1
|
Smits MM, von Voss L, Drzazga AK, Beaman EE, Brethvad AO, Holst JJ, Rosenkilde MM. Alpha-cyclodextrin increases glucagon-like peptide-1 secretion in multiple models and improves metabolic status in mice. Food Chem 2024; 460:140759. [PMID: 39142205 DOI: 10.1016/j.foodchem.2024.140759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Alpha-cyclodextrin (α-CD) is a non-absorbable and soluble fiber that causes weight loss. We studied whether this is due to an effect on GLP-1 secretion. In GLUTag cells, α-CD increased GLP-1 secretion up to 170% via adenylyl cyclase, phospholipase C, and L-type calcium channels dependent processes. In rat isolated colon perfusions, luminal α-CD increased GLP-1 secretion with 20%. In lean mice, once daily α-CD versus saline caused weight loss and lowered the peak in glucose after an oral glucose tolerance test (OGTT). In obese mice, α-CD added to high-fat diet caused weight loss similar to the control group (receiving cellulose). However, compared to cellulose, the α-CD group ate less. During an OGTT, no differences were observed in glucose, insulin and GLP-1. Thus, α-CD increases GLP-1 secretion in a dose-dependent manner and could be a safe and easy addition to food products to help reduce body weight.
Collapse
Affiliation(s)
- Mark M Smits
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Liv von Voss
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Katarzyna Drzazga
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Lodz, Poland.
| | - Emily Eufaula Beaman
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
2
|
Zhang Y, Zhou Z, Luan H, Zhang X, Liu M, Wang K, Wang F, Feng W, Xu W, Song P. Advances in the biosynthesis of D-allulose. World J Microbiol Biotechnol 2024; 40:375. [PMID: 39487344 DOI: 10.1007/s11274-024-04166-w] [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: 07/24/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
D-allulose is a rare monosaccharide and a C-3 epimer of D-fructose. It has physiological functions, such as antihyperglycemic, obesity-preventing, neuroprotective, and reactive oxygen species (ROS) scavenging effects, making it an ideal sugar substitute. The synthesis methods for D-allulose include chemical synthesis and biosynthesis. Chemical synthesis requires strict reaction conditions and tends to produce byproducts. Biosynthesis is mainly an enzymatic process. Enzymatic catalysis for the conversion of starch or glycerol to D-allulose is performed mainly by enzymes such as isoamylase (IA), glucose isomerase (GI), D-allulose 3-epimerase (DPE), D-allulose-6-phosphate 3-epimerase (A6PE), D-allulose 6-phosphate phosphatase (A6PP), ribitol 2-dehydrogenase (RDH), glycerophosphate kinase (GK), glycerophosphate oxidase (GPO), and dihydroxyacetone phosphate (DHAP)-dependent aldolase. Biosynthesis is a more energy-efficient process, producing fewer harmful by-products and pollutants, and significantly reducing negative environmental impacts. Furthermore, the specific catalytic activity of enzymes facilitates the production of compounds of higher purity, thereby facilitating the isolation and purification of the products. It has thus become the main method for producing D-allulose. This article reviews the progress in research on the biosynthetic production of D-allulose, focusing on the enzymes involved and their enzymatic properties, and discusses the production prospects for D-allulose.
Collapse
Affiliation(s)
- Yue Zhang
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Zhengsong Zhou
- Shandong Aocter Biotechnology Co., Ltd, Liaocheng, 252000, China
| | - Haoni Luan
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Xue Zhang
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Mengyu Liu
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Kuiming Wang
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Fei Wang
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Wei Feng
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Wei Xu
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Peng Song
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China.
| |
Collapse
|
3
|
Shintani T, Yanai S, Kanasaki A, Iida T, Endo S. Long-term d-allose administration ameliorates age-related cognitive impairment and loss of bone strength in male mice. Exp Gerontol 2024; 196:112555. [PMID: 39179160 DOI: 10.1016/j.exger.2024.112555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Age-related physical and cognitive decline may be ameliorated by consuming functional foods. d-Allose, reported to have multiple health benefits, may temper aging phenotypes, particularly brain function. We investigated whether d-allose supplementation improves cognitive function. A standard battery of behavioral tests was administered to 18-month-old male mice after consuming diet containing 3 % d-allose for 6 months. Following a wire-hanging test, an open-field test, Morris water maze, fear-conditioning, and an analgesia test were sequentially performed. Bone density and strength were assessed afterwards. Possible mechanism(s) under-lying memory changes in hippocampus were also examined with a DNA microarray. d-Allose failed to influence muscle strength, locomotor activity and anxiety, fear memory, or pain sensitivity. However, d-allose improved hippocampus-dependent spatial learning and memory, and it may contribute to increase bone strength. d-Allose also changed the expression of some genes in hippocampus involved in cognitive functions. Long-term d-allose supplementation appears to modestly change aging phenotypes and improve spatial memory.
Collapse
Affiliation(s)
- Tomoya Shintani
- Research and Development, Matsutani Chemical Industry Co., Ltd., 5-3 Kita-Itami, Itami City, Hyogo 664-8508, Japan
| | - Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan; Animal Facility, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Akane Kanasaki
- Research and Development, Matsutani Chemical Industry Co., Ltd., 5-3 Kita-Itami, Itami City, Hyogo 664-8508, Japan.
| | - Tetsuo Iida
- Research and Development, Matsutani Chemical Industry Co., Ltd., 5-3 Kita-Itami, Itami City, Hyogo 664-8508, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
4
|
Khamees Thabet H, Ammar YA, Imran M, Hamdy Helal M, Ibrahim Alaqel S, Alshehri A, Ash Mohd A, Abusaif MS, Ragab A. Unveiling anti-diabetic potential of new thiazole-sulfonamide derivatives: Design, synthesis, in vitro bio-evaluation targeting DPP-4, α-glucosidase, and α-amylase with in-silico ADMET and docking simulation. Bioorg Chem 2024; 151:107671. [PMID: 39067419 DOI: 10.1016/j.bioorg.2024.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Diabetes mellitus type 2 (T2DM) can be managed by targeting dipeptidyl peptidase-4 (DPP-4), an enzyme that breaks down and deactivates peptides such as GIP and GLP-1. In this context, a new series of 2-(2-substituted hydrazineyl)thiazole derivatives 4, 5, 6, 8, 10, and 11 conjugated with the 2-hydroxy-5-(pyrrolidin-1-ylsulfonyl)benzylidene fragment were designed and synthesized. The virtual screening of the designed derivatives inside DPP-4 demonstrated good to moderate activity, with binding affinity ranging from -6.86 to -5.36 kcal/mol compared to Sitagliptin (S=-5.58 kcal/mol). These results encourage us to evaluate DPP-4 using in-vitro fluorescence-based assay. The in-vitro results exhibited inhibitory percentage (IP) values ranging from 40.66 to 75.62 % in comparison to Sitagliptin (IP=63.14 %) at 100 µM. Subsequently, the IC50 values were determined, and the 5-aryl thiazole derivatives 10 and 11 revealed strong potent IC50 values 2.75 ± 0.27 and 2.51 ± 0.27 µM, respectively, compared to Sitagliptin (3.32 ± 0.22 µM). The SAR study exhibited the importance of the substituents on the thiazole scaffold, especially with the hydrophobic fragment at C5 of the thiazole, which has a role in the activity. Compounds 10 and 11 were further assessed toward α-glucosidase and α-amylase enzymes and give promising results. Compound 10 showed good activity against α-glucosidase with IC50 value of 3.02 ± 0.23 µM compared to Acarbose 3.05 ± 0.22 µM and (11 = 3.34 ± 0.10 µM). On the other hand, for α-amylase, compound 11 was found to be most effective with IC50 value of 2.91 ± 0.23 µM compared to compound 10 = 3.30 ± 0.16 µM and Acarbose (2.99 ± 0.21 µM) indicating that these derivatives could reduce glucose by more than one target. The most active derivatives 10 and 11 attracted great interest as candidates for oral bioavailability and safe toxicity profiles compared to positive controls. The in-silico docking simulation was performed to understand the binding interactions inside the DPP-4, α-glucosidase, and α-amylase pockets, and it was found to be promising antidiabetic agents through a number of interactions.
Collapse
Affiliation(s)
- Hamdy Khamees Thabet
- Department of Chemistry, College of Sciences and Arts, Northern Border University, Rafha, 91911, Saudi Arabia.
| | - Yousry A Ammar
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City 11884, Cairo, Egypt
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mohamed Hamdy Helal
- Department of Chemistry, College of Sciences and Arts, Northern Border University, Rafha, 91911, Saudi Arabia
| | - Saleh Ibrahim Alaqel
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Ahmed Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, King Faisal Road, Dammam 31441, Saudi Arabia
| | - Abida Ash Mohd
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Moustafa S Abusaif
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City 11884, Cairo, Egypt
| | - Ahmed Ragab
- Department of Chemistry, Faculty of Science (Boys), Al-Azhar University, Nasr City 11884, Cairo, Egypt.
| |
Collapse
|
5
|
Ohbayashi K, Sugiyama Y, Nohmi T, Nishimura K, Nakazaki T, Sato YI, Masumura T, Iwasaki Y. Anekomochi glutinous rice provides low postprandial glycemic response by enhanced insulin action via GLP-1 release and vagal afferents activation. J Physiol Sci 2024; 74:47. [PMID: 39333851 PMCID: PMC11428336 DOI: 10.1186/s12576-024-00940-5] [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: 07/31/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
Glutinous rice (mochi rice), compared to non-glutinous rice (uruchi rice), exhibits a wide range of glycemic index (GI) values, from low to high. However, the underlying mechanisms behind the variation in GI values remain poorly understood. In this study, we aimed to identify rice cultivars with a low postprandial glycemic response and investigate the mechanisms, focusing on insulin and incretin hormones. We examined seven glutinous rice cultivars and three non-glutinous rice cultivars. We discovered that Anekomochi, a glutinous rice cultivar, has the lowest postprandial glycemic response. Anekomochi significantly enhanced glucagon-like peptide-1 (GLP-1) secretion while suppressing insulin secretion. These effects were completely blunted by inhibiting GLP-1 receptor signaling and denervating the common hepatic branch of vagal afferent nerves that are crucial for sensing intestinal GLP-1. Our findings demonstrate that Anekomochi markedly enhances insulin action via GLP-1 release and vagal afferent neural pathways, thereby leading to a lower postprandial glycemic response.
Collapse
Affiliation(s)
- Kento Ohbayashi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Yudai Sugiyama
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Taichi Nohmi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Kazusa Nishimura
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, 1-1-1 Tsushimanaka, Kita-Ku, Okayama, 700-0082, Japan
- Graduate School of Agriculture, Kyoto University, 4-2-1, Shiroyamadai, Kizugawa, Kyoto, 619-0218, Japan
| | - Tetsuya Nakazaki
- Graduate School of Agriculture, Kyoto University, 4-2-1, Shiroyamadai, Kizugawa, Kyoto, 619-0218, Japan
- Office of Institutional Advancement and Communications, Kyoto University, Yoshida-Honmachi, Sakyo-Ku, Kyoto, 606-8501, Japan
| | - Yo-Ichiro Sato
- Research Center for Japanese Food Culture, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
- Museum of Natural and Environmental History, Shizuoka, 5762 Oya, Suruga-Ku, Shizuoka, 422-8017, Japan
| | - Takehiro Masumura
- Laboratory of Genetic Engineering, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan.
| |
Collapse
|
6
|
Binvignat M, Sellam J, Berenbaum F, Felson DT. The role of obesity and adipose tissue dysfunction in osteoarthritis pain. Nat Rev Rheumatol 2024; 20:565-584. [PMID: 39112603 DOI: 10.1038/s41584-024-01143-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 08/29/2024]
Abstract
Obesity has a pivotal and multifaceted role in pain associated with osteoarthritis (OA), extending beyond the mechanistic influence of BMI. It exerts its effects both directly and indirectly through various modifiable risk factors associated with OA-related pain. Adipose tissue dysfunction is highly involved in OA-related pain through local and systemic inflammation, immune dysfunction, and the production of pro-inflammatory cytokines and adipokines. Adipose tissue dysfunction is intricately connected with metabolic syndrome, which independently exerts specific effects on OA-related pain, distinct from its association with BMI. The interplay among obesity, adipose tissue dysfunction and metabolic syndrome influences OA-related pain through diverse pain mechanisms, including nociceptive pain, peripheral sensitization and central sensitization. These complex interactions contribute to the heightened pain experience observed in individuals with OA and obesity. In addition, pain management strategies are less efficient in individuals with obesity. Importantly, therapeutic interventions targeting obesity and metabolic syndrome hold promise in managing OA-related pain. A deeper understanding of the intricate relationship between obesity, metabolic syndrome and OA-related pain is crucial and could have important implications for improving pain management and developing innovative therapeutic options in OA.
Collapse
Affiliation(s)
- Marie Binvignat
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Sorbonne University, INSERM UMRS_959, I3 Lab Immunology Immunopathology Immunotherapy, Paris, France
| | - Jérémie Sellam
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France.
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.
| | - Francis Berenbaum
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - David T Felson
- Boston University School of Medicine, Department of Medicine, Section of Rheumatology, Boston, MA, USA
| |
Collapse
|
7
|
Hoekx CA, Straat ME, Bizino MB, van Eyk HJ, Lamb HJ, Smit JWA, Jazet IM, de Jager SCA, Boon MR, Martinez‐Tellez B. Growth differentiation factor 15 is not modified after weight loss induced by liraglutide in South Asians and Europids with type 2 diabetes mellitus. Exp Physiol 2024; 109:1292-1304. [PMID: 38965822 PMCID: PMC11291866 DOI: 10.1113/ep091815] [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: 02/21/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists induce weight loss in patients with type 2 diabetes mellitus (T2DM), but the underlying mechanism is unclear. Recently, the mechanism by which metformin induces weight loss could be explained by an increase in growth differentiation factor 15 (GDF15), which suppresses appetite. Therefore, we aimed to investigate whether the GLP-1R agonist liraglutide modifies plasma GDF15 levels in patients with T2DM. GDF15 levels were measured in plasma samples obtained from Dutch Europids and Dutch South Asians with T2DM before and after 26 weeks of treatment with daily liraglutide (n = 44) or placebo (n = 50) added to standard care. At baseline, circulating GDF15 levels did not differ between South Asians and Europids with T2DM. Treatment with liraglutide, compared to placebo, decreased body weight, but did not modify plasma GDF15 levels in all patients, or when data were split by ethnicity. Also, the change in plasma GDF15 levels after treatment with liraglutide did not correlate with changes in body weight or HbA1c levels. In addition, the dose of metformin used did not correlate with baseline plasma GDF15 levels. Compared to placebo, liraglutide treatment for 26 weeks does not modify plasma GDF15 levels in Dutch Europid or South Asian patients with T2DM. Thus, the weight loss induced by liraglutide is likely explained by other mechanisms beyond the GDF15 pathway. HIGHLIGHTS: What is the central question of this study? Growth differentiation factor 15 (GDF15) suppresses appetite and is increased by metformin: does the GLP-1R agonist liraglutide modify plasma GDF15 levels in patients with type 2 diabetes mellitus (T2DM)? What is the main finding and its importance? Plasma GDF15 levels did not differ between South Asians and Europids with T2DM and were not modified by 26 weeks of liraglutide in either ethnicity. Moreover, there was no correlation between the changes in plasma GDF15 levels and dosage of metformin administered, changes in body weight or HbA1c levels. The appetite-suppressing effect of liraglutide is likely exerted via pathways other than GDF15.
Collapse
Affiliation(s)
- Carlijn A. Hoekx
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Maaike E. Straat
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Maurice B. Bizino
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Huub J. van Eyk
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | | | - Johannes W. A. Smit
- Department of MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Ingrid M. Jazet
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Translational ImmunologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Borja Martinez‐Tellez
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Nursing Physiotherapy and Medicine, SPORT Research Group (CTS‐1024), CERNEP Research CenterUniversity of AlmeríaAlmeríaSpain
- Biomedical Research UnitTorrecárdenas University HospitalAlmeríaSpain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIGranadaSpain
| |
Collapse
|
8
|
Perez PA, Wiley MB, Makriyannis A, DiPatrizio NV. Cannabinoids Block Fat-induced Incretin Release via CB 1-dependent and CB 1-independent Pathways in Intestinal Epithelium. GASTRO HEP ADVANCES 2024; 3:931-941. [PMID: 39318720 PMCID: PMC11419882 DOI: 10.1016/j.gastha.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/11/2024] [Indexed: 09/26/2024]
Abstract
Background and Aims Glucose homeostasis is regulated by a dynamic interplay between hormones along the gastro-insular axis. For example, enteroendocrine L- and K- cells that line the intestine produce the incretins glucagon-like peptide-1 (GLP1) and glucose-dependent insulinotropic polypeptide (GIP), respectively, which are secreted following a meal. Broadly, incretin signaling enhances insulin release from the endocrine pancreas and participates in the control of food intake, and therapeutics that mimic their activity have recently been developed for the treatment of type-2 diabetes and obesity. Notably, genes for cannabinoid subtype-1 receptor (CB1R) are expressed in these cell subpopulations; however, roles for CB1Rs in controlling fat-induced incretin release are unclear. To address this gap in our understanding, we tested the hypothesis that intestinal epithelial CB1Rs control fat-induced incretin secretion. Methods We treated mice with conditional deletion of CB1Rs in the intestinal epithelium (IntCB1-/-) or controls (IntCB1+/+) with oil gavage to stimulate incretin release in the presence of the cannabinoid receptor agonists, WIN55,212-2 or Δ9 tetrahydrocannabinol (THC), and the peripherally-restricted CB1R antagonist AM6545. Circulating incretin levels were measured in plasma. Results Oral gavage of corn oil increased levels of bioactive GLP1 and GIP in IntCB1+/+ mouse plasma. Pretreatment with the WIN55,212-2 or THC blocked this response, which was largely reversed by coadministration with AM6545. WIN55,212-2 failed to inhibit fat-induced GIP release, but not GLP1, in IntCB1-/- mice. In contrast, THC inhibited the secretion of incretins irrespective of CB1R expression in intestinal epithelial cells. Conclusion These results indicate that cannabinoid receptor agonists can differentially inhibit incretin release via mechanisms that include intestinal epithelial CB1R-dependent and CB1R-independent mechanisms.
Collapse
Affiliation(s)
- Pedro Antonio Perez
- Center for Cannabinoid Research (UCRCCR), School of Medicine, University of California, Riverside, Riverside, California
- Department of Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California
| | - Mark Benjamin Wiley
- Center for Cannabinoid Research (UCRCCR), School of Medicine, University of California, Riverside, Riverside, California
| | | | - Nicholas Vincent DiPatrizio
- Center for Cannabinoid Research (UCRCCR), School of Medicine, University of California, Riverside, Riverside, California
| |
Collapse
|
9
|
Nishida K, Ueno S, Seino Y, Hidaka S, Murao N, Asano Y, Fujisawa H, Shibata M, Takayanagi T, Ohbayashi K, Iwasaki Y, Iizuka K, Okuda S, Tanaka M, Fujii T, Tochio T, Yabe D, Yamada Y, Sugimura Y, Hirooka Y, Hayashi Y, Suzuki A. Impaired Fat Absorption from Intestinal Tract in High-Fat Diet Fed Male Mice Deficient in Proglucagon-Derived Peptides. Nutrients 2024; 16:2270. [PMID: 39064713 PMCID: PMC11280123 DOI: 10.3390/nu16142270] [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: 06/19/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: Proglucagon-derived peptides (PDGPs) including glucagon (Gcg), GLP-1, and GLP-2 regulate lipid metabolism in the liver, adipocytes, and intestine. However, the mechanism by which PGDPs participate in alterations in lipid metabolism induced by high-fat diet (HFD) feeding has not been elucidated. (2) Methods: Mice deficient in PGDP (GCGKO) and control mice were fed HFD for 7 days and analyzed, and differences in lipid metabolism in the liver, adipose tissue, and duodenum were investigated. (3) Results: GCGKO mice under HFD showed lower expression levels of the genes involved in free fatty acid (FFA) oxidation such as Hsl, Atgl, Cpt1a, Acox1 (p < 0.05), and Pparα (p = 0.05) mRNA in the liver than in control mice, and both FFA and triglycerides content in liver and adipose tissue weight were lower in the GCGKO mice. On the other hand, phosphorylation of hormone-sensitive lipase (HSL) in white adipose tissue did not differ between the two groups. GCGKO mice under HFD exhibited lower expression levels of Pparα and Cd36 mRNA in the duodenum as well as increased fecal cholesterol contents compared to HFD-controls. (4) Conclusions: GCGKO mice fed HFD exhibit a lesser increase in hepatic FFA and triglyceride contents and adipose tissue weight, despite reduced β-oxidation in the liver, than in control mice. Thus, the absence of PGDP prevents dietary-induced fatty liver development due to decreased lipid uptake in the intestinal tract.
Collapse
Affiliation(s)
- Koki Nishida
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Shinji Ueno
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Yusuke Seino
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
| | - Shihomi Hidaka
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Naoya Murao
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
| | - Yuki Asano
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Haruki Fujisawa
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Megumi Shibata
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Takeshi Takayanagi
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Kento Ohbayashi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan; (K.O.); (Y.I.)
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan; (K.O.); (Y.I.)
| | - Katsumi Iizuka
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan;
| | - Shoei Okuda
- Graduate School of Bioscience and Biotechnology, College of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan; (S.O.); (M.T.)
| | - Mamoru Tanaka
- Graduate School of Bioscience and Biotechnology, College of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan; (S.O.); (M.T.)
| | - Tadashi Fujii
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake 470-1192, Japan; (T.F.); (T.T.); (Y.H.)
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake 470-1101, Japan
- BIOSIS Lab. Co., Ltd., Toyoake 470-1192, Japan
| | - Takumi Tochio
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake 470-1192, Japan; (T.F.); (T.T.); (Y.H.)
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake 470-1101, Japan
- BIOSIS Lab. Co., Ltd., Toyoake 470-1192, Japan
| | - Daisuke Yabe
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
- Center for One Medicine Innovative Translational Research, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yuuichiro Yamada
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
| | - Yoshihisa Sugimura
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Yoshiki Hirooka
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake 470-1192, Japan; (T.F.); (T.T.); (Y.H.)
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake 470-1101, Japan
- BIOSIS Lab. Co., Ltd., Toyoake 470-1192, Japan
| | - Yoshitaka Hayashi
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan;
- Department of Endocrinology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Atsushi Suzuki
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| |
Collapse
|
10
|
Fathi PA, Bales MB, Ayala JE. Time-dependent changes in feeding behavior and energy balance associated with weight gain in mice fed obesogenic diets. Obesity (Silver Spring) 2024; 32:1373-1388. [PMID: 38932722 DOI: 10.1002/oby.24052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Obesity is characterized by dysregulated homeostatic mechanisms resulting in positive energy balance; however, when this dysregulation occurs is unknown. We assessed the time course of alterations to behaviors promoting weight gain in male and female mice switched to an obesogenic high-fat diet (HFD). METHODS Male and female C57BL/6J mice were housed in metabolic chambers and were switched from chow to a 60% or 45% HFD for 4 and 3 weeks, respectively. Food intake, meal patterns, energy expenditure (EE), and body weight were continuously measured. A separate cohort of male mice was switched from chow to a 60% HFD and was given access to locked or unlocked running wheels. RESULTS Switching mice to obesogenic diets promotes transient bouts of hyperphagia during the first 2 weeks followed by persistent caloric hyperphagia. EE increases but not sufficiently enough to offset increased caloric intake, resulting in a sustained net positive energy balance. Hyperphagia is associated with consumption of calorically larger meals (impaired satiation) more frequently (impaired satiety), particularly during the light cycle. Running wheel exercise delays weight gain in male mice fed a 60% HFD by enhancing satiation and increasing EE. However, exercise effects on satiation are no longer apparent after 2 weeks, coinciding with weight gain. CONCLUSIONS Exposure to obesogenic diets engages homeostatic regulatory mechanisms for ~2 weeks that ultimately fail, and consequent weight gain is characterized by impaired satiation and satiety. Insights into the etiology of obesity can be obtained by investigating changes to satiation and satiety mechanisms during the initial ~2 weeks of HFD exposure.
Collapse
Affiliation(s)
- Payam A Fathi
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Michelle B Bales
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Julio E Ayala
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, Tennessee, USA
- Vanderbilt Center for Addiction Research, Nashville, Tennessee, USA
| |
Collapse
|
11
|
Lee CY, So YS, Yoo SH, Lee BH, Seo DH. Impact of artificial sweeteners and rare sugars on the gut microbiome. Food Sci Biotechnol 2024; 33:2047-2064. [PMID: 39130663 PMCID: PMC11315849 DOI: 10.1007/s10068-024-01597-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/03/2024] [Accepted: 04/30/2024] [Indexed: 08/13/2024] Open
Abstract
Alternative sugars are often used as sugar substitutes because of their low calories and glycemic index. Recently, consumption of these sweeteners in diet foods and beverages has increased dramatically, raising concerns about their health effects. This review examines the types and characteristics of artificial sweeteners and rare sugars and analyzes their impact on the gut microbiome. In the section on artificial sweeteners, we have described the chemical structures of different sweeteners, their digestion and absorption processes, and their effects on the gut microbiota. We have also discussed the biochemical properties and production methods of rare sugars and their positive and negative effects on gut microbial communities. Finally, we have described how artificial sweeteners and rare sugars alter the gut microbiome and how these changes affect the gut environment. Our observations aim to improve our understanding regarding the potential health implications of the consumption of artificial sweeteners and low-calorie sugars.
Collapse
Affiliation(s)
- Chang-Young Lee
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, 54896 Republic of Korea
| | - Yun-Sang So
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul, 05006 Republic of Korea
| | - Sang-Ho Yoo
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul, 05006 Republic of Korea
| | - Byung-Hoo Lee
- Department of Food Science & Biotechnology, Gachon University, Seongnam, 13120 Republic of Korea
| | - Dong-Ho Seo
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, 54896 Republic of Korea
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul, 05006 Republic of Korea
| |
Collapse
|
12
|
Cayabyab KB, Shin MJ, Heimuli MS, Kim IJ, D’Agostino DP, Johnson RJ, Koutnik AP, Bellissimo N, Diamond DM, Norwitz NG, Arroyo JA, Reynolds PR, Bikman BT. The Metabolic and Endocrine Effects of a 12-Week Allulose-Rich Diet. Nutrients 2024; 16:1821. [PMID: 38931176 PMCID: PMC11207032 DOI: 10.3390/nu16121821] [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] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
The global rise in type 2 diabetes (T2D) and obesity necessitates innovative dietary interventions. This study investigates the effects of allulose, a rare sugar shown to reduce blood glucose, in a rat model of diet-induced obesity and T2D. Over 12 weeks, we hypothesized that allulose supplementation would improve body weight, insulin sensitivity, and glycemic control. Our results showed that allulose mitigated the adverse effects of high-fat, high-sugar diets, including reduced body weight gain and improved insulin resistance. The allulose group exhibited lower food consumption and increased levels of glucagon-like peptide-1 (GLP-1), enhancing glucose regulation and appetite control. Additionally, allulose prevented liver triglyceride accumulation and promoted mitochondrial uncoupling in adipose tissue. These findings suggest that allulose supplementation can improve metabolic health markers, making it a promising dietary component for managing obesity and T2D. Further research is needed to explore the long-term benefits and mechanisms of allulose in metabolic disease prevention and management. This study supports the potential of allulose as a safe and effective intervention for improving metabolic health in the context of dietary excess.
Collapse
Affiliation(s)
- Kevin B. Cayabyab
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Marley J. Shin
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Micah S. Heimuli
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Iris J. Kim
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Dominic P. D’Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
| | | | | | - Nick Bellissimo
- School of Nutrition, Toronto Metropolitan University, Toronto, ON M5S 1A8, Canada
| | - David M. Diamond
- Department of Psychology, University of South Florida, Tampa, FL 33602, USA
| | | | - Juan A. Arroyo
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Paul R. Reynolds
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Benjamin T. Bikman
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
13
|
Abdullah bin Ahmed I. A Comprehensive Review on Weight Gain following Discontinuation of Glucagon-Like Peptide-1 Receptor Agonists for Obesity. J Obes 2024; 2024:8056440. [PMID: 38765635 PMCID: PMC11101251 DOI: 10.1155/2024/8056440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
Obesity is considered the leading public health problem in the medical sector. The phenotype includes overweight conditions that lead to several other comorbidities that drastically decrease health. Glucagon-like receptor agonists (GLP-1RAs) initially designed for treating type 2 diabetes mellitus (T2DM) had demonstrated weight loss benefits in several clinical trials. In vivo studies showed that GLP-1RA encourages reduced food consumption and consequent weight reduction by stimulating brown fat and enhancing energy outlay through the action of the sympathetic nervous system (SNS) pathways. Additionally, GLP-1RAs were found to regulate food intake through stimulation of sensory neurons in the vagus, interaction with the hypothalamus and hindbrain, and through inflammation and intestinal microbiota. However, the main concern with the use of GLP-1RA treatment was weight gain after withdrawal or discontinuation. We could identify three different ways that could lead to weight gain. Potential factors might include temporary hormonal adjustment in response to weight reduction, the central nervous system's (CNS) incompetence in regulating weight augmentation owing to the lack of GLP-1RA, and β-cell malfunction due to sustained exposure to GLP-1RA. Here, we also review the data from clinical studies that reported withdrawal symptoms. Although the use of GLP-1RA could be beneficial in multiple ways, withdrawal after years has the symptoms reversed. Clinical studies should emphasize the downside of these views we highlighted, and mechanistic studies must be carried out for a better outcome with GLP-1RA from the laboratory to the bedside.
Collapse
Affiliation(s)
- Ibrahim Abdullah bin Ahmed
- Department of Family Medicine, Faculty of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Zhang W, Ren H, Chen J, Ni D, Xu W, Mu W. Enhancement of the d-Allulose 3-Epimerase Expression in Bacillus subtilis through Both Transcriptional and Translational Regulations. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8052-8059. [PMID: 38563420 DOI: 10.1021/acs.jafc.4c01122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
d-Allulose, a functional bulk sweetener, has recently attracted increasing attention because of its low-caloric-ness properties and diverse health effects. d-Allulose is industrially produced by the enzymatic epimerization of d-fructose, which is catalyzed by ketose 3-epimerase (KEase). In this study, the food-grade expression of KEase was studied using Bacillus subtills as the host. Clostridium sp. d-allulose 3-epimerase (Clsp-DAEase) was screened from nine d-allulose-producing KEases, showing better potential for expression in B. subtills WB600. Promoter-based transcriptional regulation and N-terminal coding sequence (NCS)-based translational regulation were studied to enhance the DAEase expression level. In addition, the synergistic effect of promoter and NCS on the Clsp-DAEase expression was studied. Finally, the strain with the combination of a PHapII promoter and gln A-Up NCS was selected as the best Clsp-DAEase-producing strain. It efficiently produced Clsp-DAEase with a total activity of 333.2 and 1860.6 U/mL by shake-flask and fed-batch cultivations, respectively.
Collapse
Affiliation(s)
- Wenli Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Hu Ren
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - JiaJun Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Dawei Ni
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wei Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
15
|
Molonia MS, Salamone FL, Speciale A, Saija A, Cimino F. D-Allulose Reduces Hypertrophy and Endoplasmic Reticulum Stress Induced by Palmitic Acid in Murine 3T3-L1 Adipocytes. Int J Mol Sci 2024; 25:4059. [PMID: 38612868 PMCID: PMC11012259 DOI: 10.3390/ijms25074059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/13/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Natural rare sugars are an alternative category of sweeteners with positive physiologic and metabolic effects both in in vitro and animal models. D-allulose is a D-fructose epimer that combines 70% sucrose sweetness with the advantage of an extremely low energy content. However, there are no data about the effect of D-allulose against adipose dysfunction; thus, it remains to be confirmed whether D-allulose is useful in the prevention and in treatment of adipose tissue alterations. With this aim, we evaluated D-allulose's preventive effects on lipid accumulation in 3T3-L1 murine adipocytes exposed to palmitic acid (PA), a trigger for hypertrophic adipocytes. D-allulose in place of glucose prevented adipocyte hypertrophy and the activation of adipogenic markers C/EBP-β and PPARγ induced by high PA concentrations. Additionally, D-allulose pretreatment inhibited the NF-κB pathway and endoplasmic reticulum stress caused by PA, through activation of the Nrf2 pathway. Interestingly, these effects were also observed as D-allulose post PA treatment. Although our data need to be confirmed through in vivo models, our findings suggest that incorporating D-allulose as a glucose substitute in the diet might have a protective role in adipocyte function and support a unique mechanism of action in this sugar as a preventive or therapeutic compound against PA lipotoxicity through the modulation of pathways connected to lipid transport and metabolism.
Collapse
Affiliation(s)
- Maria Sofia Molonia
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy; (M.S.M.); (F.L.S.); (A.S.); (F.C.)
- “Prof. Antonio Imbesi” Foundation, University of Messina, 98100 Messina, Italy
| | - Federica Lina Salamone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy; (M.S.M.); (F.L.S.); (A.S.); (F.C.)
| | - Antonio Speciale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy; (M.S.M.); (F.L.S.); (A.S.); (F.C.)
| | - Antonella Saija
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy; (M.S.M.); (F.L.S.); (A.S.); (F.C.)
| | - Francesco Cimino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy; (M.S.M.); (F.L.S.); (A.S.); (F.C.)
| |
Collapse
|
16
|
Fathi PA, Bales MB, Ayala JE. Time dependent changes in feeding behavior and energy balance associated with weight gain in mice fed obesogenic diets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575043. [PMID: 38260337 PMCID: PMC10802492 DOI: 10.1101/2024.01.10.575043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Obesity is characterized by dysregulated homeostatic mechanisms resulting in positive energy balance, yet when this dysregulation occurs is unknown. We assessed the time course of alterations to behaviors promoting weight gain in male and female mice switched to obesogenic 60% or 45% high fat diet (HFD). Switching mice to obesogenic diets promotes transient bouts of hyperphagia during the first 2 weeks followed by persistent caloric hyperphagia. Energy expenditure increases but not sufficiently to offset increased caloric intake, resulting in a sustained net positive energy balance. Hyperphagia is associated with consumption of calorically larger meals (impaired satiation) more frequently (impaired satiety) particularly during the light-cycle. Running wheel exercise delays weight gain in 60% HFD-fed male mice by enhancing satiation and increasing energy expenditure. However, exercise effects on satiation are no longer apparent after 2 weeks, coinciding with weight gain. Thus, exposure to obesogenic diets engages homeostatic regulatory mechanisms for ∼2 weeks that ultimately fail, and consequent weight gain is characterized by impaired satiation and satiety. Insights into the etiology of obesity can be obtained by investigating changes to satiation and satiety mechanisms during the initial ∼2 weeks of HFD exposure. What is already known about this subject? Obesity is associated with dysregulated homeostatic mechanisms.Increased caloric consumption contributes to obesity.Obese rodents tend to eat larger, more frequent meals. What are the new findings in your manuscript? Exposure to obesogenic diets promotes transient attempts to maintain weight homeostasis.After ∼2 weeks, caloric hyperphagia exceeds increased energy expenditure, promoting weight gain.This is associated with consumption of larger, more frequent meals. How might your results change the direction of research or the focus of clinical practice? Our findings suggest that molecular studies focusing on mechanisms that regulate meal size and frequency, particularly those engaged during the first ∼2 weeks of obesogenic diet feeding that eventually fail, can provide unique insight into the etiology of obesity.
Collapse
|
17
|
Iyoshi A, Makura Y, Tanaka M, Ueda A. Stereocontrolled synthesis of α-d-allulofuranosides using α-selective d-fructofuranosidation reaction. Carbohydr Res 2024; 536:109044. [PMID: 38325068 DOI: 10.1016/j.carres.2024.109044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/09/2024]
Abstract
Stereocontrolled synthesis of rare sugar derivatives, namely α-d-allulofuranosides, was achieved using d-fructose, one of the most abundant carbohydrates in nature. The following are the key steps of the α-d-allulofuranosides' synthesis. (1) An α-selective glycosidation reaction of 1,3,4,6-tetra-O-benzoylated d-fructofuranosyl donor to obtain α-d-fructofuranosides with 98 %-75 % isolated yields. (2) A regioselective 1,4,6-tri-O-pivaloylation reaction of the tetraol of α-d-fructofuranosides with the C3-hydroxy group remaining intact. (3) The oxidation of the C3-hydroxy group followed by the stereoselective reduction of the C3-carbonyl group. Primary and secondary alcohols and sugars can be used as glycosyl acceptors and aglycones for the following pivaloylation and stereoinversion reactions to obtain α-d-allulofuranosides.
Collapse
Affiliation(s)
- Akihiro Iyoshi
- Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan
| | - Yui Makura
- Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan
| | - Masakazu Tanaka
- Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan
| | - Atsushi Ueda
- Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan.
| |
Collapse
|
18
|
Li RJW, Barros DR, Kuah R, Lim YM, Gao A, Beaudry JL, Zhang SY, Lam TKT. Small intestinal CaSR-dependent and CaSR-independent protein sensing regulates feeding and glucose tolerance in rats. Nat Metab 2024; 6:39-49. [PMID: 38167726 DOI: 10.1038/s42255-023-00942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024]
Abstract
Proteins activate small intestinal calcium sensing receptor (CaSR) and/or peptide transporter 1 (PepT1) to increase hormone secretion1-8, but the effect of small intestinal protein sensing and the mechanistic potential of CaSR and/or PepT1 in feeding and glucose regulation remain inconclusive. Here we show that, in male rats, CaSR in the upper small intestine is required for casein infusion to increase glucose tolerance and GLP1 and GIP secretion, which was also dependent on PepT1 (ref. 9). PepT1, but not CaSR, is required for casein infusion to lower feeding. Upper small intestine casein sensing fails to regulate feeding, but not glucose tolerance, in high-fat-fed rats with decreased PepT1 but increased CaSR expression. In the ileum, a CaSR-dependent but PepT1-independent pathway is required for casein infusion to lower feeding and increase glucose tolerance in chow-fed rats, in parallel with increased PYY and GLP1 release, respectively. High fat decreases ileal CaSR expression and disrupts casein sensing on feeding but not on glucose control, suggesting an ileal CaSR-independent, glucose-regulatory pathway. In summary, we discover small intestinal CaSR- and PepT1-dependent and -independent protein sensing mechanisms that regulate gut hormone release, feeding and glucose tolerance. Our findings highlight the potential of targeting small intestinal CaSR and/or PepT1 to regulate feeding and glucose tolerance.
Collapse
Affiliation(s)
- Rosa J W Li
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Daniel R Barros
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Rachel Kuah
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Yu-Mi Lim
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Anna Gao
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Jacqueline L Beaudry
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Song-Yang Zhang
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - Tony K T Lam
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Kishida K, Iida T, Yamada T, Toyoda Y. Intestinal absorption of D-fructose isomers, D-allulose, D-sorbose and D-tagatose, via glucose transporter type 5 (GLUT5) but not sodium-dependent glucose cotransporter 1 (SGLT1) in rats. Br J Nutr 2023; 130:1852-1858. [PMID: 38713062 DOI: 10.1017/s0007114523001113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
D-allulose, D-sorbose and D-tagatose are D-fructose isomers that are called rare sugars. These rare sugars have been studied intensively in terms of biological production and food application as well as physiological effects. There are limited papers with regard to the transporters mediating the intestinal absorption of these rare sugars. We examined whether these rare sugars are absorbed via sodium-dependent glucose cotransporter 1 (SGLT1) as well as via GLUT type 5 (GLUT5) using rats. High-fructose diet fed rats, which express more intestinal GLUT5, exhibited significantly higher peripheral concentrations, Cmax and AUC0–180 min when D-allulose, D-sorbose and D-tagatose were orally administrated. KGA-2727, a selective SGLT1 inhibitor, did not affect the peripheral and portal vein concentrations and pharmacokinetic parameters of these rare sugars. The results suggest that D-allulose, D-sorbose and D-tagatose are likely transported via GLUT5 but not SGLT1 in rat small intestine.
Collapse
Affiliation(s)
- Kunihiro Kishida
- Department of Science and Technology on Food Safety, Kindai University, 930 Nishimitani, Kinokawa, Wakayama649-6493, Japan
| | - Tetsuo Iida
- Research and Development, Matsutani Chemical Industry Company, Limited, 5-3 Kita-Itami, Itami, Hyogo664-8508, Japan
| | - Takako Yamada
- Research and Development, Matsutani Chemical Industry Company, Limited, 5-3 Kita-Itami, Itami, Hyogo664-8508, Japan
| | - Yukiyasu Toyoda
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi468-8503, Japan
| |
Collapse
|
20
|
Gao Y, Li F, Wang Y, Chen Z, Li Z. An artificial multienzyme cascade for the whole-cell synthesis of rare ketoses from glycerol. Biotechnol Lett 2023; 45:1355-1364. [PMID: 37486554 DOI: 10.1007/s10529-023-03415-6] [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: 01/15/2023] [Revised: 05/25/2023] [Accepted: 06/10/2023] [Indexed: 07/25/2023]
Abstract
PURPOSE In our previous study, we constructed a one-pot multi-enzyme system for rare ketoses synthesis based on L-rhamnulose-1-phosphate aldolase (RhaD) from accessible glycerol in vitro. To eliminate tedious purification of enzymes, a facile Escherichia coli whole-cell cascade platform was established in this study. METHODS To enhance the conversion rate, the reaction conditions, substrate concentrations and expressions of related enzymes were extensively optimized. RESULTS The biosynthetic route for the cascade synthesis of rare ketoses in whole cells was successfully constructed and three rare ketoses including D-allulose, D-sorbose and L-fructose were produced using glycerol and D/L-glyceraldehyde (GA). Under optimized conditions, the conversion rates of rare ketoses were 85.0% and 93.0% using D-GA and L-GA as the receptor, respectively. Furthermore, alditol oxidase (AldO) was introduced to the whole-cell system to generate D-GA from glycerol, and the total production yield of D-sorbose and D-allulose was 8.2 g l-1 only from the sole carbon source glycerol. CONCLUSION This study demonstrates a feasible and cost-efficient method for rare sugars synthesis and can also be applied to the green synthesis of other value-added chemicals from glycerol.
Collapse
Affiliation(s)
- Yahui Gao
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Fen Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yulu Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zhou Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zijie Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
21
|
Bae HR, Shin SK, Han Y, Yoo JH, Kim S, Young HA, Kwon EY. D-Allulose Ameliorates Dysregulated Macrophage Function and Mitochondrial NADH Homeostasis, Mitigating Obesity-Induced Insulin Resistance. Nutrients 2023; 15:4218. [PMID: 37836502 PMCID: PMC10574141 DOI: 10.3390/nu15194218] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
D-allulose, a rare sugar, has been proposed to have potential benefits in addressing metabolic disorders such as obesity and type 2 diabetes (T2D). However, the precise mechanisms underlying these effects remain poorly understood. We aimed to elucidate the mechanisms by which D-allulose influences obesity-induced insulin resistance. We conducted gene set enrichment analysis on the liver and white adipose tissue of mice exposed to a high-fat diet (HFD) along with the white adipose tissue of individuals with obesity. Our study revealed that D-allulose effectively suppressed IFN-γ, restored chemokine signaling, and enhanced macrophage function in the livers of HFD-fed mice. This implies that D-allulose curtails liver inflammation, alleviating insulin resistance and subsequently impacting adipose tissue. Furthermore, D-allulose supplementation improved mitochondrial NADH homeostasis and translation in both the liver and white adipose tissue of HFD-fed mice. Notably, we observed decreased NADH homeostasis and mitochondrial translation in the omental tissue of insulin-resistant obese subjects compared to their insulin-sensitive counterparts. Taken together, these results suggest that supplementation with allulose improves obesity-induced insulin resistance by mitigating the disruptions in macrophage and mitochondrial function. Furthermore, our data reinforce the crucial role that mitochondrial energy expenditure plays in the development of insulin resistance triggered by obesity.
Collapse
Affiliation(s)
- Heekyong R. Bae
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Youngji Han
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ji-Hyeon Yoo
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Suntae Kim
- Omixplus, LLC., Gaithersburg, MD 20850, USA
| | - Howard A. Young
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA;
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Beautiful Aging, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
22
|
Rakhat Y, Wang L, Han W, Rustemova A, Kulzhanova N, Yamada Y, Yabe D, Seino Y, Yada T. Oral Semaglutide under Human Protocols and Doses Regulates Food Intake, Body Weight, and Glycemia in Diet-Induced Obese Mice. Nutrients 2023; 15:3765. [PMID: 37686797 PMCID: PMC10489807 DOI: 10.3390/nu15173765] [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: 08/07/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The first oral form of the glucagon-like peptide-1 receptor agonist, oral semaglutide, has recently been launched and potently controls glycemia and body weight in subjects with type 2 diabetes. This drug carries the absorption enhancer and requires specific protocols of administration. The mechanism of action of oral semaglutide is not fully understood, for which an appropriate experimental model is required. This study explores the metabolic effects of oral semaglutide in mice under human protocols and doses. Oral semaglutide was bolus and once daily injected into high-fat diet-induced obese (DIO) mice under human protocols, followed by monitoring blood glucose, food intake, and body weight. Oral semaglutide 0.23 mg/kg, a comparable human dose (14 mg) in a small volume of water under human protocols rapidly decreased blood glucose and food intake and continuously reduced food intake and weight gain for 3 days in DIO mice. At 0.7 mg/kg (42 mg), this drug was somewhat more potent. Oral semaglutide with human protocols and doses rapidly reduces blood glucose and food intake and continuously suppresses feeding and weight in DIO mice. This study establishes mice as a model suitable for analyzing the mechanism of anti-obesity/diabetes actions of oral semaglutide.
Collapse
Affiliation(s)
- Yermek Rakhat
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Lei Wang
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Wanxin Han
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Aktolkyn Rustemova
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Nazymgul Kulzhanova
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yuichiro Yamada
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka 553-0003, Japan
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka 553-0003, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu 501-1193, Japan
| | - Yutaka Seino
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka 553-0003, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (Y.R.)
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu 501-1193, Japan
| |
Collapse
|
23
|
Gao Y, Chen Z, Nakanishi H, Li Z. Highly Efficient Synthesis of Rare Sugars from Glycerol in Endotoxin-Free ClearColi by Fermentation. Foods 2023; 12:3078. [PMID: 37628077 PMCID: PMC10453619 DOI: 10.3390/foods12163078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Rare sugars possess potential applications as low-calorie sweeteners, especially for anti-obesity and anti-diabetes. In this study, a fermentation biosystem based on the "DHAP-dependent aldolases strategy" was established for D-allulose and D-sorbose production from glycerol in endotoxin-free ClearColi BL21 (DE3). Several engineering strategies were adopted to enhance rare sugar production. Firstly, the combination of different plasmids for aldO, rhaD, and yqaB expression was optimized. Then, the artificially constructed ribosomal binding site (RBS) libraries of aldO, rhaD, and yqaB genes were assembled individually and combinatorially. In addition, a peroxidase was overexpressed to eliminate the damage or toxicity from hydrogen peroxide generated by alditol oxidase (AldO). Finally, stepwise improvements in rare sugar synthesis were elevated to 15.01 g/L with a high yield of 0.75 g/g glycerol in a 3 L fermenter. This research enables the effective production of rare sugars from raw glycerol in high yields.
Collapse
Affiliation(s)
- Yahui Gao
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhou Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Hideki Nakanishi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Zijie Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
24
|
Fukunaga K, Yoshimura T, Imachi H, Kobayashi T, Saheki T, Sato S, Saheki N, Jiang W, Murao K. A Pilot Study on the Efficacy of a Diabetic Diet Containing the Rare Sugar D-Allulose in Patients with Type 2 Diabetes Mellitus: A Prospective, Randomized, Single-Blind, Crossover Study. Nutrients 2023; 15:2802. [PMID: 37375710 DOI: 10.3390/nu15122802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/12/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
High sugar consumption increases the risk of diabetes, obesity, and cardiovascular diseases. Regarding the diet of patients with diabetes, artificial sweeteners are considered a safe alternative to sugar; however, there is also a risk that artificial sweeteners exacerbate glucose metabolism. D-allulose (C-3 isomer of d-fructose), which is a rare sugar, has been reported to have antidiabetic and antiobesity effects. In this study, the efficacy of a diabetic diet containing D-allulose was investigated in patients with type 2 diabetes using an intermittently scanned continuous glucose monitoring system (isCGM). This study was a validated, prospective, single-blind, randomized, crossover comparative study. Comparison of peak postprandial blood glucose (PPG) levels after consumption of a standard diabetic diet and a diabetic diet containing 8.5 g of D-allulose was the primary endpoint. A D-allulose-containing diabetic diet improved PPG levels in type two diabetes patients compared with a strictly energy-controlled diabetic diet. The results also showed a protective effect on endogenous pancreatic insulin secretory capacity owing to reduced insulin requirement. In patients with type two diabetes mellitus, diabetic diets containing 8.5 g D-allulose were effective in improving PPG levels.
Collapse
Affiliation(s)
- Kensaku Fukunaga
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Takafumi Yoshimura
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Hitomi Imachi
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Toshihiro Kobayashi
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Takanobu Saheki
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Seisuke Sato
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Nao Saheki
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Wenyi Jiang
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| | - Koji Murao
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun 761-0793, Kagawa, Japan
| |
Collapse
|
25
|
Sclafani A, Castillo A, Carata I, Pines R, Berglas E, Joseph S, Sarker J, Nashed M, Roland M, Arzayus S, Williams N, Glendinning JI, Bodnar RJ. Conditioned preference and avoidance induced in mice by the rare sugars isomaltulose and allulose. Physiol Behav 2023; 267:114221. [PMID: 37146897 DOI: 10.1016/j.physbeh.2023.114221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Isomaltulose, a slowly digested isocaloric analog of sucrose, and allulose, a noncaloric fructose analog, are promoted as "healthful" sugar alternatives in human food products. Here we investigated the appetite and preference conditioning actions of these sugar analogs in inbred mouse strains. In brief-access lick tests (Experiment 1), C57BL/6 (B6) mice showed similar concentration dependent increases in licking for allulose and fructose, but less pronounced concentration-dependent increases in licking for isomaltulose than sucrose. In Experiment 2, B6 male were given one-bottle training with a CS+ flavor (e.g., grape) mixed with 8% isomaltulose or allulose and a CS- flavor (e.g., cherry) mixed in water followed by two-bottle CS flavor tests. The isomaltulose mice showed only a weak CS+ flavor preference but a strong preference for the sugar over water. The allulose mice strongly preferred the CS- flavor and water over the sugar. The allulose avoidance may be due to gut discomfort as reported in humans consuming high amounts of the sugar. Experiment 3 found that the preference for 8% sucrose over 8% isomaltulose could be reversed or blocked by adding different concentrations of a noncaloric sweetener mixture (sucralose + saccharin, SS) to the isomaltulose. Experiment 4 revealed that the preference of B6 or FVB/N mice for isomaltulose+0.01%SS or sucrose over 0.1%SS increased after separate experience with the sugars and SS. This indicates that isomaltulose, like sucrose, has postoral appetition effects that enhances the appetite for the sugar. In Experiments 5 and 6, the appetition actions of the two sugars were directly compared by giving mice isomaltulose+0.05%SS vs. sucrose choice tests before and after separate experience with the two sugars. In general, the initial preference the mice displayed for isomaltulose+0.05%SS was reduced or reversed after separate experience with the two sugars although some strain and sex differences were obtained. This indicates that isomaltulose has weaker postoral appetition effects than sucrose.
Collapse
|
26
|
Han W, Wang L, Ohbayashi K, Takeuchi M, O'Farrell L, Coskun T, Rakhat Y, Yabe D, Iwasaki Y, Seino Y, Yada T. Glucose-dependent insulinotropic polypeptide counteracts diet-induced obesity along with reduced feeding, elevated plasma leptin and activation of leptin-responsive and proopiomelanocortin neurons in the arcuate nucleus. Diabetes Obes Metab 2023; 25:1534-1546. [PMID: 36852745 DOI: 10.1111/dom.15001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 03/01/2023]
Abstract
AIM To clarify the effects of glucose-dependent insulinotropic polypeptide (GIP) receptor agonists (GIPRAs) on feeding and body weight. MATERIALS AND METHODS Acute and subchronic effects of subcutaneous GIPFA-085, a long-acting GIPRA, on blood glucose, food intake, body weight, respiratory exchange ratio and plasma leptin levels were measured in diet-induced obese (DIO) mice and/or functional leptin-deficient ob/ob mice. The effects of GIPFA-085 on the hypothalamic arcuate nucleus (ARC) neurons from lean and DIO mice were studied by measuring cytosolic Ca2+ concentration ([Ca2+ ]i ). RESULTS Single bolus GIPFA-085 (30, 300 nmol/kg) dose-dependently reduced blood glucose in glucose tolerance tests, elevated plasma leptin levels at 0.5-6 hours and inhibited food intake at 2-24 hours after injection in DIO mice. Daily GIPFA-085 (300 nmol/kg) inhibited food intake and increased fat utilization on day 1, and reduced body weight gain on days 3-12 of treatment in DIO, but not ob/ob, mice. GIPFA-085 increased [Ca2+ ]i in the ARC leptin-responsive and proopiomelanocortin (POMC) neurons. GIPFA-085 and leptin cooperated to increase [Ca2+ ]i in ARC neurons and inhibit food intake. CONCLUSIONS GIPFA-085 acutely inhibits feeding and increases lipid utilization, and sustainedly lowers body weight in DIO mice via mechanisms involving rises in leptin and activation of ARC leptin-responsive and POMC neurons. This study highlights the therapeutic potential of GIPRAs for treating obesity and diabetes.
Collapse
Affiliation(s)
- Wanxin Han
- Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto, Japan
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
- Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Lei Wang
- Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto, Japan
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
- Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kento Ohbayashi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | | | | | | | - Yermek Rakhat
- Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto, Japan
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
- Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Hospital, Osaka, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Yutaka Seino
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Hospital, Osaka, Japan
| | - Toshihiko Yada
- Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kyoto, Japan
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
- Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
27
|
Kohara Y, Ikai S, Yoshihara A, Murao K, Sugiyama Y. Effect of chronic exposure to ketohexoses on pancreatic β-cell function in INS-1 rat insulinoma cells. Biosci Biotechnol Biochem 2023; 87:163-170. [PMID: 36413460 DOI: 10.1093/bbb/zbac190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Glucotoxicity, impaired insulin secretion, suppression of insulin gene expression, and apoptosis, in pancreatic β-cells caused by chronic hyperglycemia is a key component of the pathogenesis of type 2 diabetes. Recently, it has been reported that rare sugar d-allulose has antihyperglycemic and antihyperlipidemic effects in diabetic rats. However, the direct effects of rare sugars including d-allulose on pancreatic β-cell function are unclear. In this study, we investigated whether chronic exposure to ketohexoses causes glucotoxicity, suppression of insulin gene expression, and apoptosis, in INS-1 rat pancreatic insulinoma cells. d-Fructose, d-tagatose, l-allulose, and l-sorbose treatment for 1-week reduced insulin gene expression, whereas d-allulose, d-sorbose, l-fructose, and l-tagatose did not. All ketohexoses were transported into INS-1 cells, but were not metabolized. In addition, the ketohexoses did not induce apoptosis and did not affect glucose metabolism. These results suggest that long-term administration of d-allulose, d-sorbose, l-fructose, and l-tagatose does not affect pancreatic β-cell function.
Collapse
Affiliation(s)
- Yuri Kohara
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Miki, Kagawa, Japan
| | - Shuta Ikai
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Miki, Kagawa, Japan
| | - Akihide Yoshihara
- International Institute of Rare Sugar Research and Education, Kagawa University, Miki, Kagawa, Japan
| | - Koji Murao
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, Miki, Kagawa, Japan
| | - Yasunori Sugiyama
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Miki, Kagawa, Japan
| |
Collapse
|
28
|
Li Z, Feng L, Chen Z, Hu Y, Fei K, Xu H, Gao XD. Efficient enzymatic synthesis of d-allulose using a novel d-allulose-3-epimerase from Caballeronia insecticola. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:339-348. [PMID: 35871484 DOI: 10.1002/jsfa.12147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Rare sugars have become promising 'sugar alternatives' because of their low calories and unique physiological functions. Among the family of rare sugars, d-allulose is one of the sugars attracting interest. Ketose 3-epimerases (KEase), including d-tagatose 3-epimerase (DTEase) and d-allulose 3-epimerase (DAEase), are mainly used for d-allulose production. RESULTS In this study, a putative xylose isomerase from Caballeronia insecticola was characterized and identified as a novel DAEase. Caballeronia insecticola DAEase displayed prominent enzymatic properties, and 150 g L-1 d-allulose was produced from 500 g L-1 d-fructose in 45 min with a conversion rate of 30% and high productivity of 200 g L-1 h-1 . Furthermore, DAEase was employed in a phosphorylation-dephosphorylation cascade reaction, which significantly increased the conversion rate of d-allulose. Under optimized conditions, the conversion rate of d-allulose was approximately 100% when the concentration of d-fructose was 50 mmol L-1 . CONCLUSION This research described a very beneficial and facile approach for d-allulose production based on C. insecticola DAEase. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zijie Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Linxue Feng
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Zhou Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Yangfan Hu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Kangqing Fei
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Huilin Xu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiao-Dong Gao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
29
|
Medak KD, Weber AJ, Shamshoum H, McKie GL, Hahn MK, Wright DC. Enhancing endogenous levels of GLP1 dampens acute olanzapine induced perturbations in lipid and glucose metabolism. Front Pharmacol 2023; 14:1127634. [PMID: 36937886 PMCID: PMC10014622 DOI: 10.3389/fphar.2023.1127634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/17/2023] [Indexed: 03/05/2023] Open
Abstract
Olanzapine is a second-generation antipsychotic (SGA) used in the treatment of schizophrenia and several on- and off-label conditions. While effective in reducing psychoses, acute olanzapine treatment causes rapid hyperglycemia, insulin resistance, and dyslipidemia and these perturbations are linked to an increased risk of developing cardiometabolic disease. Pharmacological agonists of the glucagon-like peptide-1 (GLP1) receptor have been shown to offset weight-gain associated with chronic SGA administration and mitigate the acute metabolic side effects of SGAs. The purpose of this study was to determine if increasing endogenous GLP1 is sufficient to protect against acute olanzapine-induced impairments in glucose and lipid homeostasis. Male C57BL/6J mice were treated with olanzapine, in the absence or presence of an oral glucose tolerance test (OGTT), and a combination of compounds to increase endogenous GLP1. These include the non-nutritive sweetener allulose which acts to induce GLP1 secretion but not other incretins, the DPPiv inhibitor sitagliptin which prevents degradation of active GLP1, and an SSTR5 antagonist which relieves inhibition on GLP1 secretion. We hypothesized that this cocktail of agents would increase circulating GLP1 to supraphysiological concentrations and would protect against olanzapine-induced perturbations in glucose and lipid homeostasis. We found that 'triple treatment' increased both active and total GLP1 and protected against olanzapine-induced perturbations in lipid and glucose metabolism under glucose stimulated conditions and this was paralleled by an attenuation in the olanzapine induced increase in the glucagon:insulin ratio. Our findings provide evidence that pharmacological approaches to increase endogenous GLP1 could be a useful adjunct approach to reduce acute olanzapine-induced perturbations in lipid and glucose metabolism.
Collapse
Affiliation(s)
- Kyle D. Medak
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, ON, Canada
| | - Alyssa J. Weber
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, ON, Canada
| | - Hesham Shamshoum
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, ON, Canada
| | - Greg L. McKie
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, ON, Canada
| | - Margaret K. Hahn
- Centre for Addition and Mental Health, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - David C. Wright
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
- Faculty of Food and Land Systems, University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- *Correspondence: David C. Wright,
| |
Collapse
|
30
|
Hironao KY, Ashida H, Yamashita Y. Black soybean seed coat polyphenol ameliorates the abnormal feeding pattern induced by high-fat diet consumption. Front Nutr 2022; 9:1006132. [PMID: 36299984 PMCID: PMC9589235 DOI: 10.3389/fnut.2022.1006132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/21/2022] [Indexed: 12/04/2022] Open
Abstract
High-fat diet (HFD) consumption induces chronic inflammation and microglial accumulation in the mediobasal hypothalamus (MBH), the central regulator of feeding behavior and peripheral metabolism. As a result, the diurnal feeding rhythm is disrupted, leading to the development of obesity. Diet-induced obesity (DIO) can be prevented by restoring the normal feeding pattern. Therefore, functional foods and drugs that ameliorate hypothalamic inflammation and restore the normal feeding pattern may prevent or ameliorate DIO. Numerous functional foods and food-derived compounds with anti-obesity effects have been identified; however, few studies have been performed that assessed their potential to prevent the HFD-induced hypothalamic inflammation and disruption of feeding rhythm. In the present study, we found that polyphenols derived from black soybean seed coat (BE) significantly ameliorated the accumulation of activated microglia and pro-inflammatory cytokine expression in the arcuate nucleus of the hypothalamus of HFD-fed mice, and restored their feeding pattern to one comparable to that of standard diet-fed mice, thereby ameliorating DIO. Furthermore, cyanidin 3-O-glucoside—the principal anthocyanin in BE—was found to be a strong candidate mediator of these effects. This is the first study to show that BE has the potential to provide a variety of beneficial effects on health, which involve amelioration of the HFD-induced hypothalamic inflammation and abnormal feeding pattern. The results of this study provide new evidence for the anti-obesity effects of black soybean polyphenols.
Collapse
|
31
|
Kong H, Yu L, Li C, Ban X, Gu Z, Li Z. Short-Clustered Maltodextrin Activates Ileal Glucose-Sensing and Induces Glucagon-like Peptide 1 Secretion to Ameliorate Glucose Homeostasis in Type 2 Diabetic Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:12604-12619. [PMID: 36125960 DOI: 10.1021/acs.jafc.2c04978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Reconstructing molecular structure is an effective approach to attenuating glycemic response to starch. Previously, we rearranged α-1,4 and α-1,6-glycosidic bonds in starch molecules to produce short-clustered maltodextrin (SCMD). The present study revealed that SCMD slowly released glucose until the distal ileum. The activated ileal glucose-sensing enabled SCMD to be a potent inducer for glucagon-like peptide-1 (GLP-1). Furthermore, SCMD was found feasible to serve as the dominant dietary carbohydrate to rescue mice from diabetes. Interestingly, a mixture of normal maltodextrin and resistant dextrin (MD+RD), although it caused an attenuated glycemic response similar to that of SCMD, failed to ameliorate glucose homeostasis because it hardly induced GLP-1 secretion. The serum GLP-1 levels seen in MD+RD-fed mice (5.25 ± 1.51 pmol/L) were significantly lower than those seen in SCMD-fed mice (8.25 ± 2.01 pmol/L, p < 0.05). Further investigation revealed that the beneficial effects of SCMD could be abolished by a GLP-1 receptor (GLP-1R) antagonist. These results identify GLP-1R signaling as a critical contributor to SCMD-exerted health benefits and highlight the role of ileal glucose-sensing in designing dietary carbohydrates.
Collapse
Affiliation(s)
- Haocun Kong
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Luxi Yu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Caiming Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Xiaofeng Ban
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Zhaofeng Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
32
|
Central and peripheral regulations mediated by short-chain fatty acids on energy homeostasis. Transl Res 2022; 248:128-150. [PMID: 35688319 DOI: 10.1016/j.trsl.2022.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
The human gut microbiota influences obesity, insulin resistance, and the subsequent development of type 2 diabetes (T2D). The gut microbiota digests and ferments nutrients resulting in the production of short-chain fatty acids (SCFAs), which generate various beneficial metabolic effects on energy and glucose homeostasis. However, their roles in the central nervous system (CNS)-mediated outputs on the metabolism have only been minimally studied. Here, we explore what is known and future directions that may be worth exploring in this emerging area. Specifically, we searched studies or data in English by using PubMed, Google Scholar, and the Human Metabolome Database. Studies were filtered by time from 1978 to March 2022. As a result, 195 studies, 53 reviews, 1 website, and 1 book were included. One hundred and sixty-five of 195 studies describe the production and metabolism of SCFAs or the effects of SCFAs on energy homeostasis, glucose balance, and mental diseases through the gut-brain axis or directly by a central pathway. Thirty of 195 studies show that inappropriate metabolism and excessive of SCFAs are metabolically detrimental. Most studies suggest that SCFAs exert beneficial metabolic effects by acting as the energy substrate in the TCA cycle, regulating the hormones related to satiety regulation and insulin secretion, and modulating immune cells and microglia. These functions have been linked with AMPK signaling, GPCRs-dependent pathways, and inhibition of histone deacetylases (HDACs). However, the studies focusing on the central effects of SCFAs are still limited. The mechanisms by which central SCFAs regulate appetite, energy expenditure, and blood glucose during different physiological conditions warrant further investigation.
Collapse
|
33
|
Comparative Effects of Allulose, Fructose, and Glucose on the Small Intestine. Nutrients 2022; 14:nu14153230. [PMID: 35956407 PMCID: PMC9370476 DOI: 10.3390/nu14153230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Despite numerous studies on the health benefits of the rare sugar allulose, its effects on intestinal mucosal morphology and function are unclear. We therefore first determined its acute effects on the small intestinal transcriptome using DNA microarray analysis following intestinal allulose, fructose and glucose perfusion in rats. Expression levels of about 8-fold more genes were altered by allulose compared to fructose and glucose perfusion, suggesting a much greater impact on the intestinal transcriptome. Subsequent pathway analysis indicated that nutrient transport, metabolism, and digestive system development were markedly upregulated, suggesting allulose may acutely stimulate these functions. We then evaluated whether allulose can restore rat small intestinal structure and function when ingested orally following total parenteral nutrition (TPN). We also monitored allulose effects on blood levels of glucagon-like peptides (GLP) 1 and 2 in TPN rats and normal mice. Expression levels of fatty acid binding and gut barrier proteins were reduced by TPN but rescued by allulose ingestion, and paralleled GLP-2 secretion potentially acting as the mechanism mediating the rescue effect. Thus, allulose can potentially enhance disrupted gut mucosal barriers as it can more extensively modulate the intestinal transcriptome relative to glucose and fructose considered risk factors of metabolic disease.
Collapse
|
34
|
Lei P, Chen H, Ma J, Fang Y, Qu L, Yang Q, Peng B, Zhang X, Jin L, Sun D. Research progress on extraction technology and biomedical function of natural sugar substitutes. Front Nutr 2022; 9:952147. [PMID: 36034890 PMCID: PMC9414081 DOI: 10.3389/fnut.2022.952147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Improved human material living standards have resulted in a continuous increase in the rate of obesity caused by excessive sugar intake. Consequently, the number of diabetic patients has skyrocketed, not only resulting in a global health problem but also causing huge medical pressure on the government. Limiting sugar intake is a serious problem in many countries worldwide. To this end, the market for sugar substitute products, such as artificial sweeteners and natural sugar substitutes (NSS), has begun to rapidly grow. In contrast to controversial artificial sweeteners, NSS, which are linked to health concepts, have received particular attention. This review focuses on the extraction technology and biomedical function of NSS, with a view of generating insights to improve extraction for its large-scale application. Further, we highlight research progress in the use of NSS as food for special medical purpose (FSMP) for patients.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Haojie Chen
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Yimen Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Linkai Qu
- College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Bo Peng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Xingxing Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| |
Collapse
|
35
|
d-Allulose Inhibits Ghrelin-Responsive, Glucose-Sensitive and Neuropeptide Y Neurons in the Arcuate Nucleus and Central Injection Suppresses Appetite-Associated Food Intake in Mice. Nutrients 2022; 14:nu14153117. [PMID: 35956293 PMCID: PMC9370451 DOI: 10.3390/nu14153117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 12/18/2022] Open
Abstract
d-allulose, a rare sugar, has sweetness with few calories. d-allulose regulates feeding and glycemia, and ameliorates hyperphagia, obesity and diabetes. All these functions involve the central nervous system. However, central mechanisms underlying these effects of d-allulose remain unknown. We recently reported that d-allulose activates the anorexigenic neurons in the hypothalamic arcuate nucleus (ARC), the neurons that respond to glucagon-like peptide-1 and that express proopiomelanocortin. However, its action on the orexigenic neurons remains unknown. This study investigated the effects of d-allulose on the ARC neurons implicated in hunger, by measuring cytosolic Ca2+ concentration ([Ca2+]i) in single neurons. d-allulose depressed the increases in [Ca2+]i induced by ghrelin and by low glucose in ARC neurons and inhibited spontaneous oscillatory [Ca2+]i increases in neuropeptide Y (NPY) neurons. d-allulose inhibited 10 of 35 (28%) ghrelin-responsive, 18 of 60 (30%) glucose-sensitive and 3 of 8 (37.5%) NPY neurons in ARC. Intracerebroventricular injection of d-allulose inhibited food intake at 20:00 and 22:00, the early dark phase when hunger is promoted. These results indicate that d-allulose suppresses hunger-associated feeding and inhibits hunger-promoting neurons in ARC. These central actions of d-allulose represent the potential of d-allulose to inhibit the hyperphagia with excessive appetite, thereby counteracting obesity and diabetes.
Collapse
|
36
|
Abstract
D-Allulose, also referred to as psicose, is a C3-epimer of D-fructose used as a sugar substitute in low energy products. It can be formed naturally during processing of food and drinks containing sucrose and fructose or is prepared by chemical synthesis or via enzymatic treatment with epimerases from fructose. Estimated intakes via Western style diets including sweetened beverages are below 500 mg per d but, when used as a sugar replacement, intake may reach 10 to 30 g per d depending on the food consumed. Due to its structural similarity with fructose, allulose uses the same transport and distribution pathways. But in contrast to fructose, the human genome does not encode for enzymes that are able to metabolise allulose leading to an almost complete renal excretion of the absorbed dose and near-to-zero energetic yield. However, in vitro studies have shown that certain bacteria such as Klebsiella pneumonia are able to utilise allulose as a substrate. This finding has been a subject of concern, since Klebsiella pneumoniae represents an opportunistic human pathogen. It therefore raised the question of whether a high dietary intake of allulose may cause an undesirable growth advantage for potentially harmful bacteria at mucosal sites such as the intestine or at systemic sites following invasive infection. In this brief review, we discuss the current state of science on these issues and define the research needs to better understand the fate of allulose and its metabolic and microbiological effects when ingested as a sugar substitute.
Collapse
|
37
|
D-Allulose cooperates with glucagon-like peptide-1 and activates proopiomelanocortin neurons in the arcuate nucleus and central injection inhibits feeding in mice. Biochem Biophys Res Commun 2022; 613:159-165. [PMID: 35561584 DOI: 10.1016/j.bbrc.2022.04.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/19/2022] [Accepted: 04/06/2022] [Indexed: 11/21/2022]
Abstract
A rare sugar D-Allulose has sweetness without calorie. Previous studies have shown that D-Allulose improves glucose and energy metabolism and ameliorates obesity. However, underlying mechanisms remain elusive. This study explored the effect of central injection of D-Allulose on feeding behavior in mice. We also examined direct effects of D-Allulose on the neurons in the hypothalamic arcuate nucleus (ARC) that regulate feeding, including the anorexigenic glucagon-like peptide-1 (GLP-1)-responsive neurons and proopiomelanocortin (POMC) neurons. Single neurons were isolated from ARC and cytosolic Ca2+ concentration ([Ca2+]i) was measured by fura-2 microfluorometry. Administration of D-Allulose at 5.6, 16.7 and 56 mM concentration-dependently increased [Ca2+]i in ARC neurons. The [Ca2+]i increases took place similarly when the osmolarity of superfusion solution was kept constant. The majority (40%) of the D-Allulose-responsive neurons also responded to GLP-1 with [Ca2+]i increases. D-Allulose increased [Ca2+]i in 33% of POMC neurons in ARC. D-Allulose potentiated the GLP-1 action to increase [Ca2+]i in ARC neurons including POMC neurons. Intracerebroventricular injection of D-Allulose significantly decreased food intake at 1 and 2 h after injection. These results demonstrate that D-Allulose cooperates with glucagon-like peptide-1 and activates the ARC neurons including POMC neurons. Furthermore, central injection of D-Allulose inhibits feeding. These central actions of D-Allulose may underlie the ability of D-Allulose to counteract obesity and diabetes.
Collapse
|
38
|
Khan I, Kumar R, Prasad M, Srivastav RK, Vishwakarma VK, Akhtar J. Co-Adjuvancy of Solasodine & CoQ10 Against High Fat Diet-Induced Insulin Resistance Rats Via Modulating IRS-I and PPAR-γ Proteins Expression. Drug Res (Stuttg) 2022; 72:327-335. [PMID: 35724671 DOI: 10.1055/a-1806-1366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Insulin resistance (IR) is a condition in which target cells become insensitive to normal insulin concentrations in order to deliver glucose. The goal of this study was to see if solasodine combined with coenzyme Q10 could help rats with insulin resistance caused by a high-fat diet (HFD) by regulating the expression of IRS-I and PPAR-γ proteins.One of the six groups (n=6) got a conventional diet for 16 weeks as a control (normal), the HFD was given to the other five groups for 16 weeks, which further classified as-one group as HFD control while others treated with pioglitazone (10 mg/kg), coenzyme Q10 (50 mg/kg), solasodine (50 mg/kg) and combination of solasodine and coenzyme Q10i.e. SDQ10 (total 50 mg/kg) for the last 4 weeks orally once daily. Blood and tissue samples were collected by the end of study period for the biochemical and histological studies. As a result, HFD fed rats exhibited a significant increase in food and energy intake, body mass index, kidney and pancreas weight, fasting glucose, glycosylated haemoglobin, insulin level, liver enzyme ALT and AST and decrease antioxidant activity of superoxide dismutase and catalase. HFD received animals also produced a lower level of p-IRS1 and PPAR-y protein expression in western blot analysis. SDQ10 in combination successfully restored the above-mentioned complexity of insulin resistance caused by aHFD. Besides, increasesthe antioxidant activity of superoxide dismutase and catalase and normalized the architecture of kidney, pancreas and adipose tissue as well astreatment with SDQ10 raised the level of p-IRS1 and PPAR-y protein in liver tissue. As a result, supplementing with solasodine and coenzyme Q10 reversed the effect of the HFD on p-IRS1 and PPAR-y protein in liver tissue while also alleviating insulin resistance symptoms.
Collapse
Affiliation(s)
- Irfan Khan
- Faculty of Pharmacy, Integral University, Lucknow, U.P, India
| | - Rajesh Kumar
- Faculty of Pharmacy, Kamla Nehru Institute of Management & Technology, Sultanpur, U.P., India
| | - Mahesh Prasad
- Faculty of Pharmacy, Kamla Nehru Institute of Management & Technology, Sultanpur, U.P., India.,Faculty of Pharmacy, Integral University, Lucknow, U.P, India
| | - Ritesh Kumar Srivastav
- Faculty of Pharmacy, Kamla Nehru Institute of Management & Technology, Sultanpur, U.P., India
| | | | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, U.P, India
| |
Collapse
|
39
|
Japar S, Fukunaga K, Kobayashi T, Imachi H, Sato S, Saheki T, Ibata T, Yoshimura T, Soh KL, Ong SL, Muhamed Z, Murao K. A pilot study on the effect of D-allulose on postprandial glucose levels in patients with type 2 diabetes mellitus during Ramadan fasting. Diabetol Metab Syndr 2022; 14:86. [PMID: 35729673 PMCID: PMC9209837 DOI: 10.1186/s13098-022-00856-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND During Ramadan fasting, postprandial hyperglycemia is commonly observed after iftar (break of fast at sunset) meal. D-allulose is a rare sugar and is reported to have several health benefits, including the suppression of increase in postprandial glucose levels. This study investigates whether D-allulose (a C-3 epimer of D-fructose) improves the postprandial glucose in patients with type 2 diabetes mellitus (T2DM) during Ramadan. METHODS This was a pilot, prospective single-arm study design that was conducted for 10 consecutive days; 5 days of control and 5 days of consumption. The primary outcome was postprandial peak glucose levels. During the consumption period, 8.5 g of D-allulose was consumed by the participants before iftar meal. Postprandial glucose was measured using a continuous glucose monitoring system. RESULTS A total of 12 participants completed the study. Significant lower (p < 0.01) postprandial glucose values and the glucose incremental area under the curve (iAUC) were observed from 0 to 180 min during the consumption period compared to the control period. The consumption period demonstrated significantly higher percentages of time in which glucose values were found in the target range (p = 0.0032), and when the glucose levels above the target range were reduced (p = 0.0015). CONCLUSIONS The supplementation with D-allulose has the potential to improve postprandial hyperglycemia in patients with T2DM after iftar during Ramadan. Further studies are needed to confirm these findings. Trial registration ClinicalTrials.gov NCT05071950. Retrospectively registered, 8 October 2021.
Collapse
Affiliation(s)
- Salimah Japar
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan.
- Department of Nursing, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Kensaku Fukunaga
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Toshihiro Kobayashi
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Hitomi Imachi
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Seisuke Sato
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Takanobu Saheki
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Tomohiro Ibata
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Takafumi Yoshimura
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Kim Lam Soh
- Department of Nursing, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Swee Leong Ong
- School of Nursing Science, Faculty of Medicine, Universiti Sultan Zainal Abidin, Kampung Gong Badak, 21300, Terengganu, Malaysia
| | - Zamri Muhamed
- Department of Medicine, Hospital Sultanah Nur Zahirah, Ministry of Health Malaysia, 20400, Kuala Terengganu, Malaysia
| | - Koji Murao
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| |
Collapse
|
40
|
Dietary Gamma-Aminobutyric Acid (GABA) Induces Satiation by Enhancing the Postprandial Activation of Vagal Afferent Nerves. Nutrients 2022; 14:nu14122492. [PMID: 35745222 PMCID: PMC9227210 DOI: 10.3390/nu14122492] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/06/2022] [Accepted: 06/13/2022] [Indexed: 12/07/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is present in the mammalian brain as the main inhibitory neurotransmitter and in foods. It is widely used as a supplement that regulates brain function through stress-reducing and sleep-enhancing effects. However, its underlying mechanisms remain poorly understood, as it is reportedly unable to cross the blood–brain barrier. Here, we explored whether a single peroral administration of GABA affects feeding behavior as an evaluation of brain function and the involvement of vagal afferent nerves. Peroral GABA at 20 and 200 mg/kg immediately before refeeding suppressed short-term food intake without aversive behaviors in mice. However, GABA administration 30 min before refeeding demonstrated no effects. A rise in circulating GABA concentrations by the peroral administration of 200 mg/kg GABA was similar to that by the intraperitoneal injection of 20 mg/kg GABA, which did not alter feeding. The feeding suppression by peroral GABA was blunted by the denervation of vagal afferents. Unexpectedly, peroral GABA alone did not alter vagal afferent activities histologically. The coadministration of a liquid diet and GABA potentiated the postprandial activation of vagal afferents, thereby enhancing postprandial satiation. In conclusion, dietary GABA activates vagal afferents in collaboration with meals or meal-evoked factors and regulates brain function including feeding behavior.
Collapse
|
41
|
Chen Z, Gao XD, Li Z. Recent Advances Regarding the Physiological Functions and Biosynthesis of D-Allulose. Front Microbiol 2022; 13:881037. [PMID: 35495640 PMCID: PMC9048046 DOI: 10.3389/fmicb.2022.881037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/11/2022] [Indexed: 01/11/2023] Open
Abstract
D-Allulose, a generally regarded as safe (GRAS) sugar, is rare in nature. It is among the most promising sweeteners for future use due to its low caloric content, sucrose-like taste, and unique functions. D-Allulose has many physiological effects, such as antiobesity, antihyperglycemia, antidiabetes, anti-inflammatory, antioxidant, and neuroprotective effects. Therefore, D-allulose has important application value in the food, pharmaceutical, and healthcare industries. However, the high cost of D-allulose production limits its large-scale application. Currently, biotransformation is very attractive for D-allulose synthesis, with the two main methods of biosynthesis being the Izumoring strategy and the DHAP-dependent aldolase strategy. This article reviews recent advances regarding the physiological functions and biosynthesis of D-allulose. In addition, future perspectives on the production of D-allulose are presented.
Collapse
Affiliation(s)
- Zhou Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiao-Dong Gao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Zijie Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
42
|
Esmaeili M, Ajami M, Barati M, Javanmardi F, Houshiarrad A, Mousavi Khaneghah A. The significance and potential of functional food ingredients for control appetite and food intake. Food Sci Nutr 2022; 10:1602-1612. [PMID: 35592279 PMCID: PMC9094468 DOI: 10.1002/fsn3.2783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/23/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022] Open
Abstract
Dramatically rising global levels of obesity have raised consumers’ commercial and public health interest in foods that may help control appetite and weight. The satiety cascade consists of sensory, cognitive, physical, and hormonal events following food intake, preventing overeating, and the desire to eat for a long time. Functional foods can be one of the most influential factors in reducing appetite as long as effective ingredients, such as fiber and protein, are used to design these products. Also, functional foods should be designed to reduce appetite at different levels of oral processing, stomach, small intestine, and large intestine by various mechanisms. Therefore, the satiety power of functional foods depends on the type of ingredients and their amount. Because each compound has a different mechanism of action, it is recommended to use different compounds to influence satiety in functional foods.
Collapse
Affiliation(s)
- Mina Esmaeili
- Department of Nutrition Research National Nutrition and Food Technology Research Institute/School of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Marjan Ajami
- Department of Food and Nutrition Policy and Planning National Nutrition and Food Technology Research Institute School of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Meisam Barati
- Department of Cellular and Molecular Nutrition Faculty of Nutrition and Food Technology National Nutrition and Food Technology Research Institute Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Fardin Javanmardi
- Department of Food Science and Technology National Nutrition and Food Technology Research Institute/School of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Anahita Houshiarrad
- Department of Nutrition Research National Nutrition and Food Technology Research Institute/School of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Amin Mousavi Khaneghah
- Department of Food Science and Nutrition Faculty of Food Engineering University of Campinas (UNICAMP) Campinas Brazil
| |
Collapse
|
43
|
Chen J, Chen D, Chen Q, Xu W, Zhang W, Mu W. Computer-Aided Targeted Mutagenesis of Thermoclostridium caenicola d-Allulose 3-Epimerase for Improved Thermostability. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1943-1951. [PMID: 35107285 DOI: 10.1021/acs.jafc.1c07256] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
d-Allulose 3-epimerase (DAEase) is a key enzyme in d-allulose bioproduction. DAEase from Thermoclostridium caenicola suffers from poor thermostability, hampering its large-scale applications in industry. In this study, mutants A70P, G107P, F155Y, and D162T with increased melting point temperature (Tm) were obtained by targeted mutagenesis based on the calculation of the free energy of folding. The optimal single-point mutant G107P showed 11.08 h, 5, and 5.70 °C increases in the values of half-life (t1/2) at 60 °C, the optimum temperature (Topt), and Tm, respectively. Beneficial mutations were combined by ordered recombination mutagenesis, and the combinational mutant Var3 (G107P/F155Y/D162T/A70P) was generated with ΔTopt of 10 °C and ΔTm of 12.25 °C. Its t1/2 value at 65 °C was more than 140 times higher than that of the wild-type enzyme. Molecular dynamics simulations and homology modeling analysis indicated that the enhanced overall rigidity, increased hydrogen bonds between subunits, and redistributed surface electrostatic charges might be responsible for the improved thermostability of the mutant Var3.
Collapse
Affiliation(s)
- Jiajun Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ding Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qiuming Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Xu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Shandong Haizhibao Ocean Technology Co., Ltd, Weihai, Shandong 264333, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
44
|
Teysseire F, Bordier V, Budzinska A, Weltens N, Rehfeld JF, Holst JJ, Hartmann B, Beglinger C, Van Oudenhove L, Wölnerhanssen BK, Meyer-Gerspach AC. The Role of D-allulose and Erythritol on the Activity of the Gut Sweet Taste Receptor and Gastrointestinal Satiation Hormone Release in Humans: A Randomized, Controlled Trial. J Nutr 2022; 152:1228-1238. [PMID: 35135006 PMCID: PMC9071322 DOI: 10.1093/jn/nxac026] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/29/2021] [Accepted: 02/01/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Glucose induces the release of gastrointestinal (GI) satiation hormones, such as glucagon-like peptide 1 (GLP-1) and peptide tyrosine tyrosine (PYY), in part via the activation of the gut sweet taste receptor (T1R2/T1R3). OBJECTIVES The primary objective was to investigate the importance of T1R2/T1R3 for the release of cholecystokinin (CCK), GLP-1, and PYY in response to D-allulose and erythritol by assessing the effect of the T1R2/T1R3 antagonist lactisole on these responses and as secondary objectives to study the effect of the T1R2/T1R3 blockade on gastric emptying, appetite-related sensations, and GI symptoms. METHODS In this randomized, controlled, double-blind, crossover study, 18 participants (5 men) with a mean ± SD BMI (in kg/m2) of 21.9 ± 1.7 and aged 24 ± 4 y received an intragastric administration of 25 g D-allulose, 50 g erythritol, or tap water, with or without 450 parts per million (ppm) lactisole, respectively, in 6 different sessions. 13C-sodium acetate was added to all solutions to determine gastric emptying. At fixed time intervals, blood and breath samples were collected, and appetite-related sensations and GI symptoms were assessed. Data were analyzed with linear mixed-model analysis. RESULTS D-allulose and erythritol induced a significant release of CCK, GLP-1, and PYY compared with tap water (all PHolm < 0.0001, dz >1). Lactisole did not affect the D-allulose- and erythritol-induced release of CCK, GLP-1, and PYY (all PHolm > 0.1). Erythritol significantly delayed gastric emptying, increased fullness, and decreased prospective food consumption compared with tap water (PHolm = 0.0002, dz = -1.05; PHolm = 0.0190, dz = 0.69; and PHolm = 0.0442, dz = -0.62, respectively). CONCLUSIONS D-allulose and erythritol stimulate the secretion of GI satiation hormones in humans. Lactisole had no effect on CCK, GLP-1, and PYY release, indicating that D-allulose- and erythritol-induced GI satiation hormone release is not mediated via T1R2/T1R3 in the gut.
Collapse
Affiliation(s)
- Fabienne Teysseire
- St. Clara Research Ltd at St. Claraspital, Basel, Switzerland,Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Valentine Bordier
- St. Clara Research Ltd at St. Claraspital, Basel, Switzerland,Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Aleksandra Budzinska
- Laboratory for Brain-Gut Axis Studies, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium,Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Nathalie Weltens
- Laboratory for Brain-Gut Axis Studies, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium,Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Lukas Van Oudenhove
- Laboratory for Brain-Gut Axis Studies, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium,Leuven Brain Institute, KU Leuven, Leuven, Belgium,Cognitive and Affective Neuroscience Lab, Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Bettina K Wölnerhanssen
- St. Clara Research Ltd at St. Claraspital, Basel, Switzerland,Faculty of Medicine, University of Basel, Basel, Switzerland
| | | |
Collapse
|
45
|
Niibo M, Kanasaki A, Iida T, Ohnishi K, Ozaki T, Akimitsu K, Minamino T. d-allulose protects against diabetic nephropathy progression in Otsuka Long-Evans Tokushima Fatty rats with type 2 diabetes. PLoS One 2022; 17:e0263300. [PMID: 35100325 PMCID: PMC8803202 DOI: 10.1371/journal.pone.0263300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/17/2022] [Indexed: 11/18/2022] Open
Abstract
d-allulose is a rare sugar that has been reported to possess anti-hyperglycemic effects. In the present study, we hypothesized that d-allulose is effective in attenuating the progression of diabetic nephropathy in the Otsuka Long-Evans Tokushima Fatty (OLETF) rat model of type 2 diabetes mellitus. Drinking water with or without 3% d-allulose was administered to OLETF rats for 13 weeks. Long-Evans Tokushima Otsuka rats that received drinking water without d-allulose were used as non-diabetic control rats. d-allulose significantly attenuated the increase in blood glucose levels and progressive mesangial expansion in the glomerulus, which is regarded as a characteristic of diabetic nephropathy, in OLETF rats. d-allulose also attenuated the significant increases in renal IL-6 and tumor necrosis factor-α mRNA levels in OLETF rats, which is a proinflammatory parameter. Additionally, we showed that d-allulose suppresses mesangial matrix expansion, but its correlation with suppressing renal inflammation in OLETF rats should be investigated further. Collectively, our results support the hypothesis that d-allulose can prevent diabetic nephropathy in rats.
Collapse
Affiliation(s)
- Misato Niibo
- Research and Development, Matsutani Chemical Industry Co., Ltd, Itami City, Hyogo, Japan
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Kagawa, Japan
| | - Akane Kanasaki
- Research and Development, Matsutani Chemical Industry Co., Ltd, Itami City, Hyogo, Japan
| | - Tetsuo Iida
- Research and Development, Matsutani Chemical Industry Co., Ltd, Itami City, Hyogo, Japan
| | - Keisuke Ohnishi
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Kagawa, Japan
| | - Taro Ozaki
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Kagawa, Japan
| | - Kazuya Akimitsu
- International Institute of Rare Sugar Research and Education & Faculty of Agriculture, Kagawa University, Miki, Kagawa, Japan
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Kagawa, Japan
| |
Collapse
|
46
|
Liu B, Gou Y, Tsuzuki T, Yamada T, Iida T, Wang S, Banno R, Toyoda Y, Koike T. d-Allulose Improves Endurance and Recovery from Exhaustion in Male C57BL/6J Mice. Nutrients 2022; 14:nu14030404. [PMID: 35276765 PMCID: PMC8838150 DOI: 10.3390/nu14030404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 01/07/2023] Open
Abstract
d-Allulose, a rare sugar, improves glucose metabolism and has been proposed as a candidate calorie restriction mimetic. This study aimed to investigate the effects of d-allulose on aerobic performance and recovery from exhaustion and compared them with the effects of exercise training. Male C57BL/6J mice were subjected to exercise and allowed to run freely on a wheel. Aerobic performance was evaluated using a treadmill. Glucose metabolism was analyzed by an intraperitoneal glucose tolerance test (ipGTT). Skeletal muscle intracellular signaling was analyzed by Western blotting. Four weeks of daily oral administration of 3% d-allulose increased running distance and shortened recovery time as assessed by an endurance test. d-Allulose administration also increased the maximal aerobic speed (MAS), which was observed following treatment for >3 or 7 days. The improved performance was associated with lower blood lactate levels and increased liver glycogen levels. Although d-allulose did not change the overall glucose levels as determined by ipGTT, it decreased plasma insulin levels, indicating enhanced insulin sensitivity. Finally, d-allulose enhanced the phosphorylation of AMP-activated protein kinase and acetyl-CoA carboxylase and the expression of peroxisome proliferator-activated receptor γ coactivator 1α. Our results indicate that d-allulose administration enhances endurance ability, reduces fatigue, and improves insulin sensitivity similarly to exercise training. d-Allulose administration may be a potential treatment option to alleviate obesity and enhance aerobic exercise performance.
Collapse
Affiliation(s)
- Bingyang Liu
- Department of Sports Medicine, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Japan; (B.L.); (Y.G.); (S.W.); (R.B.)
| | - Yang Gou
- Department of Sports Medicine, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Japan; (B.L.); (Y.G.); (S.W.); (R.B.)
| | - Takamasa Tsuzuki
- Faculty of Pharmacy, Meijo University, Nagoya 468-8503, Japan; (T.T.); (Y.T.)
| | - Takako Yamada
- Research and Development, Matsutani Chemical Industry Co. Ltd., Itami 664-8508, Japan; (T.Y.); (T.I.)
| | - Tetsuo Iida
- Research and Development, Matsutani Chemical Industry Co. Ltd., Itami 664-8508, Japan; (T.Y.); (T.I.)
| | - Sixian Wang
- Department of Sports Medicine, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Japan; (B.L.); (Y.G.); (S.W.); (R.B.)
| | - Ryoichi Banno
- Department of Sports Medicine, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Japan; (B.L.); (Y.G.); (S.W.); (R.B.)
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya 464-8601, Japan
| | - Yukiyasu Toyoda
- Faculty of Pharmacy, Meijo University, Nagoya 468-8503, Japan; (T.T.); (Y.T.)
| | - Teruhiko Koike
- Department of Sports Medicine, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Japan; (B.L.); (Y.G.); (S.W.); (R.B.)
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya 464-8601, Japan
- Correspondence: ; Tel.: +81-52-789-3963
| |
Collapse
|
47
|
Haas MJ, Parekh S, Kalidas P, Richter A, Warda F, Wong NCW, Tokuda M, Mooradian AD. Insulin mimetic effect of D-allulose on apolipoprotein A-I gene. J Food Biochem 2022; 46:e14064. [PMID: 34984676 DOI: 10.1111/jfbc.14064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/23/2021] [Accepted: 12/17/2021] [Indexed: 12/07/2022]
Abstract
Several nutrients modulate the transcriptional activity of the apolipoprotein A-I (apo A-I) gene. To determine the influence of rare sugars on apo A-I expression in hepatic (HepG2) and intestinal derived (Caco-2) cell lines, apo A-I, albumin, and SP1 were quantified with enzyme immunoassay and Western blots while mRNA levels were quantified with real-time polymerase chain reaction. The promoter activity was measured using transient transfection assays with plasmids containing various segments and mutations in the promoter. D-allulose and D-tagatose, increased apo A-I concentration in culture media while D-sorbose and D-allose did not have any measurable effects. D-allulose did not increase apo A-I levels in Caco-2 cells. These changes paralleled the increased mRNA levels and promoter activity. D-allulose-response was mapped at the insulin response core element (IRCE). Mutation of the IRCE decreased the ability of D-allulose and insulin to activate the promoter. Treatment of HepG2 cells, but not Caco-2 cells, with D-alluose and insulin increased SP1 expression relative to control cells. D-allulose augmented the expression and IRCE binding of SP1, an essential transcription factor for the insulin on apo A-I promoter activity. D-allulose can modulate some insulin-responsive genes and may have anti-atherogenic properties, in part due to increasing apo A-I production. PRACTICAL APPLICATIONS: Coronary artery disease (CAD) is the number one cause of mortality in industrialized countries. A risk factor associated with CAD is low high-density lipoprotein (HDL) cholesterol and apolipoprotein A-I (apo A-I) concentrations in plasma. Thus, novel therapeutic agents or nutrients that upregulate apo A-I production should be identified. D-allulose and D-tagatose are used as sweeteners and may have favorable effects on insulin resistance and diabetes. This study shows that D-allulose and D-tagatose increases apo A-I production through increased transcription factor SP1-binding to insulin response element of the promoter. These sweeteners modulate some insulin responsive genes, increase the production of apo-A-I, and therefore may have anti-atherogenic properties.
Collapse
Affiliation(s)
- Michael J Haas
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Shrina Parekh
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Poonam Kalidas
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Angela Richter
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Firas Warda
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida College of Medicine, Jacksonville, Florida, USA
| | | | - Masaaki Tokuda
- Department of Cell Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Arshag D Mooradian
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida College of Medicine, Jacksonville, Florida, USA
| |
Collapse
|
48
|
Lee GH, Peng C, Lee HY, Park SA, Hoang TH, Kim JH, Sa S, Kim GE, Han JS, Chae HJ. D-allulose ameliorates adiposity through the AMPK-SIRT1-PGC-1α pathway in HFD-induced SD rats. Food Nutr Res 2021; 65:7803. [PMID: 35221861 PMCID: PMC8829832 DOI: 10.29219/fnr.v65.7803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/10/2021] [Accepted: 08/28/2021] [Indexed: 11/20/2022] Open
Abstract
Background Objective Design Results Conclusion
Collapse
Affiliation(s)
- Geum-Hwa Lee
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Cheng Peng
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Hwa-Young Lee
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Seon-Ah Park
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - The-Hiep Hoang
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Jung Hyun Kim
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, Jeonbuk, Republic of Korea
| | - Soonok Sa
- Samyang Corp., Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Go-Eun Kim
- Samyang Corp., Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jung-Sook Han
- Samyang Corp., Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Han-Jung Chae
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
- School of Pharmacy, Jeonbuk National University, Jeonju, Jeonbuk, Republic of Korea
- Han-Jung Chae, Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Jeonbuk 54907 Republic of Korea.
| |
Collapse
|
49
|
Matsubara Y, Kiyohara H, Teratani T, Mikami Y, Kanai T. Organ and brain crosstalk: The liver-brain axis in gastrointestinal, liver, and pancreatic diseases. Neuropharmacology 2021; 205:108915. [PMID: 34919906 DOI: 10.1016/j.neuropharm.2021.108915] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022]
Abstract
The liver is the largest organ in the human body and is responsible for the metabolism and storage of the three principal nutrients: carbohydrates, fats, and proteins. In addition, the liver contributes to the breakdown and excretion of alcohol, medicinal agents, and toxic substances and the production and secretion of bile. In addition to its role as a metabolic centre, the liver has recently attracted attention for its function in the liver-brain axis, which interacts closely with the central nervous system via the autonomic nervous system, including the vagus nerve. The liver-brain axis influences the control of eating behaviour in the central nervous system through stimuli from the liver. Conversely, neural signals from the central nervous system influence glucose, lipid, and protein metabolism in the liver. The liver also receives a constant influx of nutrients and hormones from the intestinal tract and compounds of bacterial origin via the portal system. As a result, the intestinal tract and liver are involved in various immunological interactions. A good example is the co-occurrence of primary sclerosing cholangitis and ulcerative colitis. These heterogeneous roles of the liver-brain axis are mediated via the vagus nerve in an asymmetrical manner. In this review, we provide an overview of these interactions, mainly with the liver but also with the brain and gut.
Collapse
Affiliation(s)
- Yuta Matsubara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroki Kiyohara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan.
| |
Collapse
|
50
|
Hu M, Li M, Jiang B, Zhang T. Bioproduction of D-allulose: Properties, applications, purification, and future perspectives. Compr Rev Food Sci Food Saf 2021; 20:6012-6026. [PMID: 34668314 DOI: 10.1111/1541-4337.12859] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022]
Abstract
D-allulose is the C-3 epimer of D-fructose, which rarely exists in nature, and can be biosynthesized from D-fructose by the catalysis of D-psicose 3-epimerase. D-allulose is safe for human consumption and was recently approved by the United States Food and Drug Administration for food applications. It is not only able be used in food and dietary supplements as a low-calorie sweetener, but also modulates a variety of physiological functions. D-allulose has gained increasing attention owing to its excellent properties. This article presents a review of recent progress on the properties, applications, and bioproduction progress of D-allulose.
Collapse
Affiliation(s)
- Mengying Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Mengli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Bo Jiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|