in 20 human volunteers. Rectal biopsy, feces, and serum samples were collected. The rectal mucosal endpoints were DNA methylation at 16 CpG island loci and LINE-1, epithelial proliferation (Ki67 immunohistochemistry), and crypt cellularity. The fecal endpoints were short-chain fatty acid concentrations, pH, ammonia, and microbiological profiles (by denaturing gradient gel electrophoresis and sequencing). Serum endpoints were a panel of cytokines and high-sensitivity C-reactive protein. RESULTS Seventeen subjects completed the entire study. The synbiotic intervention fostered a significantly different fecal stream bacterial community than did either the prebiotic (P = 0.032) or the probiotic (P = 0.001) intervention alone, in part because of a greater proportion of patients harboring fecal Lachnospiraceae spp. These changes developed in the absence of any significant differences in fecal chemistry. There were no differences in epithelial kinetics. CONCLUSIONS This synbiotic supplementation with B. lactis and resistant starch, in the doses used, induced unique changes in fecal microflora but did not significantly alter any other fecal, serum, or epithelial variables. This trial was registered in the Australian New Zealand Clinical Trials Registry at www.anzctr.org.au as ACTRN012606000115538.
Collapse
Affiliation(s)
- Daniel L Worthley
- Royal Brisbane and Women's Hospital Research Foundation, Clinical Research Centre, Brisbane, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Matsumura M, Monden T, Miyashita Y, Kawagoe Y, Shimizu H, Nakatani Y, Domeki N, Yanagi K, Ikeda S, Kasai K. Effects of changeover from voglibose to acarbose on postprandial triglycerides in type 2 diabetes mellitus patients. Adv Ther 2009; 26:660-6. [PMID: 19568704 DOI: 10.1007/s12325-009-0040-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Indexed: 10/20/2022]
Abstract
INTRODUCTION In this study, we examined the effects of the alpha-glucosidase inhibitors acarbose and voglibose on postprandial plasma glucose and serum triglyceride levels in patients with type 2 diabetes mellitus. METHODS Twenty-one Japanese patients with type 2 diabetes were enrolled in this study. Subjects had been treated with voglibose for at least 3 months. They underwent a 400 kcal balanced food meal tolerance test before and 8 weeks after the changeover from voglibose to acarbose. Subjects were divided into two groups: the first group (low-dose group; n=11) was changed over from 0.6 mg/day voglibose to 150 mg/day acarbose, and the other (high-dose group; n=10) from 0.9 mg/day voglibose to 300 mg/day acarbose. RESULTS The increment rate of postprandial plasma glucose ([plasma glucose 2 hours after test meal - fasting glucose]/fasting glucose) decreased from 34.7%+/-23.9% to 25.0%+/-24.6% (P=0.13) in the low-dose group, and decreased significantly from 56.1%+/-53.1% to 31.5%+/-36.0% (P=0.03) in the high-dose group after changeover. However, there were no significant changes in blood glycated hemoglobin (HbA(1c)) levels before and after changeover in either group. The increment rate of postprandial serum triglyceride (TG) ([serum TG 2 hours after test meal - fasting TG]/fasting TG) decreased significantly only in the high-dose group (52.4%+/-60.0% to 24.3%+/-16.6%) (P=0.05). No significant changes in serum high-density lipoprotein cholesterol levels were observed in either group, whereas serum low-density lipoprotein cholesterol levels decreased significantly from 3.20+/-0.25 to 2.65+/-0.18 mmol/L (P=0.04), only in the high-dose group. CONCLUSIONS In patients with type 2 diabetes our findings suggest that acarbose 300 mg/day is superior to voglibose 0.9 mg/day in improving postprandial hyperglycemia and hypertriglyceridemia.
Collapse
|
30
|
Kishino E, Ito T, Fujita K, Kiuchi Y. A mixture of Salacia reticulata (Kotala himbutu) aqueous extract and cyclodextrin reduces body weight gain, visceral fat accumulation, and total cholesterol and insulin increases in male Wistar fatty rats. Nutr Res 2009; 29:55-63. [DOI: 10.1016/j.nutres.2008.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2008] [Revised: 10/23/2008] [Accepted: 11/17/2008] [Indexed: 11/25/2022]
|
31
|
Sharma A, Yadav BS, Ritika. Resistant Starch: Physiological Roles and Food Applications. FOOD REVIEWS INTERNATIONAL 2008. [DOI: 10.1080/87559120801926237] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Alka Sharma
- a Department of Food Technology , Guru Jambheshwar University of Science & Technology , Haryana , India
| | - Baljeet Singh Yadav
- a Department of Food Technology , Guru Jambheshwar University of Science & Technology , Haryana , India
| | - Ritika
- a Department of Food Technology , Guru Jambheshwar University of Science & Technology , Haryana , India
| |
Collapse
|
32
|
Korenaga K, Korenaga M, Uchida K, Yamasaki T, Sakaida I. Effects of a late evening snack combined with alpha-glucosidase inhibitor on liver cirrhosis. Hepatol Res 2008; 38:1087-97. [PMID: 18647188 DOI: 10.1111/j.1872-034x.2008.00391.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM A late evening snack (LES) is recommended for protein-energy malnutrition in patients with liver cirrhosis. However, many cases of liver cirrhosis have accompanying impaired glucose tolerance and there are concerns that LESs might aggravate glucose intolerance. In this study, we concomitantly used an alpha-glucosidase inhibitor with a LES and examined the effects on glucose tolerance. In addition, we examined whether or not there was an improvement in energy metabolism by slowing glucose absorption with the concomitant use of the alpha-glucosidase inhibitor. METHODS The subjects were 11 patients with liver cirrhosis. From before the study, all the patients had been taking a LES supplementation with a branched-chain amino acid (BCAA)-enriched nutrient mixture. The patients were started on the concomitant use of alpha-glucosidase inhibitor (0.2 mg) taken just prior to the LES. The change of glucose tolerance and energy metabolism were examined using a 75-g oral glucose tolerance test and indirect calorimetry. RESULTS One week and three months after the start of the concomitant use of the alpha-glucosidase inhibitor, the area under the concentration curve for plasma glucose was significantly decreased. Three months after the concomitant use, the non-protein respiratory quotient was significantly improved. There were no serious side effects during the follow-ups. CONCLUSION The concomitant use of the alpha-glucosidase inhibitor use with LES showed the possibility of improving glucose tolerance and energy metabolism. In patients with impaired glucose tolerance, the concomitant use of an alpha-glucosidase inhibitor with LES might be a useful measure for nutritional management.
Collapse
Affiliation(s)
- Keiko Korenaga
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | | | | | | | | |
Collapse
|
33
|
Abstract
The present review assesses the potential of thePhaseolus vulgarisα-amylase inhibitor isoform 1 (α-AI1) starch blockers as a widely used remedy against obesity and diabetes. Consumption of the α-amylase inhibitor causes marginal intraluminal α-amylase activity facilitated by the inhibitor's appropriate structural, physico-chemical and functional properties. As a result there is decreased postprandial plasma hyperglycaemia and insulin levels, increased resistance of starch to digestion and increased activity of colorectal bacteria. The efficacy and safety of the amylase inhibitor extracts, however, depend on the processing and extraction techniques used. The extracts are potential ingredients in foods for increased carbohydrate tolerance in diabetics, decreased energy intake for reducing obesity and for increased resistant starch. Research developments in the distribution and biosynthesis of the α-amylase inhibitor, relevant physico-chemical properties, the molecular starch-blocking mechanism, anti-obesity and anti-diabetes effects, safety of extracts and the need for research into their potential anti-colorectal cancer effect are discussed.
Collapse
|
34
|
Wachters-Hagedoorn RE, Priebe MG, Heimweg JAJ, Heiner AM, Elzinga H, Stellaard F, Vonk RJ. Low-dose acarbose does not delay digestion of starch but reduces its bioavailability. Diabet Med 2007; 24:600-6. [PMID: 17381499 DOI: 10.1111/j.1464-5491.2007.02115.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS Slowly digestible starch is associated with beneficial health effects. The glucose-lowering drug acarbose has the potential to retard starch digestion since it inhibits alpha-amylase and alpha-glucosidases. We tested the hypothesis that a low dose of acarbose delays the rate of digestion of rapidly digestible starch without reducing its bioavailability and thereby increasing resistant starch flux into the colon. METHODS In a crossover study, seven healthy males ingested corn pasta (50.3 g dry weight), naturally enriched with (13)C, with and without 12.5 mg acarbose. Plasma glucose and insulin concentrations, and (13)CO(2) and hydrogen excretion in breath were monitored for 6 h after ingestion of the test meals. Using a primed continuous infusion of D-[6,6-(2)H(2)] glucose, the rate of appearance of starch-derived glucose was estimated, reflecting intestinal glucose absorption. RESULTS Areas under the 2-h postprandial curves of plasma glucose and insulin concentrations were significantly decreased by acarbose (-58.1 +/- 8.2% and -72.7 +/- 7.4%, respectively). Acarbose reduced the overall 6-h appearance of exogenous glucose (bioavailability) by 22 +/- 7% (mean +/-se) and the 6-h cumulative (13)CO(2) excretion by 30 +/- 6%. CONCLUSIONS These data show that in healthy volunteers a low dose of 12.5 mg acarbose decreases the appearance of starch-derived glucose substantially. Reduced bioavailability seems to contribute to this decrease to a greater extent than delay of digestion. This implies that the treatment effect of acarbose could in part be ascribed to the metabolic effects of colonic starch fermentation.
Collapse
Affiliation(s)
- R E Wachters-Hagedoorn
- Department of Paediatrics and Department of Medical Biomics, University of Groningen Medical Centre, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
35
|
Bang MH, Chio OS, Kim WK. Soyoligosaccharide increases fecal bifidobacteria counts, short-chain fatty acids, and fecal lipid concentrations in young Korean women. J Med Food 2007; 10:366-70. [PMID: 17651076 DOI: 10.1089/jmf.2005.096] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The effects of soyoligosaccharide intake on human fecal characteristics were investigated by measuring the fecal water content, bifidobacteria counts, pH, and concentrations of short-chain fatty acids (SCFAs) and lipids. Sixteen young women were randomly assigned to one of two groups: (1) 1.5 g/day soyoligosaccharide intake group [low soyoligosaccharide (LSO)] or (2) 3 g/day soyoligosaccharide intake group [high soyoligosaccharide (HSO)]. The experimental period was 30 days, and fecal samples were collected every 7 days. The number of bifidobacteria in feces was increased significantly in the HSO group. Of the fecal SCFAs, propionate and butyrate concentrations were significantly increased in the HSO group. The excretions of total lipids in feces were significantly increased in both the LSO and HSO groups. These results demonstrate that a soyoligosaccharide intake of 3 g/day increases fecal bifidobacteria counts, SCFA concentrations, and fecal lipid output in Korean young women.
Collapse
Affiliation(s)
- Myung Hee Bang
- Department of Food Science and Nutrition, Dankook University, Seoul, Republic of Korea
| | | | | |
Collapse
|
36
|
Abstract
The fermentation of starchin vitroproduces a higher proportion of butyrate than the fermentation of most other substrates. The α-glucosidase inhibitor acarbose increases the amount of starch entering the colon, and has been shown to increase faecal butyrate in humans. It is generally considered that colonic butyrate is quantitatively removed by the colonic mucosa and liver and does not appear in peripheral blood. However, studies in animals suggest that a small proportion of colonic butyrate reaches peripheral blood. Thus, we hypothesised that an increase in colonic butyrate production would result in a rise in serum butyrate in human subjects. To test this, subjects with impaired glucose tolerance were randomly treated in a double-blind fashion with placebo (n11) or acarbose (n11) (100 mg three times per day). Serum short-chain fatty acid concentrations were measured twelve times over 12 h with subjects eating a standard diet before randomization and after 4 months of therapy. At baseline, 12 h mean serum butyrate concentrations were similar in the placebo and acarbose groups (2·8 (SE 0·7) AND 3·3 (se 0·6) μm, respectively). After 4 months on placebo, mean serum butyrate (2·6 (se 0·5) μm) was no different from baseline. However, after 4 months on acarbose, serum butyrate had increased to 4·2 (se 1·0) μm, a value which differed significantly from both the baseline value in the acarbose group and the treatment value in the placebo group. We conclude that acarbose increased serum butyrate in subjects with impaired glucose tolerance. These results support the hypothesis that increased colonic butyrate production in human subjects can be detected by an increase in serum butyrate.
Collapse
|
37
|
Oku T, Yamada M, Nakamura M, Sadamori N, Nakamura S. Inhibitory effects of extractives from leaves of Morus alba on human and rat small intestinal disaccharidase activity. Br J Nutr 2007; 95:933-8. [PMID: 16611383 DOI: 10.1079/bjn20061746] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The inhibitory effect on human and rat intestinal disaccharidase by the extractive from the leaves ofMorus alba (ELM) containing 0·24% 1-deoxynojirimycin equivalent and its inhibitory activities were investigated by the modified Dahlqvist method. In the presence of 1000-fold diluted ELM solution, the sucrase activity of four human samples was inhibited by 96% and that of maltase and isomaltase by 95 and 99 %, respectively. The activities of trehalase and lactase were inhibited by 44 and 38 %, respectively. The human disaccharidase activities varied from sample to sample because the samples were obtained from different resected regions after surgery. However, the ratio of the inhibitory effect for sucrase, maltase, isomaltase, trehalase and lactase was very similar among the four samples, and also that of resembled rat intestinal disaccharides. The inhibitory constant of the 1-deoxynojirimycin equivalent for sucrase, maltase and isomaltase was 2·1× 10−4, 2·5 × 10−4 and 4·5 10−4μM, respectively, and these inhibitory activities were shown, using rat brush border membrane vesicles, to be competitive. These results demonstrate that digestion is inhibited when an appropriate amount of ELM is orally ingested with sucrose or polysaccharide in man. When ELM was orally administered in a sucrose solution to fasted rats, the elevation in blood glucose was significantly suppressed, depending on the concentration of ELM given. These results suggest that ELM could be used as an ingredient in health foods and in foods that help to prevent diabetes.
Collapse
Affiliation(s)
- Tsuneyuki Oku
- Graduate School of Human Health Science, Siebold University of Nagasaki, Japan.
| | | | | | | | | |
Collapse
|
38
|
Abstract
Interest has been recently rekindled in short chain fatty acids (SCFAs) with the emergence of prebiotics and probiotics aimed at improving colonic and systemic health. Dietary carbohydrates, specifically resistant starches and dietary fiber, are substrates for fermentation that produce SCFAs, primarily acetate, propionate, and butyrate, as end products. The rate and amount of SCFA production depends on the species and amounts of microflora present in the colon, the substrate source and gut transit time. SCFAs are readily absorbed. Butyrate is the major energy source for colonocytes. Propionate is largely taken up by the liver. Acetate enters the peripheral circulation to be metabolized by peripheral tissues. Specific SCFA may reduce the risk of developing gastrointestinal disorders, cancer, and cardiovascular disease. Acetate is the principal SCFA in the colon, and after absorption it has been shown to increase cholesterol synthesis. However, propionate, a gluconeogenerator, has been shown to inhibit cholesterol synthesis. Therefore, substrates that can decrease the acetate: propionate ratio may reduce serum lipids and possibly cardiovascular disease risk. Butyrate has been studied for its role in nourishing the colonic mucosa and in the prevention of cancer of the colon, by promoting cell differentiation, cell-cycle arrest and apoptosis of transformed colonocytes; inhibiting the enzyme histone deacetylase and decreasing the transformation of primary to secondary bile acids as a result of colonic acidification. Therefore, a greater increase in SCFA production and potentially a greater delivery of SCFA, specifically butyrate, to the distal colon may result in a protective effect. Butyrate irrigation (enema) has also been suggested in the treatment of colitis. More human studies are now needed, especially, given the diverse nature of carbohydrate substrates and the SCFA patterns resulting from their fermentation. Short-term and long-term human studies are particularly required on SCFAs in relation to markers of cancer risk. These studies will be key to the success of dietary recommendations to maximize colonic disease prevention.
Collapse
Affiliation(s)
- Julia M W Wong
- Clinical Nutrition and Risk Factor Modification Center, St Michael's Hospital, Toronto, Ont, Canada.
| | | | | | | | | |
Collapse
|
39
|
Vogt JA, Ishii-Schrade KB, Pencharz PB, Wolever TMS. L-Rhamnose increases serum propionate after long-term supplementation, but lactulose does not raise serum acetate. Am J Clin Nutr 2004; 80:1254-61. [PMID: 15531673 DOI: 10.1093/ajcn/80.5.1254] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Acute ingestion of the unabsorbed sugar l-rhamnose in humans raises serum propionate, whereas acute ingestion of lactulose raises serum acetate. It is not known whether short-chain fatty acid concentrations in urine and feces reflect those in blood. OBJECTIVE The objective was to test the effects of oral l-rhamnose and lactulose for 28 d on acetate and propionate concentrations in serum, urine, and feces. DESIGN Eleven subjects ingested 25 g l-rhamnose, lactulose, or d-glucose (control) for 28 d in a partially randomized crossover design. One fecal sample, hourly blood samples, and all urine samples were collected over 12 h on the last day of each phase. RESULTS The increase in serum propionate was greater after l-rhamnose than after lactulose (P < 0.05). The effect of lactulose on serum acetate was not significant, but lactulose raised the acetate:propionate ratio compared with d-glucose or l-rhamnose in serum (P < 0.005) and urine (P < 0.02). Flatulence was significantly greater after lactulose and l-rhamnose than after d-glucose (P < 0.0001), an effect that lasted 4 wk with lactulose but only 1 wk with l-rhamnose. CONCLUSIONS This study confirmed that l-rhamnose ingestion over 28 d continues to selectively raise serum propionate in humans. Although serum acetate did not increase significantly after lactulose, the serum acetate:propionate ratio was significantly different after l-rhamnose and lactulose, which suggests that these substrates could be used to examine the role of colonic acetate and propionate production in the effect of dietary fiber on lipid metabolism. Changes in the ratio of urinary acetate to propionate reflected those in serum.
Collapse
Affiliation(s)
- Janet A Vogt
- University of Toronto, Department of Nutritional Sciences, Toronto, ON, Canada
| | | | | | | |
Collapse
|
40
|
Dehghan-Kooshkghazi M, Mathers JC. Starch digestion, large-bowel fermentation and intestinal mucosal cell proliferation in rats treated with the alpha-glucosidase inhibitor acarbose. Br J Nutr 2004; 91:357-65. [PMID: 15005821 DOI: 10.1079/bjn20031063] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Acarbose (Glucobay; Bayer) is an alpha-glucosidase inhibitor used to treat diabetes and which may have a role in the prevention of type 2 diabetes. The present study investigated the effects of acarbose treatment on the site and extent of starch digestion, large-bowel fermentation and intestinal mucosal cell proliferation. Eighteen young male Wistar rats were fed "Westernised" diets containing 0, 250 and 500 mg acarbose/kg (six rats/diet) for 21 d. For most variables measured, both acarbose doses had similar effects. Acarbose treatment suppressed starch digestion in the small bowel but there was compensatory salvage by bacterial fermentation in the large bowel. This was accompanied by a substantial hypertrophy of small- and large-bowel tissue and a consistent increase in crypt width along the intestine. Caecal total SCFA pool size was increased more than 4-fold, with even bigger increases for butyrate. These changes in butyrate were reflected in increased molar proportions of butyrate in blood from both the portal vein and heart. There was little effect of acarbose administration on crypt-cell proliferation (significant increase for mid-small intestine only). This is strong evidence against the hypothesis that increased fermentation and increased supply of butyrate enhances intestinal mucosal cell proliferation. In conclusion, apart from the increased faecal loss of starch, there was no evidence of adverse effects of acarbose on the aspects of large-bowel function investigated.
Collapse
Affiliation(s)
- Mahshid Dehghan-Kooshkghazi
- Human Nutrition Research Centre, School of Clinical Medical Sciences, University of Newcastle, Newcastle-upon-Tyne NE1 7RU, UK
| | | |
Collapse
|
41
|
Dolara P, Caderni G, Salvadori M, Morozzi G, Fabiani R, Cresci A, Orpianesi C, Trallori G, Russo A, Palli D. Fecal levels of short-chain fatty acids and bile acids as determinants of colonic mucosal cell proliferation in humans. Nutr Cancer 2003; 42:186-90. [PMID: 12416258 DOI: 10.1207/s15327914nc422_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
We studied the correlation between fecal levels of short-chain fatty acids (SCFA), bile acids (BA), and colonic mucosal proliferation in humans on a free diet. Subjects [n = 43: 27 men and 16 women; 61 +/- 7 and 59 +/- 6 (SE) yr old, respectively] were outpatients who previously underwent resection of at least two sporadic colon polyps. Mucosal proliferation was determined by [3H]thymidine incorporation in vitro in three colorectal biopsies obtained without cathartics and was expressed as labeling index (LI). BA were analyzed in feces by mass spectrometry and SCFA by gas chromatography. We found that increasing levels of BA in feces did not correlate with higher LI. On the contrary, higher levels of SCFA were significantly associated with lower LI in the colonic mucosa (P for trend = 0.02). In conclusion, in humans on a free diet, intestinal proliferation seems to be regulated by the levels of SCFA in feces and not by BA. Because a lower intestinal proliferation is associated with a decreased colon cancer risk, treatments or diets that increase colonic levels of SCFA might be beneficial for colonic mucosa.
Collapse
Affiliation(s)
- Piero Dolara
- Department of Pharmacology, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Behall KM, Howe JC, Anderson RA. Apparent mineral retention is similar in control and hyperinsulinemic men after consumption of high amylose cornstarch. J Nutr 2002; 132:1886-91. [PMID: 12097664 DOI: 10.1093/jn/132.7.1886] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The effects on apparent mineral retention after long-term consumption of a high amylose diet containing 30 g resistant starch (RS) were investigated in 10 control and 14 hyperinsulinemic men. Subjects consumed products (bread, muffins, cookies, corn flakes and cheese puffs) made with standard (70% amylopectin, 30% amylose; AP) or high amylose (70% amylose, 30% amylopectin; AM) cornstarch for two 14-wk periods in a crossover pattern. Starch products replaced usual starches in the habitual diet for 10 wk followed by 4 wk of consuming the controlled diets. During wk 12, all urine, feces and duplicate foods were collected for 7 d. Urinary chromium losses after a glucose tolerance test or 24-h collections of the hyperinsulinemic and control subjects did not differ and were not altered by diet. Except for zinc, the two subject types did not differ significantly in apparent mineral balance. Apparent retentions of calcium and magnesium were not significantly affected by diet (AM vs. AP) or type-by-diet interaction. Apparent iron retention tended to be greater after AM than AP consumption (P < 0.09). Apparent copper retention was greater after consuming AP than after AM (P < 0.02), whereas apparent zinc retention was greater after consuming AM than after AP (P < 0.018). Zinc also showed a significant type-by-diet interaction (P < 0.034) with control subjects retaining less zinc after consuming AP than after AM. In summary, a high amylose cornstarch diet containing 30 g RS could be consumed long term without markedly affecting, and possibly enhancing, retention of some minerals.
Collapse
Affiliation(s)
- Kay M Behall
- Diet and Human Performance, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, MD, USA.
| | | | | |
Collapse
|
43
|
Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol Rev 2001; 81:1031-64. [PMID: 11427691 DOI: 10.1152/physrev.2001.81.3.1031] [Citation(s) in RCA: 1993] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Resistant starch (RS) is starch and products of its small intestinal digestion that enter the large bowel. It occurs for various reasons including chemical structure, cooking of food, chemical modification, and food mastication. Human colonic bacteria ferment RS and nonstarch polysaccharides (NSP; major components of dietary fiber) to short-chain fatty acids (SCFA), mainly acetate, propionate, and butyrate. SCFA stimulate colonic blood flow and fluid and electrolyte uptake. Butyrate is a preferred substrate for colonocytes and appears to promote a normal phenotype in these cells. Fermentation of some RS types favors butyrate production. Measurement of colonic fermentation in humans is difficult, and indirect measures (e.g., fecal samples) or animal models have been used. Of the latter, rodents appear to be of limited value, and pigs or dogs are preferable. RS is less effective than NSP in stool bulking, but epidemiological data suggest that it is more protective against colorectal cancer, possibly via butyrate. RS is a prebiotic, but knowledge of its other interactions with the microflora is limited. The contribution of RS to fermentation and colonic physiology seems to be greater than that of NSP. However, the lack of a generally accepted analytical procedure that accommodates the major influences on RS means this is yet to be established.
Collapse
Affiliation(s)
- D L Topping
- Commonwealth Scientific and Industrial Research Organization, Health Sciences and Nutrition, Adelaide, Australia.
| | | |
Collapse
|
44
|
Fujita H, Yamagami T, Ohshima K. Fermented soybean-derived water-soluble Touchi extract inhibits alpha-glucosidase and is antiglycemic in rats and humans after single oral treatments. J Nutr 2001; 131:1211-3. [PMID: 11285328 DOI: 10.1093/jn/131.4.1211] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A water-soluble extract of Touchi, a traditional Chinese food, was found to exert a strong inhibitory activity against rat intestinal alpha-glucosidase. We orally administered sucrose (2 g/kg) with or without Touchi extract (TE) to normal rats at 100 and 500 mg/kg. Postprandial increases in blood glucose levels at 30 and 60 min after the administration of TE were significantly depressed compared with controls. In humans, eight borderline diabetic subjects were administered 0.1-10.0 g TE before sucrose loading (75 g). TE decreased the glycemic response dose dependently after sucrose loading. Compared with the area under the curve of the postprandial rise in blood glucose with various doses, TE elicited a significant antiglycemic effect at a minimum effective dose of 0.3 g. In addition, when four diabetics were administered 0.3 g TE before eating 200 g of cooked rice, the postprandial increases in blood glucose and mean insulin levels were significantly depressed at 60 and 120 min, respectively, after ingestion compared with levels when no TE was administered. TE, which exhibits alpha-glucosidase inhibitory activity, demonstrated an antihyperglycemic effect and may have potential use in the management of patients with non-insulin-dependent diabetic mellitus.
Collapse
Affiliation(s)
- H Fujita
- Research and Development Department, Nippon Supplement, Inc., Kita-Ku, Osaka, 531-0076, Japan.
| | | | | |
Collapse
|
45
|
Abstract
The importance of glucose control in reducing the complications of diabetes mellitus has been clearly demonstrated. The emergency physician routinely is expected to treat a wide range of problems related to this disease, including making the initial diagnosis of type 2 and occasionally type 1 diabetes. Also common are patients with poorly controlled diabetes. The recent introduction of new classes of agents to lower blood glucose, especially in type 2 diabetes, should improve the control in this category of patient and reduce the complication rate. Some of these agents, such as troglitazone, have potentially fatal complications and require careful monitoring. Emergency physicians should be aware of the common complications of these drugs because patients can present to the ED with them. Hypoglycemia, a common cause of 911 calls and emergency visits, is not a side effect of either metformin or acarbose. Insulin lispro has improved postprandial glycemic control for type 1 and some insulin-requiring type 2 diabetics. Hypoglycemia is less of a risk with insulin lispro, and quality of life is better with this rapidly acting insulin. Newer methods of insulin delivery, such as continuous subcutaneous infusion, have greatly improved glucose control, given greater freedom to patients, and reduced the risks of hypoglycemia.
Collapse
Affiliation(s)
- F B Carlton
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| |
Collapse
|
46
|
Weaver GA, Tangel CT, Krause JA, Parfitt MM, Stragand JJ, Jenkins PL, Erb TA, Davidson RH, Alpern HD, Guiney WB, Higgins PJ. Biomarkers of human colonic cell growth are influenced differently by a history of colonic neoplasia and the consumption of acarbose. J Nutr 2000; 130:2718-25. [PMID: 11053512 DOI: 10.1093/jn/130.11.2718] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The nutritional effects of butyrate on the colonic mucosa and studies of transformed cells suggest that butyrate has anti-colon cancer effects. If butyrate has antineoplastic effects, mucosal growth contrasts between normal subjects and those with a history of colonic neoplasia would parallel changes in growth characteristics caused by butyrate in a colon neoplasia population. To test this hypothesis, rectal biopsies from a survey of colonoscopy patients (n = 50) with and without a history of colonic neoplasia (controls) were compared. Similarly, rectal biopsies were compared from subjects (n = 44) with a colon neoplasia history in an acarbose-placebo crossover trial. Control subjects in the colonoscopy survey had higher bromodeoxyuridine (BrdU) uptake than subjects with a history of neoplasia (P = 0.05). The control subjects also had a higher correlation of BrdU and Ki-67 labeling (P = 0.003). Both findings were paralleled by acarbose use. Acarbose augmented BrdU uptake (P = 0.0001) and improved the correlation of BrdU and Ki-67 labeling (P = 0.013). Acarbose also augmented fecal butyrate (P = 0.0001), which was positively correlated with Ki-67 labeling (P = 0.003). p52 antigen had an earlier pattern of crypt distribution in subjects with a history of colon neoplasia but was not affected by acarbose use. Lewis-Y antigen was expressed earlier in the crypt with acarbose but had similar expression in the colonoscopy survey groups. The use of acarbose to enhance fecal butyrate concentration produced mucosal changes paralleling the findings in control subjects as opposed to those with neoplasia, supporting the concept of an antineoplastic role for butyrate.
Collapse
Affiliation(s)
- G A Weaver
- Department of Medicine, Research Institute and. Department of Pathology, The Mary Imogene Bassett Hospital, Cooperstown, NY 13326, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wolin MJ, Miller TL, Yerry S, Zhang Y, Bank S, Weaver GA. Changes of fermentation pathways of fecal microbial communities associated with a drug treatment that increases dietary starch in the human colon. Appl Environ Microbiol 1999; 65:2807-12. [PMID: 10388668 PMCID: PMC91421 DOI: 10.1128/aem.65.7.2807-2812.1999] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Acarbose inhibits starch digestion in the human small intestine. This increases the amount of starch available for microbial fermentation to acetate, propionate, and butyrate in the colon. Relatively large amounts of butyrate are produced from starch by colonic microbes. Colonic epithelial cells use butyrate as an energy source, and butyrate causes the differentiation of colon cancer cells. In this study we investigated whether colonic fermentation pathways changed during treatment with acarbose. We examined fermentations by fecal suspensions obtained from subjects who participated in an acarbose-placebo crossover trial. After incubation with [1-13C]glucose and 12CO2 or with unlabeled glucose and 13CO2, the distribution of 13C in product C atoms was determined by nuclear magnetic resonance spectrometry and gas chromatography-mass spectrometry. Regardless of the treatment, acetate, propionate, and butyrate were produced from pyruvate formed by the Embden-Meyerhof-Parnas pathway. Considerable amounts of acetate were also formed by the reduction of CO2. Butyrate formation from glucose increased and propionate formation decreased with acarbose treatment. Concomitantly, the amounts of CO2 reduced to acetate were 30% of the total acetate in untreated subjects and 17% of the total acetate in the treated subjects. The acetate, propionate, and butyrate concentrations were 57, 20, and 23% of the total final concentrations, respectively, for the untreated subjects and 57, 13, and 30% of the total final concentrations, respectively, for the treated subjects.
Collapse
Affiliation(s)
- M J Wolin
- Wadsworth Center for Laboratories and Research, New York State Department of Health, Albany, New York 12201-0509, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Jenkins DJ, Vuksan V, Rao AV, Vidgen E, Kendall CW, Tariq N, Würsch P, Koellreutter B, Shiwnarain N, Jeffcoat R. Colonic bacterial activity and serum lipid risk factors for cardiovascular disease. Metabolism 1999; 48:264-8. [PMID: 10024093 DOI: 10.1016/s0026-0495(99)90045-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Antibiotics are being proposed for the treatment of cardiovascular disease. In the past, antibiotics were advocated for the control of hypercholesterolemia. We have therefore investigated the relation between colonic bacterial activity and serum lipids. In a four-phase randomized crossover study, we fed a different starch supplement during each 2-week phase to 24 healthy subjects. In two phases, supplements containing resistant starches were fed that reach the colon and are largely fermented by colonic bacteria. Fecal starch recovery therefore reflects the metabolic activity of colonic microflora. The control treatments were conventional starches. Blood lipid levels were obtained at the start and 4-day fecal collections at the end of each phase. Resistant starch supplements increased fecal starch excretion by 3.8 +/- 1.2 g/d more than conventional starches (P = .006). Mean starch excretion was related positively to pretreatment serum high-density lipoprotein (HDL) cholesterol (r = -.57, P = .003) and negatively to low-density lipoprotein (LDL) cholesterol (r = -.57, P = .004), apolipoprotein B:AI (r = -.56, P = .005), and fecal output of fusobacteria (r = -.73, P = .003) and bacteroides (r = -.72, P = .003). The ratio of fusobacteria to total anaerobes was also related to pretreatment LDL cholesterol (r = .56, P = .037). Differences in starch excretion between healthy subjects, as a measure of bacterial activity, accounted for 32% of the variation in pretreatment LDL cholesterol. The activity of colonic microflora therefore appears to influence serum lipid levels. Alterations of bacterial number and activity may provide an additional strategy to control serum lipid risk factors for cardiovascular disease.
Collapse
Affiliation(s)
- D J Jenkins
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
It is widely accepted that the most challenging goal in the management of patients with diabetes mellitus is to achieve blood glucose levels as close to normal as possible. In general, normalising postprandial blood glucose levels is more difficult than normalising fasting hyperglycaemia. In addition, some epidemiological studies suggest that postprandial hyperglycaemia (PPHG) or hyperinsulinaemia are independent risk factors for the development of macrovascular complications of diabetes mellitus. Recently, several drugs with differing pharmacodynamic profiles have been developed which target PPHG. These include insulin lispro, amylin analogues, alpha-glucosidase inhibitors and meglitinide analogues. Insulin lispro has a more rapid onset of action and shorter duration of efficacy compared with regular human insulin. In clinical trials, the use of insulin lispro was associated with improved control of PPHG and a reduced incidence of hypoglycaemic episodes. Repaglinide, a meglitinide analogue, is a short-acting insulinotropic agent which. when given before meals, stimulates endogenous insulin secretions and lowers postprandial hyperglycaemic excursions. Both insulin lispro and repaglinide are associated with postprandial hyperinsulinaemia. In contrast, amylin analogues reduce PPHG by slowing gastric emptying and delivery of nutrients to the absorbing surface of the gut. Alpha-Glucosidase inhibitors such as acarbose, miglitol and voglibose also reduce PPHG primarily by interfering with the carbohydrate-digesting enzymes and delaying glucose absorption. With the availability of agents which preferentially reduce postprandial blood glucose excursions, it is now possible to achieve glycaemic goals in a larger proportion of individuals with diabetes mellitus.
Collapse
Affiliation(s)
- A D Mooradian
- Department of Internal Medicine, St Louis University Medical School, Missouri 63104, USA.
| | | |
Collapse
|
50
|
Quesada CF, Kimata H, Mori M, Nishimura M, Tsuneyoshi T, Baba S. Piroxicam and acarbose as chemopreventive agents for spontaneous intestinal adenomas in APC gene 1309 knockout mice. Jpn J Cancer Res 1998; 89:392-6. [PMID: 9617344 PMCID: PMC5921824 DOI: 10.1111/j.1349-7006.1998.tb00576.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The use of nonsteroidal anti-inflammatory drugs has been suggested to have a chemopreventive effect against colon carcinoma, through the inhibition of cyclooxygenases 1 and 2, in patients with familial adenomatous polyposis and in animal models. Acarbose, an alpha-glycosidase inhibitor, may also be chemopreventive. In order to examine the effects of these drugs we employed APC gene knockout mice randomized into 3 groups, one for treatment with piroxicam (0.05% concentration in drinking water), one for acarbose (0.04% concentration in food) and another for the control. After 14 weeks of treatment, mice were killed for quantitation of gastric and intestinal adenomas. Tumor multiplicity in the whole gastrointestinal tract decreased from 33.89 +/- 13.07 tumors/mouse in the control group to 17.05 +/- 7 tumors/mouse in the piroxicam-treated group (P < 0.001). The decrease in the acarbose-treated group (29.68 +/- 12.86 tumors/mouse) was not significant (P < 0.05). The number of tumors > or = 3 mm in diameter was also quantified in all gastrointestinal segments. The number of such tumors in the piroxicam group was decreased to 0.56 +/- 1.2 tumors/mouse from the control value of 3.78 +/- 1.17 tumors/mouse (P < 0.001), while in the acarbose-treated group the number decreased to 2.36 +/- 1.7 tumors/mouse (P < 0.01). Thus, piroxicam decreases the size and number of gastrointestinal adenomas in APC 1309 knockout mice, while acarbose decreases only the size.
Collapse
Affiliation(s)
- C F Quesada
- Second Department of Surgery, Hamamatsu University School of Medicine, Shizuoka
| | | | | | | | | | | |
Collapse
|