51
|
Zearalenone Exposure Affects the Keap1-Nrf2 Signaling Pathway and Glucose Nutrient Absorption Related Genes of Porcine Jejunal Epithelial Cells. Toxins (Basel) 2022; 14:toxins14110793. [PMID: 36422967 PMCID: PMC9696209 DOI: 10.3390/toxins14110793] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 11/15/2022] Open
Abstract
This study aims to examine the impact of zearalenone (ZEA) on glucose nutrient absorption and the role of the Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway in zearalenone-induced oxidative stress of porcine jejunal epithelial cells (IPEC-J2). For 24 and 36 h, the IPEC-J2 cells were exposed to ZEA at concentrations of 0, 10, 20, and 40 (Control, ZEA10, ZEA20, ZEA40) mol/L. With the increase of ZEA concentration and prolongation of the action time, the apoptosis rate and malondialdehyde level and relative expression of sodium-dependent glucose co-transporter 1 (Sglt1), glucose transporter 2 (Glut2), Nrf2, quinone oxidoreductase 1 (Nqo1), and hemeoxygenase 1 (Ho1) at mRNA and protein level, fluorescence intensity of Nrf2 and reactive oxygen species increased significantly (p < 0.05), total superoxide dismutase and glutathione peroxidase activities and relative expression of Keap1 at mRNA and protein level, fluorescence intensity of Sglt1 around the cytoplasm and the cell membrane of IPEC-J2 reduced significantly (p < 0.05). In conclusion, ZEA can impact glucose absorption by affecting the expression of Sglt1 and Glut2, and ZEA can activate the Keap1-Nrf2 signaling pathway by enhancing Nrf2, Nqo1, and Ho1 expression of IPEC-J2.
Collapse
|
52
|
Jan R, Khan M, Asaf S, Lubna, Asif S, Kim KM. Bioactivity and Therapeutic Potential of Kaempferol and Quercetin: New Insights for Plant and Human Health. PLANTS (BASEL, SWITZERLAND) 2022; 11:2623. [PMID: 36235488 PMCID: PMC9571405 DOI: 10.3390/plants11192623] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 07/25/2023]
Abstract
Plant secondary metabolites, especially flavonoids, are major metabolites widely found in plants that play several key roles in plant defence and signalling in response to stress conditions. The most studied among these flavonoids are kaempferol and quercetin due to their anti-oxidative potential and their key roles in the defence system, making them more critical for plant adaptation in stress environments. Kaempferol and quercetin in plants have great therapeutic potential for human health. Despite being well-studied, some of their functional aspects regarding plants and human health need further evaluation. This review summarizes the emerging potential of kaempferol and quercetin in terms of antimicrobial activity, bioavailability and bioactivity in the human body as well as in the regulation of plant defence in response to stresses and as a signalling molecule in terms of hormonal modulation under stress conditions. We also evaluated the safe use of both metabolites in the pharmaceutical industry.
Collapse
Affiliation(s)
- Rahmatullah Jan
- Department of Applied Biosciences, Graduate School, Kyungpook National University, Daegu 41566, Korea
- Coastal Agriculture Research Institute, Kyungpook National University, Daegu 41566, Korea
| | - Murtaza Khan
- Department of Horticulture and Life Science, Yeungnam University, Gyeongsan 38541, Korea
| | - Sajjad Asaf
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Oman
| | - Lubna
- Department of Botany, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Saleem Asif
- Department of Applied Biosciences, Graduate School, Kyungpook National University, Daegu 41566, Korea
| | - Kyung-Min Kim
- Department of Applied Biosciences, Graduate School, Kyungpook National University, Daegu 41566, Korea
- Coastal Agriculture Research Institute, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
53
|
Abstract
When it comes to food, one tempting substance is sugar. Although sweetness is detected by the tongue, the desire to consume sugar arises from the gut. Even when sweet taste is impaired, animals can distinguish sugars from non-nutritive sweeteners guided by sensory cues arising from the gut epithelium. Here, we review the molecular receptors, cells, circuits and behavioural consequences associated with sugar sensing in the gut. Recent work demonstrates that some duodenal cells, termed neuropod cells, can detect glucose using sodium-glucose co-transporter 1 and release glutamate onto vagal afferent neurons. Based on these and other data, we propose a model in which specific populations of vagal neurons relay these sensory cues to distinct sets of neurons in the brain, including neurons in the caudal nucleus of the solitary tract, dopaminergic reward circuits in the basal ganglia and homeostatic feeding circuits in the hypothalamus, that alter current and future sugar consumption. This emerging model highlights the critical role of the gut in sensing the chemical properties of ingested nutrients to guide appetitive decisions.
Collapse
Affiliation(s)
- Winston W Liu
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
54
|
Jing C, Niu J, Liu Y, Jiao N, Huang L, Jiang S, Yan L, Yang W, Li Y. Tannic Acid Extracted from Galla chinensis Supplementation in the Diet Improves Intestinal Development through Suppressing Inflammatory Responses via Blockage of NF-κB in Broiler Chickens. Animals (Basel) 2022; 12:2397. [PMID: 36139256 PMCID: PMC9495145 DOI: 10.3390/ani12182397] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/11/2022] [Accepted: 09/11/2022] [Indexed: 12/27/2022] Open
Abstract
The objective of this study was to investigate the effects of adding tannic acid (TA) extracted from Galla chinensis to the diet of broiler chickens on intestinal development. A total of 324 healthy 1-day-old broilers were used in a 42 d study, and divided into two treatment groups at random (six replicates per group). Broilers were either received a basal diet or a basal diet supplemented with 300 mg/kg microencapsulated TA extracted from Galla chinensis. The results showed that dietary supplemented with 300 mg/kg TA from Galla chinensis improved intestinal morphology, promoted intestinal mucosal barrier integrity, and elevated mucosal expressions of nutrients transporters and tight junction protein CLDN3 in broilers. Besides, 300 mg/kg TA from Galla chinensis supplementation decreased the concentrations of inflammatory cytokines in serum and intestinal mucosa and reduced the mRNA expression of NF-κB in intestinal mucosa. Above all, supplementation of 300 mg/kg microencapsulated TA extracted from Galla chinensis showed beneficial effects in improving intestinal development, which might be attributed to the suppression of inflammatory responses via blockage of NF-κB in broiler chickens. These findings will support the use of TA sourced from Galla chinensis in poultry industry.
Collapse
Affiliation(s)
- Changwei Jing
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Shizishan Street 1#, Wuhan 430070, China
| | - Jiaxing Niu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Yang Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Ning Jiao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Libo Huang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Shuzhen Jiang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Lei Yan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
- Shandong New Hope Liuhe Group Co., Ltd., Jiudongshui Road 592-26#, Qingdao 266100, China
| | - Weiren Yang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| | - Yang Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Street 61#, Tai’an 271018, China
| |
Collapse
|
55
|
Noonong K, Pranweerapaiboon K, Chaithirayanon K, Surayarn K, Ditracha P, Changklungmoa N, Kueakhai P, Hiransai P, Bunluepuech K. Antidiabetic potential of Lysiphyllum strychnifolium (Craib) A. Schmitz compounds in human intestinal epithelial Caco-2 cells and molecular docking-based approaches. BMC Complement Med Ther 2022; 22:235. [PMID: 36064352 PMCID: PMC9442914 DOI: 10.1186/s12906-022-03706-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Lysiphyllum strychnifolium (Craib) A. Schmitz, a traditional Thai medicinal plant, is mainly composed of polyphenols and flavonoids and exhibits several pharmacological activities, including antioxidant, anticancer, antimicrobial, and antidiabetic activities. However, the mechanism by which pure compounds from L. strychnifolium inhibit glucose catalysis in the small intestine and their effect on the glucose transporter remain unknown.
Methods
The objectives of this research were to examine the effect of 3,5,7-trihydroxychromone-3-O-𝛼-L-rhamnopyranoside (compound 1) and 3,5,7,3’,5’-pentahydroxy-flavanonol-3-O-𝛼-L-rhamnopyranoside (compound 2) on the inhibition of α-amylase and α-glucosidase, as well as glucose transporters, such as sodium-glucose cotransporter 1 (SGLT1), glucose transporter 2 (GLUT2), and glucose transporter 5 (GLUT5), using Caco-2 cells as a model of human intestinal epithelial cells. Additionally, the binding affinity and interaction patterns of compounds against two receptor proteins (SGLT1 and GLUT2) were determined for the first time utilizing a molecular docking approach.
Results
In the α-amylase inhibition assay, a concentration-dependent inhibitory response was observed against the enzyme. The results indicated that compound 1 inhibited α-amylase activity in a manner similar to that of acarbose (which exhibit IC50 values of 3.32 ± 0.30 µg/mL and 2.86 ± 0.10 µg/mL, respectively) in addition to a moderate inhibitory effect for compound 2 (IC50 = 10.15 ± 0.53 µg/mL). Interestingly, compounds 1 and 2 significantly inhibited α-glucosidase and exhibited better inhibition than that of acarbose, with IC50 values of 5.35 ± 1.66 µg/mL, 510.15 ± 1.46 µg/mL, and 736.93 ± 7.02 µg/mL, respectively. Additionally, α-glucosidase activity in the supernatant of the Caco-2 cell monolayer was observed. In comparison to acarbose, compounds 1 and 2 inhibited α-glucosidase activity more effectively in Caco-2 cells without cytotoxicity at a concentration of 62.5 µg/mL. Furthermore, the glucose uptake pathways mediated by SGLT1, GLUT2, and GLUT5- were downregulated in Caco-2 cells treated with compounds 1 and 2. Additionally, molecular modeling studies revealed that compounds 1 and 2 presented high binding activity with SGLT1 and GLUT2.
Conclusion
In summary, our present study was the first to perform molecular docking with compounds present in L. strychnifolium extracts. Our findings indicated that compounds 1 and 2 reduced glucose uptake in Caco-2 cells by decreasing the expression of glucose transporter genes and inhibiting the binding sites of SGLT1 and GLUT2. Therefore, compounds 1 and 2 may be used as functional foods in dietary therapy for postprandial hyperglycemia modulation of type 2 diabetes.
Collapse
|
56
|
Forouzandeh A, Blavi L, Pérez JF, D’Angelo M, González-Solé F, Monteiro A, Stein HH, Solà-Oriol D. How copper can impact pig growth: comparing the effect of copper sulfate and monovalent copper oxide on oxidative status, inflammation, gene abundance, and microbial modulation as potential mechanisms of action. J Anim Sci 2022; 100:skac224. [PMID: 35723874 PMCID: PMC9486896 DOI: 10.1093/jas/skac224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/17/2022] [Indexed: 11/12/2022] Open
Abstract
The beneficial effect of elevated concentrations of copper (Cu) on growth performance of pigs has been already demonstrated; however, their mechanism of action is not fully discovered. The objective of the present experiment was to investigate the effects of including Cu from copper sulfate (CuSO4) or monovalent copper oxide (Cu2O) in the diet of growing pigs on oxidative stress, inflammation, gene abundance, and microbial modulation. We used 120 pigs with initial body weight (BW) of 11.5 ± 0.98 kg in 2 blocks of 60 pigs, 3 dietary treatments, 5 pigs per pen, and 4 replicate pens per treatment within each block for a total of 8 pens per treatment. Dietary treatments included the negative control (NC) diet containing 20 mg Cu/kg and 2 diets in which 250 mg Cu/kg from CuSO4 or Cu2O was added to the NC. On day 28, serum samples were collected from one pig per pen and this pig was then euthanized to obtain liver samples for the analysis of oxidative stress markers (Cu/Zn superoxide dismutase, glutathione peroxidase, and malondialdehyde, MDA). Serum samples were analyzed for cytokines. Jejunum tissue and colon content were collected and used for transcriptomic analyses and microbial characterization, respectively. Results indicated that there were greater (P < 0.05) MDA levels in the liver of pigs fed the diet with 250 mg/kg CuSO4 than in pigs fed the other diets. The serum concentration of tumor necrosis factor-alpha was greater (P < 0.05) in pigs fed diets containing CuSO4 compared with pigs fed the NC diet or the diet with 250 mg Cu/kg from Cu2O. Pigs fed diets containing CuSO4 or Cu2O had a greater (P < 0.05) abundance of genes related to the intestinal barrier function and nutrient transport, but a lower (P < 0.05) abundance of pro-inflammatory genes compared with pigs fed the NC diet. Supplementing diets with CuSO4 or Cu2O also increased (P < 0.05) the abundance of Lachnospiraceae and Peptostreptococcaceae families and reduced (P < 0.05) the abundance of the Rikenellaceae family, Campylobacter, and Streptococcus genera in the colon of pigs. In conclusion, adding 250 mg/kg of Cu from CuSO4 or Cu2O regulates genes abundance in charge of the immune system and growth, and promotes changes in the intestinal microbiota; however, Cu2O induces less systemic oxidation and inflammation compared with CuSO4.
Collapse
Affiliation(s)
- Asal Forouzandeh
- Animal Nutrition and Welfare Service (SNiBA), Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laia Blavi
- Animal Nutrition and Welfare Service (SNiBA), Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jose Francisco Pérez
- Animal Nutrition and Welfare Service (SNiBA), Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Matilde D’Angelo
- Animal Nutrition and Welfare Service (SNiBA), Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Francesc González-Solé
- Animal Nutrition and Welfare Service (SNiBA), Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | - Hans H Stein
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA
| | - David Solà-Oriol
- Animal Nutrition and Welfare Service (SNiBA), Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
57
|
Glendinning JI, Williams N. Prolonged Consumption of glucose syrup enhances glucose tolerance in mice. Physiol Behav 2022; 256:113954. [PMID: 36055416 DOI: 10.1016/j.physbeh.2022.113954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 11/28/2022]
Abstract
There is debate about the metabolic impact of sugar-sweetened beverages. Here, we tested the hypothesis that ad lib consumption of glucose (Gluc) or high-fructose (HiFruc) syrups improves glucose tolerance in mice. We provided C57BL/6 mice with a control (chow and water) or experimental (chow, water and sugar solution) diet across two consecutive 28-day exposure periods, and monitored changes in body composition, glucose tolerance, cephalic-phase insulin release (CPIR) and insulin sensitivity. The sugar solutions contained 11% concentrations of Gluc or HiFruc syrup; these syrups were derived from either corn starch or cellulose. In Experiment 1, consumption of the Gluc diets reliably enhanced glucose tolerance, while consumption of the HiFruc diets did not. Mice on the Gluc diets exhibited higher CPIR (relative to baseline) by the end of exposure period 1, whereas mice on the control and HiFruc diets did not do so until the end of exposure period 2. Mice on the Gluc diets also exhibited higher insulin sensitivity than control mice at the end of exposure period 2, while mice on the HiFruc diets did not. In Experiment 2, we repeated the previous experiment, but limited testing to the corn-based Gluc and HiFruc syrups. We found, once again, that consumption of the Gluc (but not the HiFruc) diet enhanced glucose tolerance, in part by increasing CPIR and insulin sensitivity. These results show that mice can adapt metabolically to high glucose diets, and that this adaptation process involves upregulating at least two components of the insulin response system.
Collapse
Affiliation(s)
- John I Glendinning
- Departments of Biology and Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027.
| | - Niki Williams
- Departments of Biology and Neuroscience & Behavior, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027
| |
Collapse
|
58
|
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
|
59
|
Ahsan MK, dos Reis DC, Barbieri A, Sumigray KD, Nottoli T, Salas PJ, Ameen NA. Loss of Serum Glucocorticoid-Inducible Kinase 1 SGK1 Worsens Malabsorption and Diarrhea in Microvillus Inclusion Disease (MVID). J Clin Med 2022; 11:jcm11144179. [PMID: 35887942 PMCID: PMC9319011 DOI: 10.3390/jcm11144179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Microvillus inclusion disease (MVID), a lethal congenital diarrheal disease, results from loss of function mutations in the apical actin motor myosin VB (MYO5B). How loss of MYO5B leads to both malabsorption and fluid secretion is not well understood. Serum glucocorticoid-inducible kinase 1 (SGK1) regulates intestinal carbohydrate and ion transporters including cystic fibrosis transmembrane conductance regulator (CFTR). We hypothesized that loss of SGK1 could reduce CFTR fluid secretion and MVID diarrhea. Using CRISPR-Cas9 approaches, we generated R26CreER;MYO5Bf/f conditional single knockout (cMYO5BKO) and R26CreER;MYO5Bf/f;SGK1f/f double knockout (cSGK1/MYO5B-DKO) mice. Tamoxifen-treated cMYO5BKO mice resulted in characteristic features of human MVID including severe diarrhea, microvillus inclusions (MIs) in enterocytes, defective apical traffic, and depolarization of transporters. However, apical CFTR distribution was preserved in crypts and depolarized in villus enterocytes, and CFTR high expresser (CHE) cells were observed. cMYO5BKO mice displayed increased phosphorylation of SGK1, PDK1, and the PDK1 target PKCι in the intestine. Surprisingly, tamoxifen-treated cSGK1/MYO5B-DKO mice displayed more severe diarrhea than cMYO5BKO, with preservation of apical CFTR and CHE cells, greater fecal glucose and reduced SGLT1 and GLUT2 in the intestine. We conclude that loss of SGK1 worsens carbohydrate malabsorption and diarrhea in MVID.
Collapse
Affiliation(s)
- Md Kaimul Ahsan
- Department of Pediatrics, Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT 06510, USA; (M.K.A.); (D.C.d.R.)
| | - Diego Carlos dos Reis
- Department of Pediatrics, Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT 06510, USA; (M.K.A.); (D.C.d.R.)
| | - Andrea Barbieri
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Kaelyn D. Sumigray
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Timothy Nottoli
- Genome Editing Center, Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Pedro J. Salas
- Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, FL 33146, USA;
| | - Nadia A. Ameen
- Department of Pediatrics, Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT 06510, USA; (M.K.A.); (D.C.d.R.)
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Correspondence:
| |
Collapse
|
60
|
Morresi C, Vasarri M, Bellachioma L, Ferretti G, Degl′Innocenti D, Bacchetti T. Glucose Uptake and Oxidative Stress in Caco-2 Cells: Health Benefits from Posidonia oceanica (L.) Delile. Mar Drugs 2022; 20:md20070457. [PMID: 35877750 PMCID: PMC9319946 DOI: 10.3390/md20070457] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/10/2022] Open
Abstract
Posidonia oceanica (L.) Delile is an endemic Mediterranean marine plant of extreme ecological importance. Previous in vitro and in vivo studies have demonstrated the potential antidiabetic properties of P. oceanica leaf extract. Intestinal glucose transporters play a key role in glucose homeostasis and represent novel targets for the management of diabetes. In this study, the ability of a hydroalcoholic P. oceanica leaf extract (POE) to modulate intestinal glucose transporters was investigated using Caco-2 cells as a model of an intestinal barrier. The incubation of cells with POE significantly decreased glucose uptake by decreasing the GLUT2 glucose transporter levels. Moreover, POE had a positive effect on the barrier integrity by increasing the Zonulin-1 levels. A protective effect exerted by POE against oxidative stress induced by chronic exposure to high glucose concentrations or tert-butyl hydroperoxide was also demonstrated. This study highlights for the first time the effect of POE on glucose transport, intestinal barrier integrity, and its protective antioxidant effect in Caco-2 cells. These findings suggest that the P. oceanica phytocomplex may have a positive impact by preventing the intestinal cell dysfunction involved in the development of inflammation-related disease associated with oxidative stress.
Collapse
Affiliation(s)
- Camilla Morresi
- Department of Clinical Experimental Science and Odontostomatology-Biochemistry, Università Politecnica delle Marche, 60100 Ancona, Italy; (C.M.); (G.F.)
| | - Marzia Vasarri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy;
| | - Luisa Bellachioma
- Department of Life and Environmental Sciences-Biochemistry, Università Politecnica delle Marche, 60100 Ancona, Italy; (L.B.); (T.B.)
| | - Gianna Ferretti
- Department of Clinical Experimental Science and Odontostomatology-Biochemistry, Università Politecnica delle Marche, 60100 Ancona, Italy; (C.M.); (G.F.)
| | - Donatella Degl′Innocenti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy;
- Interuniversity Center of Marine Biology and Applied Ecology “G. Bacci” (CIBM), Viale N. Sauro 4, 57128 Livorno, Italy
- Correspondence:
| | - Tiziana Bacchetti
- Department of Life and Environmental Sciences-Biochemistry, Università Politecnica delle Marche, 60100 Ancona, Italy; (L.B.); (T.B.)
| |
Collapse
|
61
|
Sundaresan S, Antoun J, Banan B, Adcock J, Johnson C, Claire B, Dixon K, Flynn J, Shibao CA, Abumrad N. Botulinum Injection Into the Proximal Intestinal Wall of Diet-Induced Obese Mice Leads to Weight Loss and Improves Glucose and Fat Tolerance. Diabetes 2022; 71:1424-1438. [PMID: 35476783 PMCID: PMC9490449 DOI: 10.2337/db21-0708] [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: 08/10/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022]
Abstract
Botulinum neurotoxin (available commercially as BOTOX) has been used successfully for treatment of several neuromuscular disorders, including blepharospasm, dystonia, spasticity, and cerebral palsy in children. Our data demonstrate that injection of Botox into the proximal intestinal wall of diet-induced obese (DIO) mice induces weight loss and reduces food intake. This was associated with amelioration of hyperglycemia, hyperlipidemia, and significant improvement of glucose tolerance without alteration of energy expenditure. We also observed accelerated gastrointestinal transit and significant reductions in glucose and lipid absorption, which may account, at least in part, for the observed weight loss and robust metabolic benefits, although possible systemic effects occurring as a consequence of central and/or peripheral signaling cannot be ignored. The observed metabolic benefits were found to be largely independent of weight loss, as demonstrated by pair-feeding experiments. Effects lasted ∼8 weeks, for as long as the half-life of Botox as reported in prior rodent studies. These results have valuable clinical implications. If the observed effects are translatable in humans, this approach could lay the foundation for therapeutic approaches geared toward robust and sustained weight loss, mimicking some of the benefits of bariatric operations without its cost and complications.
Collapse
Affiliation(s)
- Sinju Sundaresan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Physiology, Midwestern University, Downers Grove, IL
- Corresponding author: Sinju Sundaresan,
| | - Joseph Antoun
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Babak Banan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Jamie Adcock
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Connor Johnson
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Brendan Claire
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Kala Dixon
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Joyce Flynn
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Cyndya A. Shibao
- Department of Physiology, Midwestern University, Downers Grove, IL
| | - Naji Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
62
|
Greenhalgh S, Chrystal PV, Lemme A, Juliano C DPD, Macelline SP, Liu SY, Selle PH. Capping dietary starch:protein ratios enhances performance of broiler chickens offered reduced-crude protein, maize-based diets. Anim Feed Sci Technol 2022. [DOI: 10.1016/j.anifeedsci.2022.115374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
63
|
Does Bentonite Cause Cytotoxic and Whole-Transcriptomic Adverse Effects in Enterocytes When Used to Reduce Aflatoxin B1 Exposure? Toxins (Basel) 2022; 14:toxins14070435. [PMID: 35878173 PMCID: PMC9322703 DOI: 10.3390/toxins14070435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/17/2022] Open
Abstract
Aflatoxin B1 (AFB1) is a major food safety concern, threatening the health of humans and animals. Bentonite (BEN) is an aluminosilicate clay used as a feed additive to reduce AFB1 presence in contaminated feedstuff. So far, few studies have characterized BEN toxicity and efficacy in vitro. In this study, cytotoxicity (WST-1 test), the effects on cell permeability (trans-epithelial electrical resistance and lucifer yellow dye incorporation), and transcriptional changes (RNA-seq) caused by BEN, AFB1 and their combination (AFB1 + BEN) were investigated in Caco-2 cells. Up to 0.1 mg/mL, BEN did not affect cell viability and permeability, but it reduced AFB1 cytotoxicity; however, at higher concentrations, BEN was cytotoxic. As to RNA-seq, 0.1 mg/mL BEN did not show effects on cell transcriptome, confirming that the interaction between BEN and AFB1 occurs in the medium. Data from AFB1 and AFB1 + BEN suggested AFB1 provoked most of the transcriptional changes, whereas BEN was preventive. The most interesting AFB1-targeted pathways for which BEN was effective were cell integrity, xenobiotic metabolism and transporters, basal metabolism, inflammation and immune response, p53 biological network, apoptosis and carcinogenesis. To our knowledge, this is the first study assessing the in vitro toxicity and whole-transcriptomic effects of BEN, alone or in the presence of AFB1.
Collapse
|
64
|
Liu G, Duan Y, Yang S, Yu M, Lv Z. Simultaneous quantification of marine neutral neoagaro-oligosaccharides and agar-oligosaccharides by the UHPLC-MS/MS method: application to the intestinal transport study by using the Caco-2 cell monolayer. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:2227-2234. [PMID: 35616101 DOI: 10.1039/d2ay00700b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A sensitive and robust ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method was established for the first time to simultaneously quantify marine neutral neoagaro-oligosaccharides (NAOS) and agar-oligosaccharides (AOS) with different degrees of polymerization (DP) in Hanks' balanced salt solution (HBSS). The separation was achieved on a BEH amide column using a mobile phase of acetonitrile-10 mmol L-1 ammonium acetate (58 : 42, v/v) with an isocratic elution program. The total analysis time was 3.5 min. The mass spectra were acquired in the multiple reaction monitoring (MRM) pattern by using a heated-electrospray ionization (H-ESI) source operating in the positive ionization mode. The linear range was 40-20 000 nmol L-1. The accuracy and precision ranged from 91.5 to 110.0% and 0.9 to 10.4%, respectively. The extraction recovery was consistent and reproducible. The stability was within 90.3-110.8%. The matrix effect, carryover, and dilution integrity were all satisfactory. Moreover, the validated method was successfully applied to the intestinal transport study by using the Caco-2 cell monolayer in vitro. The results revealed that neoagarobiose, neoagarotetraose, neoagarohexaose, agarotriose, agaropentose, and agaroheptose were transported by a paracellular pathway.
Collapse
Affiliation(s)
- Guilin Liu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
| | - Yunhai Duan
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
| | - Shuang Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, People's Republic of China
- Key Laboratory of Glycoscience & Glycotechnology of Shandong Province, Qingdao 266003, People's Republic of China
- Key Laboratory of Marine Drugs, Ministry of Education of China, Qingdao 266003, People's Republic of China
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, People's Republic of China
- Key Laboratory of Glycoscience & Glycotechnology of Shandong Province, Qingdao 266003, People's Republic of China
- Key Laboratory of Marine Drugs, Ministry of Education of China, Qingdao 266003, People's Republic of China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, People's Republic of China
- Key Laboratory of Glycoscience & Glycotechnology of Shandong Province, Qingdao 266003, People's Republic of China
- Key Laboratory of Marine Drugs, Ministry of Education of China, Qingdao 266003, People's Republic of China
| |
Collapse
|
65
|
Ma Y, Lee E, Yoshikawa H, Noda T, Miyamoto J, Kimura I, Hatano R, Miki T. Phloretin suppresses carbohydrate-induced GLP-1 secretion via inhibiting short chain fatty acid release from gut microbiome. Biochem Biophys Res Commun 2022; 621:176-182. [DOI: 10.1016/j.bbrc.2022.06.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022]
|
66
|
Zhang X, Cheng Z, Dong S, Rayner C, Wu T, Zhong M, Zhang G, Wang K, Hu S. Effects of ileal glucose infusion on enteropancreatic hormone secretion in humans: relationship to glucose absorption. Metabolism 2022; 131:155198. [PMID: 35395220 DOI: 10.1016/j.metabol.2022.155198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/13/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUNDS The distal small intestine plays an important role in regulating the secretion of entero-pancreatic hormones that are critical to the control of glucose metabolism and appetite, but the quantitative contribution of a specific segment to these effects is unknown. PURPOSES To determine the effects of 30 cm of the ileum exposed to glucose on the secretion of ghrelin, glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) insulin, C-peptide and glucagon, in relation to glucose absorption in non-diabetic subjects. BASIC PROCEDURES 10 non-diabetic subjects with a loop ileostomy after early-stage rectal cancer resection were studied on 2 days in a double-blind, randomized and crossover fashion, when a catheter was inserted retrogradely 30 cm from the ileostomy for infusion of a glucose solution containing 30 g glucose and 3 g 3-O-methylglucose (as a marker of active glucose absorption), or 0.9% saline, over 60 min. Ghrelin, GIP, GLP-1, insulin, C-peptide, glucagon and ileal glucose absorption (from concentrations of 3-O-methylglucose in serum and glucose in ileostomy effluent) were measured over 180 min. MAIN FINDINGS 12.0 ± 1.2 g glucose was absorbed over 180 min. Compared to saline, ileal glucose resulted in minimal increases in blood glucose and plasma insulin and C-peptide, but substantial increases in plasma GLP-1, without affecting ghrelin, GIP or glucagon. The magnitude of the GLP-1 response to glucose was strongly related to the increase in serum 3-O-methylglucose. PRINCIPAL CONCLUSIONS Stimulation of the terminal ileum by glucose, even over a short length (30 cm), induces substantial GLP-1 release, coupled primarily to active glucose absorption. CLINICAL REGISTRATION NCT05030376 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Zhiqiang Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Shuohui Dong
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Christopher Rayner
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia
| | - Tongzhi Wu
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia
| | - Mingwei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Kexin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.
| | - Sanyuan Hu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China; Shandong University, China.
| |
Collapse
|
67
|
Zapater JL, Wicksteed B, Layden BT. Enterocyte HKDC1 Modulates Intestinal Glucose Absorption in Male Mice Fed a High-fat Diet. Endocrinology 2022; 163:6569855. [PMID: 35435980 PMCID: PMC9078327 DOI: 10.1210/endocr/bqac050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Indexed: 11/24/2022]
Abstract
Hexokinase domain containing protein-1, or HKDC1, is a widely expressed hexokinase that is genetically associated with elevated 2-hour gestational blood glucose levels during an oral glucose tolerance test, suggesting a role for HKDC1 in postprandial glucose regulation during pregnancy. Our earlier studies utilizing mice containing global HKDC1 knockdown, as well as hepatic HKDC1 overexpression and knockout, indicated that HKDC1 is important for whole-body glucose homeostasis in aging and pregnancy, through modulation of glucose tolerance, peripheral tissue glucose utilization, and hepatic energy storage. However, our knowledge of the precise role(s) of HKDC1 in regulating postprandial glucose homeostasis under normal and diabetic conditions is lacking. Since the intestine is the main entry portal for dietary glucose, here we have developed an intestine-specific HKDC1 knockout mouse model, HKDC1Int-/-, to determine the in vivo role of intestinal HKDC1 in regulating glucose homeostasis. While no overt glycemic phenotype was observed, aged HKDC1Int-/- mice fed a high-fat diet exhibited an increased glucose excursion following an oral glucose load compared with mice expressing intestinal HKDC1. This finding resulted from glucose entry via the intestinal epithelium and is not due to differences in insulin levels, enterocyte glucose utilization, or reduction in peripheral skeletal muscle glucose uptake. Assessment of intestinal glucose transporters in high-fat diet-fed HKDC1Int-/- mice suggested increased apical GLUT2 expression in the fasting state. Taken together, our results indicate that intestinal HKDC1 contributes to the modulation of postprandial dietary glucose transport across the intestinal epithelium under conditions of enhanced metabolic stress, such as high-fat diet.
Collapse
Affiliation(s)
- Joseph L Zapater
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Medical Research Service, Chicago, IL 60612, USA
| | - Barton Wicksteed
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Brian T Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Medical Research Service, Chicago, IL 60612, USA
- Correspondence: Brian T. Layden, MD, PhD, 835 South Wolcott Avenue, Suite 625E (M/C 640), Chicago, IL, 60612, USA.
| |
Collapse
|
68
|
Macelline SP, Chrystal PV, Selle PH, Liu SY. Protein sources and starch-protein digestive dynamics manipulate growth performance in broiler chickens defined by an equilateral-triangle response surface design. ANIMAL NUTRITION 2022; 9:204-213. [PMID: 35600555 PMCID: PMC9092981 DOI: 10.1016/j.aninu.2022.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/28/2021] [Accepted: 01/27/2022] [Indexed: 12/01/2022]
Abstract
A total of 360 male, off-sex Ross 308 chicks were offered 10 dietary treatments from 14 to 35 d post–hatch in an equilateral-triangle response surface design feeding study in order to confirm the importance of protein and amino acid digestive dynamics in broiler chickens. The 3 apical diets were nutritionally-equivalent containing either soybean meal, non-bound amino acids or whey protein concentrate as the major source of dietary protein and amino acids. Appropriate blends of the 3 apical diets comprised the balance of 7 diets and each dietary treatment was offered to 6 replicate cages with 6 birds per cage. Growth performance, nutrient utilisation, apparent protein and starch digestibility coefficients were determined in 4 small intestinal segments. The optimal weight gain (2,085 g/bird) and feed conversion ratios (FCR, 1.397) were generated by Diet 50S50W which included a 50:50 blend of apical diets rich in whey protein concentrate and soybean meal. Broiler chickens offered Diet 50S50W also had the highest experimental and predicted jejunal digestibility (0.685 in proximal jejunum and 0.823 in distal jejunum). FCR was not correlated with apparent distal ileal digestibility coefficient (P > 0.05) of protein but was correlated with apparent protein digestibility in proximal jejunum (r = −0.369, P = 0.040) and distal jejunum (r = −0.316, P = 0.015). Surplus dietary starch was correlated with increased fat pad weight (r = 0.781, P = 0.008). The findings confirmed the relevance of protein digestion rate, reflected by jejunal digestibility, on feed conversion of broiler chickens. A balance between protein-bound and non-bound crystalline or synthetic amino acids may be required for optimal growth and protein digestion.
Collapse
Affiliation(s)
- Shemil P. Macelline
- Poultry Research Foundation, The University of Sydney, Camden NSW 2570, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney NSW 2006, Australia
| | - Peter V. Chrystal
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney NSW 2006, Australia
| | - Peter H. Selle
- Poultry Research Foundation, The University of Sydney, Camden NSW 2570, Australia
- Sydney School of Veterinary Science, The University of Sydney, Sydney NSW 2006, Australia
| | - Sonia Y. Liu
- Poultry Research Foundation, The University of Sydney, Camden NSW 2570, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney NSW 2006, Australia
- Corresponding author.
| |
Collapse
|
69
|
Elebring E, Wallenius V, Casselbrant A, Docherty NG, le Roux CW, Marschall HU, Fändriks L. A Fatty Diet Induces a Jejunal Ketogenesis Which Inhibits Local SGLT1-Based Glucose Transport via an Acetylation Mechanism—Results from a Randomized Cross-Over Study between Iso-Caloric High-Fat versus High-Carbohydrate Diets in Healthy Volunteers. Nutrients 2022; 14:nu14091961. [PMID: 35565929 PMCID: PMC9100393 DOI: 10.3390/nu14091961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/27/2022] Open
Abstract
Background and aims: Insights into the nature of gut adaptation after different diets enhance the understanding of how food modifications can be used to treat type 2 diabetes and obesity. The aim was to understand how diets, enriched in fat or carbohydrates, affect glucose absorption in the human healthy jejunum, and what mechanisms are involved. Methods: Fifteen healthy subjects received, in randomised order and a crossover study design, two weeks of iso-caloric high-fat diet (HFD) and high-carbohydrate diet (HCD). Following each dietary period, jejunal mucosa samples were retrieved and assessed for protein expression using immunofluorescence and western blotting. Functional characterisation of epithelial glucose transport was assessed ex vivo using Ussing chambers. Regulation of SGLT1 through histone acetylation was studied in vitro in Caco-2 and human jejunal enteroid monolayer cultures. Results: HFD, compared to HCD, decreased jejunal Ussing chamber epithelial glucose transport and the expression of apical transporters for glucose (SGLT1) and fructose (GLUT5), while expression of the basolateral glucose transporter GLUT2 was increased. HFD also increased protein expression of the ketogenesis rate-limiting enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (HMGCS2) and decreased the acetylation of histone 3 at lysine 9 (H3K9ac). Studies in Caco-2 and human jejunal enteroid monolayer cultures indicated a ketogenesis-induced activation of sirtuins, in turn decreasing SGLT1 expression. Conclusion: Jejunal glucose absorption is decreased by a fat-enriched diet, via a ketogenesis-induced alteration of histone acetylation responsible for the silencing of SGLT1 transcription. The work relates to a secondary outcome in ClinicalTrials.gov (NCT02088853).
Collapse
Affiliation(s)
- Erik Elebring
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, SE41345 Gothenburg, Sweden; (E.E.); (V.W.); (A.C.)
| | - Ville Wallenius
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, SE41345 Gothenburg, Sweden; (E.E.); (V.W.); (A.C.)
- Department of Surgery, Sahlgrenska University Hospital, SE41345 Gothenburg, Sweden
| | - Anna Casselbrant
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, SE41345 Gothenburg, Sweden; (E.E.); (V.W.); (A.C.)
| | - Neil G. Docherty
- Metabolic Medicine, School of Medicine, Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland; (N.G.D.); (C.W.l.R.)
| | - Carel W. le Roux
- Metabolic Medicine, School of Medicine, Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland; (N.G.D.); (C.W.l.R.)
| | - Hanns-Ulrich Marschall
- Institute of Medicine, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, SE41345 Gothenburg, Sweden;
- Department of Medicine, Sahlgrenska University Hospital, SE41345 Gothenburg, Sweden
| | - Lars Fändriks
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, SE41345 Gothenburg, Sweden; (E.E.); (V.W.); (A.C.)
- Department of Surgery, Sahlgrenska University Hospital, SE41345 Gothenburg, Sweden
- Correspondence: ; Tel.: +46-313424123
| |
Collapse
|
70
|
Zhu S, Li J, Li W, Li S, Yang X, Liu X, Sun L. Enzymic catalyzing affinity to substrate affects inhibitor-enzyme binding interactions: Inhibition behaviors of EGCG against starch digestion by individual and co-existing α-amylase and amyloglucosidase. Food Chem 2022; 388:133047. [PMID: 35483290 DOI: 10.1016/j.foodchem.2022.133047] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 03/11/2022] [Accepted: 04/20/2022] [Indexed: 02/09/2023]
Abstract
The inhibition of (-)-epigallocatechin-gallate (EGCG) against starch digestion by α-amylase (AA), amyloglucosidase (AMG) and co-existing enzymes (AA/AMG) were comparatively studied. EGCG inhibited AA only at slowly-digestible-starch (SDS) phase. This resulted from high catalytic efficiency of AA for rapidly-digestible-starch (RDS), counteracting the inhibition at this phase. EGCG inhibited AMG and AA/AMG during whole process. At RDS phase, the catalytic velocity of AMG was always higher than AA/AMG because of an antagonistic effect of two enzymes. However, at SDS phase with EGCG, the catalytic velocity of AA/AMG was higher than AMG. This is because binding of EGCG with both enzymes caused more unbound AMG that generated more glucose in co-existing AA/AMG than AMG. Although EGCG-AA binding affinity was higher than EGCG-AMG, competitive inhibition of EGCG against AA was weaker than AMG, indicating relatively higher binding/catalyzing affinity of AA to starch significantly weakened EGCG-AA binding due to competitive relationship between starch and EGCG.
Collapse
Affiliation(s)
- Shengnan Zhu
- College of Food Science and Engineering, Northwest A & F University, China
| | - Jing Li
- College of Food Science and Engineering, Northwest A & F University, China
| | - Wenyue Li
- College of Food Science and Engineering, Northwest A & F University, China
| | - Shuangshuang Li
- College of Food Science and Engineering, Northwest A & F University, China
| | - Xi Yang
- College of Food Science and Engineering, Northwest A & F University, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A & F University, China
| | - Lijun Sun
- College of Food Science and Engineering, Northwest A & F University, China.
| |
Collapse
|
71
|
Li S, Wu W, Li J, Zhu S, Yang X, Sun L. α-Amylase Changed the Catalytic Behaviors of Amyloglucosidase Regarding Starch Digestion Both in the Absence and Presence of Tannic Acid. Front Nutr 2022; 9:817039. [PMID: 35495955 PMCID: PMC9043763 DOI: 10.3389/fnut.2022.817039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/01/2022] [Indexed: 11/13/2022] Open
Abstract
The courses of starch digestion with individual α-amylase (AA), amyloglucosidase (AMG), and AA/AMG bi-enzyme system were performed and analyzed by first-order-reaction equations in the absence and presence of tannic acid (TA). An antagonistic effect between AA and AMG occurred at the digestion phase of readily-digestible starch due to the higher catalytic efficiency of AMG for starchy-substrates with more complex structures. This effect caused a faster rate of glucose production with AMG than with AA/AMG bi-enzyme system at this phase both in the absence and presence of TA. TA had a higher binding affinity to AA than to AMG as accessed by several methods, such as inhibition kinetics, fluorescence quenching, isothermal titration calorimetry (ITC), and molecular docking. Besides, differential scanning calorimetry (DSC) indicated that the change in the thermal and structural stabilities of enzymes in the presence of TA was related to the enzyme residues involved in binding with TA, rather than the inhibitory effects of TA. The binding characters of TA to both enzymes resulted in more “free” AMG without TA binding in AA/AMG bi-enzyme system than that in individual AMG. This binding property caused more and faster rate of glucose production at the digestion phase of slowly digestible starch (SDS) in the bi-enzyme system.
Collapse
|
72
|
Zhu H, Cai H, Wang X, Chen T, Zhen C, Zhang Z, Ruan X, Li G. Sodium-glucose co-transporter 1 (SGLT1) differentially regulates gluconeogenesis and GLP-1 receptor (GLP-1R) expression in different diabetic rats: a preliminary validation of the hypothesis of "SGLT1 bridge" as an indication for "surgical diabetes". ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:481. [PMID: 35571394 PMCID: PMC9096370 DOI: 10.21037/atm-22-1769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Sodium-glucose co-transporter 1 (SGLT1) may play a synergistic role in gluconeogenesis (GNG) and glucagon-like peptide-1 (GLP-1) expression. We proposed the hypothesis of a "SGLT1 bridge" as an indication for "surgical diabetes" that was preliminary validated in the present study. METHODS We selected nonobese diabetic Goto-Kakizaki (GK) rats and Zuker diabetic fat (ZDF) rats to represent advanced and early diabetes, respectively. Based on glucose gavage with or without SGLT1 inhibitor phlorizin, the rats were divided into 4 groups: Gk-Glu, GK-P, ZDF-Glu, and ZDF-P. The expressions of SGLT1, GLP-1 receptor (GLP-1R), glucose-6 phosphatase (G6Pase), and phosphoenolpyruvate carboxykinase-1 (Pck1) were determined by immunohistochemistry (IHC) or quantitative reverse transcription polymerase chain reaction (RT-qPCR), and the effects of phlorizin were analyzed. RESULTS Glucose tolerance was worse in GK rats and the homeostasis model assessment-insulin resistance (HOMA-IR) was higher in ZDF rats, indicating different pathophysiological conditions between the different diabetic rats. GK rats showed higher activity of duodenal SGLT1 (P=0.022) and jejunal SGLT1 mRNA expression (P=0.000) and lower SGLT1 mRNA expression in the liver (P=0.000) and pancreas (P=0.000). Phlorizin effectively inhibited the activity of duodenal SGLT1 in both GK rats (P=0.000) and ZDF rats (P=0.000). In ZDF rats, the expression of GLP-1R mRNA was downregulated in the jejunum (P=0.001) and upregulated in the pancreas (P=0.021) by phlorizin, but there were no regulatory effects on GLP-1R mRNA in the jejunum and pancreas of GK rats. As for the regulatory effects on GNG, phlorizin upregulated Pck1 mRNA in the duodenum (P=0.000) and the jejunum (P=0.038), whereas it downregulated hepatic G6Pase mRNA in ZDF rats (P=0.005) and Pck1 mRNA expression in GK rats (P=0.001), suggesting that SGLT1 inhibitor may have upregulated intestinal GNG in ZDF rats and downregulated hepatic GNG in both ZDF and GK rats. CONCLUSIONS SGLT1 showed synergistic regulatory effects on the entero-insular axis (EIA) and the gut-brain-liver axis (GBLA), preliminarily validating the hypothesis of a "SGLT1 bridge". The distinct expression of SGLT1 and its differentially regulatory effects on diabetic rats with different pathophysiological conditions may provide probable potential indications involved in the "Surgical Diabetes" that is supposed as the inclusion for diabetic surgery.
Collapse
Affiliation(s)
- Hengliang Zhu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of General Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, China
| | - Huajie Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Wang
- Department of Gastrointestinal & Hernia Surgery, Ganzhou People’s Hospital, Ganzhou, China
| | - Tao Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chaohui Zhen
- Department of General Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, China
| | - Zhenzhan Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojiao Ruan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
73
|
Impact of Feeding Fermented Palm Kernel Cake and High Dietary Fat on Nutrient Digestibility, Enzyme Activity, Intestinal Morphology and Intestinal Nutrient Transporters mRNA Expression in Broiler Chickens under Hot and Humid Conditions. Animals (Basel) 2022; 12:ani12070882. [PMID: 35405871 PMCID: PMC8997065 DOI: 10.3390/ani12070882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 01/25/2023] Open
Abstract
The study aimed at determining the ileal nutrient digestibility, digestive enzyme activity, intestinal morphology, and nutrient transporters mRNA expressions in broiler chickens fed with fermented PKC (LPKC) based diets with different levels of fat supplementation under hot and humid conditions. From day 22 to 35, broiler chickens were randomly fed with either (1) 20% LPKC-based diet with 5% palm oil, (2) 20% LPKC based diet with 9.5% palm oil, (3) 20% PKC-based diet with 5% palm oil or (4) 20% PKC-based diet with 9.5% palm oil. Feeding LPKC and PKC diets at the finisher phase have not affected the nutrient’s digestibility, but a higher level of oil supplementation does. This was seconded by changes in the digestive enzyme activity, villus height, and mRNA expression of nutrient transporters in the higher level of oil-supplemented diets fed chickens. In conclusion, the inclusion of oil at 9.5% in a 20% LPKC/PKC-based diet is necessary to ensure better nutrient digestibility in chickens via improved digestive function, especially in hot and humid tropical regions.
Collapse
|
74
|
Chen S, Wu Y, Wang S, Wu J, Wu X, Zheng Z. A risk model of gene signatures for predicting platinum response and survival in ovarian cancer. J Ovarian Res 2022; 15:39. [PMID: 35361267 PMCID: PMC8973612 DOI: 10.1186/s13048-022-00969-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the deadliest tumor in the female reproductive tract. And increased resistance to platinum-based chemotherapy represents the major obstacle in the treatment of OC currently. Robust and accurate gene expression models are crucial tools in distinguishing platinum therapy response and evaluating the prognosis of OC patients. METHODS In this study, 230 samples from The Cancer Genome Atlas (TCGA) OV dataset were subjected to mRNA expression profiling, single nucleotide polymorphism (SNP), and copy number variation (CNV) analysis comprehensively to screen out the differentially expressed genes (DEGs). An SVM classifier and a prognostic model were constructed using the Random Forest algorithm and LASSO Cox regression model respectively via R. The Gene Expression Omnibus (GEO) database was applied as the validation set. RESULTS Forty-eight differentially expressed genes (DEGs) were figured out through integrated analysis of gene expression, single nucleotide polymorphism (SNP), and copy number variation (CNV) data. A 10-gene classifier was constructed which could discriminate platinum-sensitive samples precisely with an AUC of 0.971 in the training set and of 0.926 in the GEO dataset (GSE638855). In addition, 8 optimal genes were further selected to construct the prognostic risk model whose predictions were consistent with the actual survival outcomes in the training cohort (p = 9.613e-05) and validated in GSE638855 (p = 0.04862). PNLDC1, SLC5A1, and SYNM were then identified as hub genes that were associated with both platinum response status and prognosis, which was further validated by the Fudan University Shanghai cancer center (FUSCC) cohort. CONCLUSION These findings reveal a specific risk model that could serve as effective biomarkers to identify patients' platinum response status and predict survival outcomes for OC patients. PNLDC1, SLC5A1, and SYNM are the hub genes that may serve as potential biomarkers in OC treatment.
Collapse
Affiliation(s)
- Siyu Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Simin Wang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiangchun Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Zhong Zheng
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
75
|
Jahandideh F, Bourque SL, Wu J. A comprehensive review on the glucoregulatory properties of food-derived bioactive peptides. Food Chem X 2022; 13:100222. [PMID: 35498998 PMCID: PMC9039931 DOI: 10.1016/j.fochx.2022.100222] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/03/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus, a group of metabolic disorders characterized by persistent hyperglycemia, affects millions of people worldwide and is on the rise. Dietary proteins, from a wide range of food sources, are rich in bioactive peptides with antidiabetic properties. Notable examples include AGFAGDDAPR, a black tea-derived peptide, VRIRLLQRFNKRS, a β-conglycinin-derived peptide, and milk-derived peptide VPP, which have shown antidiabetic effects in diabetic rodent models through variety of pathways including improving beta-cells function, suppression of alpha-cells proliferation, inhibiting food intake, increasing portal cholecystokinin concentration, enhancing insulin signaling and glucose uptake, and ameliorating adipose tissue inflammation. Despite the immense research on glucoregulatory properties of bioactive peptides, incorporation of these bioactive peptides in functional foods or nutraceuticals is widely limited due to the existence of several challenges in the field of peptide research and commercialization. Ongoing research in this field, however, is fundamental to pave the road for this purpose.
Collapse
Key Words
- AMPK, AMP-activated protein kinase
- Akt, Protein kinase B
- Bioactive peptides
- C/EBP-α, CCAAT/ enhancer binding protein alpha
- CCK, Cholecystokinin
- CCK-1R, CCK type 1 receptor
- DPP-IV, Dipeptidyl peptidase IV
- Diabetes mellitus
- ERK1/2, Extracellular signal regulated kinase 1/2
- GIP, Glucose-dependent insulinotropic polypeptide
- GLP-1, Glucagon-like peptide 1
- GLUT, Glucose transporter
- Glucose homeostasis
- IRS-1, Insulin receptor substrate-1
- Insulin resistance
- MAPK, Mitogen activated protein kinase
- PI3K, Phosphatidylinositol 3-kinase
- PPARγ, Peroxisome proliferator associated receptor gamma
- Reproductive dysfunction
- TZD, Thiazolidinedione
- cGMP, cyclic guanosine-monophosphate
Collapse
Affiliation(s)
- Forough Jahandideh
- Department of Anesthesiology & Pain Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada.,Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Stephane L Bourque
- Department of Anesthesiology & Pain Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada.,Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Jianping Wu
- Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
76
|
Wang G, Li X, Zhou Y, Feng J, Zhang M. Effects of Dietary Chromium Picolinate on Gut Microbiota, Gastrointestinal Peptides, Glucose Homeostasis, and Performance of Heat-Stressed Broilers. Animals (Basel) 2022; 12:ani12070844. [PMID: 35405834 PMCID: PMC8997060 DOI: 10.3390/ani12070844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/13/2022] [Accepted: 03/23/2022] [Indexed: 12/25/2022] Open
Abstract
The current research was devoted to evaluating the effects on gut microbiota, gastrointestinal peptides, and glucose homeostasis of chromium picolinate applied to heat-stressed broilers. In a 14 d experiment, 220 28-day-old AA broilers were randomly assigned into one thermal-neutral and three high-temperature groups dietary-supplemented with 0, 0.4, or 0.8 mg/kg of chromium as chromium picolinate. The temperature for the thermal-neutral group was set at 21 °C, while that for the other three groups (high temperature) was set at 31 °C. The results showed that the average daily gain and average daily feed intake of the 0.4 mg/kg chromium-supplemented group significantly increased compared with the high-temperature groups (p < 0.05). The content of cholecystokinin in the 0.4 mg/kg group significantly decreased, and the gastric inhibitory polypeptide level was significantly elevated in jejunum (p < 0.05). The cecal microbiota of heat-stressed broilers was substantially different from that of the thermal-neutral group. After diet-supplemented chromium, compared to the high-temperature groups, the 0.4 mg/kg chromium supplemented group was characterized by a reduction of Actinobacteriota and Proteobacteria at the phylum level. The Bacilli were elevated, while proportions of Coriobacteria and Gammaproteobacteria were reduced significantly at the class level. The proportions of Lactobacillaceae, Christensenellaceae, and Erysipelotrichaceae were elevated significantly, while that of Clostridiaceae was reduced significantly at the family level. The proportion of Turicibacter was elevated significantly and the proportions of Olsenella and Ruminococcus were reduced significantly at the genus level (p < 0.05). Compared to the high-temperature groups, in the 0.4 mg/kg chromium-supplemented group, the insulin concentration and insulin resistance index were reduced (p < 0.05), and sodium-glucose transporter 1 expression was up-regulated in jejunum (p < 0.05). Performance, microbiota, gastrointestinal peptides, or serum parameters of the 0.8 mg/kg group were almost unaffected by chromium compared with the high-temperature groups. In conclusion, diet supplemented with 0.4 mg/kg Cr improved performance, insulin resistance and sodium-glucose transporter 1 expression and altered gut microflora structure and secretion of gastrointestinal peptides, thus showing that supplementation with chromium is beneficial to maintain glucose homeostasis and alleviate heat stress.
Collapse
|
77
|
Ortega R, Valdés M, Alarcón-Aguilar FJ, Fortis-Barrera Á, Barbosa E, Velazquez C, Calzada F. Antihyperglycemic Effects of Salvia polystachya Cav. and Its Terpenoids: α-Glucosidase and SGLT1 Inhibitors. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11050575. [PMID: 35270046 PMCID: PMC8912538 DOI: 10.3390/plants11050575] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 06/02/2023]
Abstract
The antihyperglycemic activity of ethanolic extract from Salvia polystachya (EESpS) and its products was evaluated using in vivo, ex vivo and in silico assays; additionally, an acute toxicity assay was evaluated. EESpS was classified as a nontoxic class 5 drug. EESpS, ethyl acetate fraction (EtOAcFr), secondary-6-fraction (SeFr6), ursolic acid (UA), and oleanolic acid (OA) reduced the hyperglycemia in DM2 mice. α-glucosidase inhibition was evaluated with oral sucrose and starch tolerance tests (OSuTT and OStTT), an intestinal sucrose hydrolysis (ISH) assay and molecular docking studies using acarbose as control. SGLT1 inhibition was evaluated with oral glucose and galactose tolerance tests (OGTT and OGaTT), an intestinal glucose absorption (IGA) assay and molecular docking studies using canagliflozin as the control. During the carbohydrate tolerance tests, all the treatments reduced the postprandial peak, similar to the control drugs. During the ISH, IC50 values of 739.9 and 726.3 µM for UA and OA, respectively, were calculated. During the IGA, IC50 values of 966.6 and 849.3 for UA, OA respectively, were calculated. Finally, during the molecular docking studies, UA and OA showed ∆G values of -6.41 and -5.48 kcal/mol-1, respectively, on α-glucosidase enzymes. During SGLT1, UA and OA showed ∆G values of -10.55 and -9.65, respectively.
Collapse
Affiliation(s)
- Rocio Ortega
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, UAM-I, Mexico City CP 09340, Mexico
- Av. San Rafael Atlixco 186, Leyes de Reforma 1ra Sección, Iztapalapa, Mexico City CP 09340, Mexico
- Unidad de Investigación Médica en Farmacología, UMAE Hospital de Especialidades, 2° Piso CORSE Centro Médico Nacional Siglo XXI, IMSS, Av. Cuauhtémoc 330, Col. Doctores, Mexico City CP 06725, Mexico
| | - Miguel Valdés
- Unidad de Investigación Médica en Farmacología, UMAE Hospital de Especialidades, 2° Piso CORSE Centro Médico Nacional Siglo XXI, IMSS, Av. Cuauhtémoc 330, Col. Doctores, Mexico City CP 06725, Mexico
| | - Francisco J. Alarcón-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de CBS, Universidad Autónoma Metropolitana-Iztapalapa, UAM-I, Av. San Rafael Atlixco 186, Leyes de Reforma 1ra Sección, Mexico City CP 09340, Mexico; (F.J.A.-A.); (Á.F.-B.)
| | - Ángeles Fortis-Barrera
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de CBS, Universidad Autónoma Metropolitana-Iztapalapa, UAM-I, Av. San Rafael Atlixco 186, Leyes de Reforma 1ra Sección, Mexico City CP 09340, Mexico; (F.J.A.-A.); (Á.F.-B.)
| | - Elizabeth Barbosa
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Mexico City CP 11340, Mexico;
| | - Claudia Velazquez
- Área Académica de Farmacia, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Km 4.5, Carretera Pachuca-Tulancingo, Unidad Universitaria, Pachuca CP 42076, Mexico;
| | - Fernando Calzada
- Unidad de Investigación Médica en Farmacología, UMAE Hospital de Especialidades, 2° Piso CORSE Centro Médico Nacional Siglo XXI, IMSS, Av. Cuauhtémoc 330, Col. Doctores, Mexico City CP 06725, Mexico
| |
Collapse
|
78
|
Prescott SL, Liberles SD. Internal senses of the vagus nerve. Neuron 2022; 110:579-599. [PMID: 35051375 PMCID: PMC8857038 DOI: 10.1016/j.neuron.2021.12.020] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/30/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022]
Abstract
The vagus nerve is an indispensable body-brain connection that controls vital aspects of autonomic physiology like breathing, heart rate, blood pressure, and gut motility, reflexes like coughing and swallowing, and survival behaviors like feeding, drinking, and sickness responses. Classical physiological studies and recent molecular/genetic approaches have revealed a tremendous diversity of vagal sensory neuron types that innervate different internal organs, with many cell types remaining poorly understood. Here, we review the state of knowledge related to vagal sensory neurons that innervate the respiratory, cardiovascular, and digestive systems. We focus on cell types and their response properties, physiological/behavioral roles, engaged neural circuits and, when possible, sensory receptors. We are only beginning to understand the signal transduction mechanisms used by vagal sensory neurons and upstream sentinel cells, and future studies are needed to advance the field of interoception to the level of mechanistic understanding previously achieved for our external senses.
Collapse
|
79
|
Elferink H, Titulaer WHC, Derks MGN, Veeneman GH, Rutjes FPJT, Boltje TJ. Chloromethyl Glycosides as Versatile Synthons to Prepare Glycosyloxymethyl-Prodrugs. Chemistry 2022; 28:e202103910. [PMID: 35045197 PMCID: PMC9304170 DOI: 10.1002/chem.202103910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Indexed: 11/21/2022]
Abstract
This work investigates the addition of monosaccharides to marketed drugs to improve their pharmacokinetic properties for oral absorption. To this end, a set of chloromethyl glycoside synthons were developed to prepare a variety of glycosyloxymethyl-prodrugs derived from 5-fluorouracil, thioguanine, propofol and losartan. Drug release was studied in vitro using β-glucosidase confirming rapid conversion of the monosaccharide prodrugs to release the parent drug, formaldehyde and the monosaccharide. To showcase this prodrug approach, a glucosyloxymethyl conjugate of the tetrazole-containing drug losartan was used for in vivo experiments and showed complete release of the drug in a dog-model.
Collapse
Affiliation(s)
- Hidde Elferink
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | - Willem H. C. Titulaer
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | - Maik G. N. Derks
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | | | - Floris P. J. T. Rutjes
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | - Thomas J. Boltje
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| |
Collapse
|
80
|
Dextrose 10% drink is superior to sodium-dextrose drink in increasing blood glucose and sprint speed in soccer players: A double-blinded randomized crossover trial study. Sci Sports 2022. [DOI: 10.1016/j.scispo.2020.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
81
|
Wu C, Jeong MY, Kim JY, Lee G, Kim JS, Cheong YE, Kang H, Cho CH, Kim J, Park MK, Shin YK, Kim KH, Seol GH, Koo SH, Ko G, Lee SJ. Activation of ectopic olfactory receptor 544 induces GLP-1 secretion and regulates gut inflammation. Gut Microbes 2022; 13:1987782. [PMID: 34674602 PMCID: PMC8632334 DOI: 10.1080/19490976.2021.1987782] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Olfactory receptors are ectopically expressed in extra-nasal tissues. The gut is constantly exposed to high levels of odorants where ectopic olfactory receptors may play critical roles. Activation of ectopic olfactory receptor 544 (Olfr544) by azelaic acid (AzA), an Olfr544 ligand, reduces adiposity in mice fed a high-fat diet (HFD) by regulating fuel preference to fats. Herein, we investigated the novel function of Olfr544 in the gut. In GLUTag cells, AzA induces the cAMP-PKA-CREB signaling axis and increases the secretion of GLP-1, an enteroendocrine hormone with anti-obesity effects. In mice fed a HFD and orally administered AzA, GLP-1 plasma levels were elevated in mice. The induction of GLP-1 secretion was negated in cells with Olfr544 gene knockdown and in Olfr544-deficient mice. Gut microbiome analysis revealed that AzA increased the levels of Bacteroides acidifaciens and microbiota associated with antioxidant pathways. In fecal metabolomics analysis, the levels of succinate and trehalose, metabolites correlated with a lean phenotype, were elevated by AzA. The function of Olfr544 in gut inflammation, a key feature in obesity, was further investigated. In RNA sequencing analysis, AzA suppressed LPS-induced activation of inflammatory pathways and reduced TNF-α and IL-6 expression, thereby improving intestinal permeability. The effects of AzA on the gut metabolome, microbiome, and colon inflammation were abrogated in Olfr544-KO mice. These results collectively demonstrated that activation of Olfr544 by AzA in the gut exerts multiple effects by regulating GLP-1 secretion, gut microbiome and metabolites, and colonic inflammation in anti-obesogenic phenotypes and, thus, may be applied for obesity therapeutics.
Collapse
Affiliation(s)
- Chunyan Wu
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| | - Mi-Young Jeong
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| | - Jung Yeon Kim
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| | - Giljae Lee
- Department of Environmental Health Sciences, Seoul National University, Seoul, Republic of Korea,Center for Human and Environmental Microbiome, Seoul National University, Seoul, Republic of Korea
| | - Ji-Sun Kim
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| | - Yu Eun Cheong
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| | - Hyena Kang
- Department of Environmental Health Sciences, Seoul National University, Seoul, Republic of Korea,Center for Human and Environmental Microbiome, Seoul National University, Seoul, Republic of Korea
| | - Chung Hwan Cho
- Department of Environmental Health Sciences, Seoul National University, Seoul, Republic of Korea,Center for Human and Environmental Microbiome, Seoul National University, Seoul, Republic of Korea
| | - Jimin Kim
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| | - Min Kyung Park
- Department of Food Science and Engineering, Ewha Womans University, Seoul, Republic of Korea
| | - You Kyoung Shin
- Department of Basic Nursing Science, School of Nursing, Korea University, Seoul, Republic of Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| | - Geun Hee Seol
- Department of Basic Nursing Science, School of Nursing, Korea University, Seoul, Republic of Korea
| | - Seung Hoi Koo
- Division of Biological Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - GwangPyo Ko
- Department of Environmental Health Sciences, Seoul National University, Seoul, Republic of Korea,Center for Human and Environmental Microbiome, Seoul National University, Seoul, Republic of Korea
| | - Sung-Joon Lee
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea,CONTACT Sung-Joon Lee Department of Biotechnology, School of Life Science and Biotechnology for BK21 Plus, Korea University, Seoul, Republic of Korea
| |
Collapse
|
82
|
Gromova LV, Polozov AS, Savochkina EV, Alekseeva AS, Dmitrieva YV, Kornyushin OV, Gruzdkov AA. Effect of Type 2 Diabetes and Impaired Glucose Tolerance on Digestive Enzymes and Glucose Absorption in the Small Intestine of Young Rats. Nutrients 2022; 14:nu14020385. [PMID: 35057569 PMCID: PMC8779211 DOI: 10.3390/nu14020385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/10/2022] Open
Abstract
The reactions of intestinal functional parameters to type 2 diabetes at a young age remain unclear. The study aimed to assess changes in the activity of intestinal enzymes, glucose absorption, transporter content (SGLT1, GLUT2) and intestinal structure in young Wistar rats with type 2 diabetes (T2D) and impaired glucose tolerance (IGT). To induce these conditions in the T2D (n = 4) and IGT (n = 6) rats, we used a high-fat diet and a low dose of streptozotocin. Rats fed a high-fat diet (HFD) (n = 6) or a standard diet (SCD) (n = 6) were used as controls. The results showed that in T2D rats, the ability of the small intestine to absorb glucose was higher in comparison to HFD rats (p < 0.05). This was accompanied by a tendency towards an increase in the number of enterocytes on the villi of the small intestine in the absence of changes in the content of SGLT1 and GLUT2 in the brush border membrane of the enterocytes. T2D rats also showed lower maltase and alkaline phosphatase (AP) activity in the jejunal mucosa compared to the IGT rats (p < 0.05) and lower AP activity in the colon contents compared to the HFD (p < 0.05) and IGT (p < 0.05) rats. Thus, this study provides insights into the adaptation of the functional and structural parameters of the small intestine in the development of type 2 diabetes and impaired glucose tolerance in young representatives.
Collapse
Affiliation(s)
- Lyudmila V. Gromova
- Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Makarova emb., 199034 Saint-Petersburg, Russia; (L.V.G.); (A.S.P.); (E.V.S.); (A.S.A.); (Y.V.D.)
| | - Alexandr S. Polozov
- Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Makarova emb., 199034 Saint-Petersburg, Russia; (L.V.G.); (A.S.P.); (E.V.S.); (A.S.A.); (Y.V.D.)
| | - Elizaveta V. Savochkina
- Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Makarova emb., 199034 Saint-Petersburg, Russia; (L.V.G.); (A.S.P.); (E.V.S.); (A.S.A.); (Y.V.D.)
| | - Anna S. Alekseeva
- Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Makarova emb., 199034 Saint-Petersburg, Russia; (L.V.G.); (A.S.P.); (E.V.S.); (A.S.A.); (Y.V.D.)
| | - Yulia V. Dmitrieva
- Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Makarova emb., 199034 Saint-Petersburg, Russia; (L.V.G.); (A.S.P.); (E.V.S.); (A.S.A.); (Y.V.D.)
| | - Oleg V. Kornyushin
- Almazov National Medical Research Center, Ministry of Health of the Russian Federation, 2 Akkuratova Str., 197341 Saint-Petersburg, Russia;
| | - Andrey A. Gruzdkov
- Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Makarova emb., 199034 Saint-Petersburg, Russia; (L.V.G.); (A.S.P.); (E.V.S.); (A.S.A.); (Y.V.D.)
- Correspondence: ; Tel.: +7-960-276-3000
| |
Collapse
|
83
|
Meng Q, Culnan DM, Ahmed T, Sun M, Cooney RN. Roux-en-Y gastric bypass alters intestinal glucose transport in the obese Zucker rat. Front Endocrinol (Lausanne) 2022; 13:901984. [PMID: 36034439 PMCID: PMC9405183 DOI: 10.3389/fendo.2022.901984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION The gastrointestinal tract plays a major role in regulating glucose homeostasis and gut endocrine function. The current study examines the effects of Roux-en-Y gastric bypass (RYGB) on intestinal GLP-1, glucose transporter expression and function in the obese Zucker rat (ZR). METHODS Two groups of ZRs were studied: RYGB and sham surgery pair-fed (PF) fed rats. Body weight and food intake were measured daily. On post-operative day (POD) 21, an oral glucose test (OGT) was performed, basal and 30-minute plasma, portal venous glucose and glucagon-like peptide-1 (GLP-1) levels were measured. In separate ZRs, the biliopancreatic, Roux limb (Roux) and common channel (CC) intestinal segments were harvested on POD 21. RESULTS Body weight was decreased in the RYGB group. Basal and 30-minute OGT plasma and portal glucose levels were decreased after RYGB. Basal plasma GLP-1 levels were similar, while a 4.5-fold increase in GLP-1 level was observed in 30-minute after RYGB (vs. PF). The increase in basal and 30-minute portal venous GLP-1 levels after RYGB were accompanied by increased mRNA expressions of proglucagon and PC 1/3, GPR119 protein in the Roux and CC segments. mRNA and protein levels of FFAR2/3 were increased in Roux segment. RYGB decreased brush border glucose transport, transporter proteins (SGLT1 and GLUT2) and mRNA levels of Tas1R1/Tas1R3 and α-gustducin in the Roux and CC segments. CONCLUSIONS Reductions in intestinal glucose transport and enhanced post-prandial GLP-1 release were associated with increases in GRP119 and FFAR2/3 after RYGB in the ZR model. Post-RYGB reductions in the regulation of intestinal glucose transport and L cell receptors regulating GLP-1 secretion represent potential mechanisms for improved glycemic control.
Collapse
Affiliation(s)
- Qinghe Meng
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Derek M. Culnan
- Burn and Reconstructive Centers of America, Jackson, MS, United States
| | - Tamer Ahmed
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Mingjie Sun
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Robert N. Cooney
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
- *Correspondence: Robert N. Cooney,
| |
Collapse
|
84
|
Roberts A, Phuah P, Cheng S, Murphy KG. Targeting Enteroendocrine Cells to Treat Metabolic Disease. COMPREHENSIVE PHARMACOLOGY 2022:344-372. [DOI: 10.1016/b978-0-12-820472-6.00068-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
85
|
Phospholipase C signal mediated the glucose-induced changes of glucose absorption and lipid accumulation in the intestinal epithelial cells of yellow catfish Pelteobagrus fulvidraco. Br J Nutr 2021; 126:1601-1610. [PMID: 33504374 DOI: 10.1017/s0007114521000350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In present study, we explored the effects and the underlying mechanisms of phospholipase C (PLC) mediating glucose-induced changes in intestinal glucose transport and lipid metabolism by using U-73122 (a PLC inhibitor). We found that glucose incubation activated the PLC signal and U-73122 pre-incubation alleviated the glucose-induced increase in plcb2, plce1 and plcg1 mRNA expression. Meanwhile, U-73122 pre-treatment blunted the glucose-induced increase in sodium/glucose co-transporters 1/2 mRNA and protein expressions. U-73122 pre-treatment alleviated the glucose-induced increase in TAG content, BODIPY 493/503 fluorescence intensity, lipogenic enzymes (glucose 6-phospate dehydrogenase (G6PD), 6-phosphogluconate dehydrogenase (6PGD), malic enzyme and fatty acid synthase (FAS)) activity and the mRNA expressions of lipogenic genes and related transcription factors (6pgd, g6pd, fas, acca, srebp1 and carbohydrate response element-binding protein (chrebp)) in intestinal epithelial cells of yellow catfish. Further research found that U-73122 pre-incubation mitigated the glucose-induced increase in the ChREBP protein expression and the acetylation level of ChREBP in HEK293T cells. Taken together, these data demonstrated that the PLC played a major role in the glucose-induced changes of glucose transport and lipid metabolism and provide a new perspective for revealing the molecular mechanism of glucose-induced changes of intestinal glucose absorption, lipid deposition and metabolism.
Collapse
|
86
|
Ho H, Kikuchi K, Oikawa D, Watanabe S, Kanemitsu Y, Saigusa D, Kujirai R, Ikeda‐Ohtsubo W, Ichijo M, Akiyama Y, Aoki Y, Mishima E, Ogata Y, Oikawa Y, Matsuhashi T, Toyohara T, Suzuki C, Suzuki T, Mano N, Kagawa Y, Owada Y, Katayama T, Nakayama T, Tomioka Y, Abe T. SGLT-1-specific inhibition ameliorates renal failure and alters the gut microbial community in mice with adenine-induced renal failure. Physiol Rep 2021; 9:e15092. [PMID: 34921520 PMCID: PMC8683788 DOI: 10.14814/phy2.15092] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/20/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022] Open
Abstract
Sodium-dependent glucose cotransporters (SGLTs) have attracted considerable attention as new targets for type 2 diabetes mellitus. In the kidney, SGLT2 is the major glucose uptake transporter in the proximal tubules, and inhibition of SGLT2 in the proximal tubules shows renoprotective effects. On the other hand, SGLT1 plays a role in glucose absorption from the gastrointestinal tract, and the relationship between SGLT1 inhibition in the gut and renal function remains unclear. Here, we examined the effect of SGL5213, a novel and potent intestinal SGLT1 inhibitor, in a renal failure (RF) model. SGL5213 improved renal function and reduced gut-derived uremic toxins (phenyl sulfate and trimethylamine-N-oxide) in an adenine-induced RF model. Histological analysis revealed that SGL5213 ameliorated renal fibrosis and inflammation. SGL5213 also reduced gut inflammation and fibrosis in the ileum, which is a primary target of SGL5213. Examination of the gut microbiota community revealed that the Firmicutes/Bacteroidetes ratio, which suggests gut dysbiosis, was increased in RF and SGL5213 rebalanced the ratio by increasing Bacteroidetes and reducing Firmicutes. At the genus level, Allobaculum (a major component of Erysipelotrichaceae) was significantly increased in the RF group, and this increase was canceled by SGL5213. We also measured the effect of SGL5213 on bacterial phenol-producing enzymes that catalyze tyrosine into phenol, following the reduction of phenyl sulfate, which is a novel marker and a therapeutic target for diabetic kidney disease DKD. We found that the enzyme inhibition was less potent, suggesting that the change in the microbial community and the reduction of uremic toxins may be related to the renoprotective effect of SGL5213. Because SGL5213 is a low-absorbable SGLT1 inhibitor, these data suggest that the gastrointestinal inhibition of SGLT1 is also a target for chronic kidney diseases.
Collapse
Affiliation(s)
- Hsin‐Jung Ho
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Koichi Kikuchi
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Medical MegabankTohoku UniversitySendaiJapan
| | - Daiki Oikawa
- Department of Biomolecular Engineering Applied Life ChemistryTohoku University Graduate School of EngineeringSendaiJapan
| | - Shun Watanabe
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | | | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Ryota Kujirai
- Laboratory of OncologyPharmacy Practice and SciencesTohoku University Graduate School of Pharmaceutical SciencesSendaiJapan
| | - Wakako Ikeda‐Ohtsubo
- Laboratory of Animal Products ChemistryGraduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| | - Mariko Ichijo
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yukako Akiyama
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yuichi Aoki
- Department of Integrative Genomics, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Eikan Mishima
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshiaki Ogata
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshitsugu Oikawa
- Department of PediatricsTohoku University Graduate School of MedicineSendaiJapan
| | - Tetsuro Matsuhashi
- Department of PediatricsTohoku University Graduate School of MedicineSendaiJapan
| | - Takafumi Toyohara
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Chitose Suzuki
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Takehiro Suzuki
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Nariyasu Mano
- Department of Pharmaceutical SciencesTohoku University HospitalSendaiJapan
| | - Yoshiteru Kagawa
- Department of Organ AnatomyTohoku University Graduate School of MedicineSendaiJapan
| | - Yuji Owada
- Department of Organ AnatomyTohoku University Graduate School of MedicineSendaiJapan
| | - Takane Katayama
- Laboratory of Molecular Biology of BioresponseGraduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Toru Nakayama
- Department of Biomolecular Engineering Applied Life ChemistryTohoku University Graduate School of EngineeringSendaiJapan
| | - Yoshihisa Tomioka
- Laboratory of Molecular Biology of BioresponseGraduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Takaaki Abe
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
87
|
Effect of pecan nuts and extra-virgin olive oil on glycemic profile and nontraditional anthropometric indexes in patients with coronary artery disease: a randomized clinical trial. Eur J Clin Nutr 2021; 76:827-834. [PMID: 34811509 DOI: 10.1038/s41430-021-01045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND/OBJECTIVES The influence of cardioprotective foods on nontraditional indexes related to dysglycemia and body fat distribution is unknown in individuals with coronary artery disease (CAD). This study aimed to evaluate the effect of a healthy diet supplemented with pecan nuts or extra-virgin olive oil on glycemic profile and adipose tissue dysfunction assessed by anthropometric indexes in patients with stable CAD. SUBJECTS/METHODS In a randomized, pragmatic, parallel clinical trial lasting 12 weeks, 204 individuals were allocated to three interventions: a healthy diet (control group [CG], n = 67), a healthy diet plus 30 g/day of pecan nuts (pecan nut group [PNG], n = 68), or a healthy diet plus 30 mL/day of extra-virgin olive oil (olive oil group [OOG], n = 69). Triglyceride-glucose (TyG) index (primary outcome) and other markers of glycemic profile were evaluated, and nontraditional anthropometric indexes as well. Diet quality was assessed according to the Alternate Healthy Eating Index (mAHEI). RESULTS After adjustment for baseline values, use of antidiabetic drugs and insulin, there were no differences in both glycemic and anthropometric profiles according to groups at the end of the study. PNG improved the quality of the diet in comparison to other groups (final mAHEI scores: CG: 19 ± 7.5; PNG: 26 ± 8; OOG: 18.9 ± 6; P < 0.001). CONCLUSIONS There was no difference regarding glycemic and anthropometric parameters according to interventions in patients with stable CAD. However, adding pecan nuts to a healthy diet may improve its quality. Further studies must be conducted considering dietary interventions on secondary cardiovascular prevention setting. CLINICAL TRIALS IDENTIFIER NUMBER NCT02202265. First Posted: July 2014; Last Update: September 2020.
Collapse
|
88
|
Franco YL, Da Silva L, Cristofoletti R. Navigating Through Cell-Based In vitro Models Available for Prediction of Intestinal Permeability and Metabolism: Are We Ready for 3D? AAPS J 2021; 24:2. [PMID: 34811603 PMCID: PMC8925318 DOI: 10.1208/s12248-021-00665-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/23/2021] [Indexed: 01/07/2023] Open
Abstract
Traditionally, in vitro studies to quantify the
intestinal permeability of drugs have relied on two-dimensional cell culture models using human colorectal carcinoma cell lines, namely Caco-2, HT 29 and T84 cells. Although these models have been commonly used for high-throughput screening of xenobiotics in preclinical studies, they do not fully recapitulate the morphology and functionality of enterocytes found in the human intestine in vivo. Efforts to improve the physiological and functional relevance of in vitro intestinal models have led to the development of enteroids/intestinal organoids and microphysiological systems. These models leverage advances in three-dimensional cell culture techniques and stem cell technology (in addition to microfluidics for microphysiological systems), to mimic the architecture and microenvironment of the in vivo intestine more accurately. In this commentary, we will discuss the advantages and limitations of these established and emerging intestinal models, as well as their current and potential future applications for the pre-clinical assessment of oral therapies.
Collapse
Affiliation(s)
- Yesenia L Franco
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Lais Da Silva
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA.
| |
Collapse
|
89
|
Macelline SP, Chrystal PV, Greenhalgh S, Toghyani M, Selle PH, Liu SY. Evaluation of dietary crude protein concentrations, fishmeal, and sorghum inclusions in broiler chickens offered wheat-based diet via Box-Behnken response surface design. PLoS One 2021; 16:e0260285. [PMID: 34797900 PMCID: PMC8604292 DOI: 10.1371/journal.pone.0260285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/08/2021] [Indexed: 11/25/2022] Open
Abstract
The objective of this study was to investigate the impacts of dietary crude protein (CP), fishmeal and sorghum on nutrient utilisation, digestibility coefficients and disappearance rates of starch and protein, amino acid concentrations in systemic plasma and their relevance to growth performance of broiler chickens using the Box-Behnken response surface design. The design consisted of three factors at three levels including dietary CP (190, 210, 230 g/kg), fishmeal (0, 50, 100 g/kg), and sorghum (0, 150, 300 g/kg). A total of 390 male, off-sex Ross 308 chicks were offered experimental diets from 14 to 35 days post-hatch. Growth performance, nutrient utilisation, starch and protein digestibilities and plasma free amino acids were determined. Dietary CP had a negative linear impact on weight gain where the transition from 230 to 190 g/kg CP increased weight gain by 9.43% (1835 versus 2008 g/bird, P = 0.006). Moreover, dietary CP linearly depressed feed intake (r = -0.486. P < 0.001). Fishmeal inclusions had negative linear impacts on weight gain (r = -0.751, P < 0.001) and feed intake (r = -0.495, P < 0.001). There was an interaction between dietary CP and fishmeal for FCR. However, growth performance was not influenced by dietary inclusions of sorghum. Total plasma amino acid concentrations were negatively related to weight gain (r = -0.519, P < 0.0001). The dietary transition from 0 to 100 g/kg fishmeal increased total amino acid concentrations in systemic plasma by 35% (771 versus 1037 μg/mL, P < 0.001). It may be deduced that optimal weight gain (2157 g/bird), optimal feed intake (3330 g/bird) and minimal FCR (1.544) were found in birds offered 190 g/kg CP diets without fishmeal inclusion, irrespective of sorghum inclusions. Both fishmeal and sorghum inclusions did not alter protein and starch digestion rate in broiler chickens; however, moderate reductions in dietary CP could advantage broiler growth performance.
Collapse
Affiliation(s)
- Shemil P. Macelline
- Poultry Research Foundation, The University of Sydney, Camden, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Peter V. Chrystal
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
- Complete Feed Solutions, Hornsby, NSW, Australia; Howick, New Zealand
| | - Shiva Greenhalgh
- Poultry Research Foundation, The University of Sydney, Camden, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Mehdi Toghyani
- Poultry Research Foundation, The University of Sydney, Camden, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Peter H. Selle
- Poultry Research Foundation, The University of Sydney, Camden, NSW, Australia
- Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Sonia Y. Liu
- Poultry Research Foundation, The University of Sydney, Camden, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
90
|
Drosophila Solute Carrier 5A5 Regulates Systemic Glucose Homeostasis by Mediating Glucose Absorption in the Midgut. Int J Mol Sci 2021; 22:ijms222212424. [PMID: 34830305 PMCID: PMC8617630 DOI: 10.3390/ijms222212424] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022] Open
Abstract
The small intestine is the initial site of glucose absorption and thus represents the first of a continuum of events that modulate normal systemic glucose homeostasis. A better understanding of the regulation of intestinal glucose transporters is therefore pertinent to our efforts in curbing metabolic disorders. Using molecular genetic approaches, we investigated the role of Drosophila Solute Carrier 5A5 (dSLC5A5) in regulating glucose homeostasis by mediating glucose uptake in the fly midgut. By genetically knocking down dSLC5A5 in flies, we found that systemic and circulating glucose and trehalose levels are significantly decreased, which correlates with an attenuation in glucose uptake in the enterocytes. Reciprocally, overexpression of dSLC5A5 significantly increases systemic and circulating glucose and trehalose levels and promotes glucose uptake in the enterocytes. We showed that dSLC5A5 undergoes apical endocytosis in a dynamin-dependent manner, which is essential for glucose uptake in the enterocytes. Furthermore, we showed that the dSLC5A5 level in the midgut is upregulated by glucose and that dSLC5A5 critically directs systemic glucose homeostasis on a high-sugar diet. Together, our studies have uncovered the first Drosophila glucose transporter in the midgut and revealed new mechanisms that regulate glucose transporter levels and activity in the enterocyte apical membrane.
Collapse
|
91
|
Tan FPY, Beltranena E, Zijlstra RT. Resistant starch: Implications of dietary inclusion on gut health and growth in pigs: a review. J Anim Sci Biotechnol 2021; 12:124. [PMID: 34784962 PMCID: PMC8597317 DOI: 10.1186/s40104-021-00644-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 10/07/2021] [Indexed: 01/10/2023] Open
Abstract
Starch from cereal grains, pulse grains, and tubers is a major energy substrate in swine rations constituting up to 55% of the diet. In pigs, starch digestion is initiated by salivary and then pancreatic α-amylase, and has as final step the digestion of disaccharides by the brush-border enzymes in the small intestine that produce monosaccharides (glucose) for absorption. Resistant starch (RS) is the proportion of starch that escapes the enzymatic digestion and absorption in the small intestine. The undigested starch reaches the distal small intestine and hindgut for microbial fermentation, which produces short-chain fatty acids (SCFA) for absorption. SCFA in turn, influence microbial ecology and gut health of pigs. These fermentative metabolites exert their benefits on gut health through promoting growth and proliferation of enterocytes, maintenance of intestinal integrity and thus immunity, and modulation of the microbial community in part by suppressing the growth of pathogenic bacteria while selectively enhancing beneficial microbes. Thus, RS has the potential to confer prebiotic effects and may contribute to the improvement of intestinal health in pigs during the post-weaning period. Despite these benefits to the well-being of pigs, RS has a contradictory effect due to lower energetic efficiency of fermented vs. digested starch absorption products. The varying amount and type of RS interact differently with the digestion process along the gastrointestinal tract affecting its energy efficiency and host physiological responses including feed intake, energy metabolism, and feed efficiency. Results of research indicate that the use of RS as prebiotic may improve gut health and thereby, reduce the incidence of post-weaning diarrhea (PWD) and associated mortality. This review summarizes our current knowledge on the effects of RS on microbial ecology, gut health and growth performance in pigs.
Collapse
Affiliation(s)
- Felina P Y Tan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Eduardo Beltranena
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Ruurd T Zijlstra
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada.
| |
Collapse
|
92
|
Zhang Y, Li Y, Xia Q, Liu L, Wu Z, Pan D. Recent advances of cereal β-glucan on immunity with gut microbiota regulation functions and its intelligent gelling application. Crit Rev Food Sci Nutr 2021:1-17. [PMID: 34748438 DOI: 10.1080/10408398.2021.1995842] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
β-glucan from cereals such as wheat, barley, oats and rye are a water-soluble dietary fiber, which are composed of repeating (1→4)-β-bond β-D-glucopyranosyl units and a single (1→3)-β-D-bond separated unit. β-glucan has a series of physicochemical properties (such as viscosity, gelling properties, solubility, etc.), which can be used as a food gel and fat substitute. Its structure endows the healthy functions, including anti-oxidative stress, lowering blood glucose and serum cholesterol, regulating metabolic syndrome and exerting gut immunity via gut microbiota. Due to their unique structural properties and efficacy, cereal β-glucan are not only applied in food substrates in the food industry, but also in food coatings and packaging. This article reviewed the applications of cereal β-glucan in hydrogels, aerogels, intelligent packaging systems and targeted delivery carriers in recent years. Cereal β-glucan in edible film and gel packaging applications are becoming more diversified and intelligent in recent years. Those advances provide a potential solution based on cereal β-glucan as biodegradable substances for immune regulation delivery system and intelligent gelling material in the biomedicine field.
Collapse
Affiliation(s)
- Yunzhen Zhang
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang Province, PR China
| | - Yueqin Li
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang Province, PR China
| | - Qiang Xia
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang Province, PR China
| | - Lianliang Liu
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang Province, PR China
| | - Zufang Wu
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang Province, PR China
| | - Daodong Pan
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang Province, PR China
| |
Collapse
|
93
|
Fiorentino TV, Suraci E, De Vito F, Cimellaro A, Hribal ML, Sciacqua A, Andreozzi F, Luzza F, Sesti G. One-hour post-load hyperglycemia combined with HbA1c identifies individuals with augmented duodenal levels of sodium/glucose co-transporter 1. Diabetes Res Clin Pract 2021; 181:109094. [PMID: 34662689 DOI: 10.1016/j.diabres.2021.109094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 10/20/2022]
Abstract
AIMS Individuals with HbA1c-defined prediabetes (HbA1c 5.7-6.4%) and 1-hour post-load plasma glucose (1hPG) ≥ 155 mg/dl have an increased risk to develop type 2 diabetes (T2DM). T2DM is associated with a higher intestinal expression of sodium/glucose co-transporter 1 (SGLT-1) and glucose transporter 2 (GLUT-2). It is currently unsettled whether HbA1c-defined dysglycemic conditions combined to 1hPG ≥ 155 mg/dl are associated with changes in SGLT-1 and GLUT-2 duodenal abundance. METHODS SGLT-1 and GLUT-2 protein levels were assessed by western blot on duodenal mucosa biopsies of 57 individuals underwent an upper gastrointestinal endoscopy. RESULTS Compared with the normal group (HbA1c < 5.7%), individuals with HbA1c-defined pre-diabetes and diabetes exhibit no significant change in duodenal SGLT-1 abundance. Conversely, duodenal GLUT-2 levels were progressively increased in subjects with prediabetes and diabetes. Stratifying participants according to HbA1c and 1hPG we found that amongst subjects with HbA1c-defined normal or prediabetes condition those having 1hPG ≥ 155 mg/dl displayed higher duodenal levels of SGLT-1 as compared to their counterparts with 1hPG < 155 mg/dl; in contrast to GLUT-2 levels, which were similar between normal and with prediabetes subjects, regardless of 1hPG value. CONCLUSION A value of 1hPG ≥ 155 mg/dl may identify a subset of individuals within HbA1c-defined glycemic categories having a higher duodenal abundance of SGLT-1.
Collapse
Affiliation(s)
- Teresa Vanessa Fiorentino
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Evelina Suraci
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Francesca De Vito
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Antonio Cimellaro
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Marta Letizia Hribal
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Francesco Luzza
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, University of Rome-Sapienza, Rome 00189, Italy.
| |
Collapse
|
94
|
Kozieł MJ, Ziaja M, Piastowska-Ciesielska AW. Intestinal Barrier, Claudins and Mycotoxins. Toxins (Basel) 2021; 13:758. [PMID: 34822542 PMCID: PMC8622050 DOI: 10.3390/toxins13110758] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 02/08/2023] Open
Abstract
The intestinal barrier is the main barrier against all of the substances that enter the body. Proper functioning of this barrier guarantees maintained balance in the organism. Mycotoxins are toxic, secondary fungi metabolites, that have a negative impact both on human and animal health. It was postulated that various mycotoxins may affect homeostasis by disturbing the intestinal barrier. Claudins are proteins that are involved in creating tight junctions between epithelial cells. A growing body of evidence underlines their role in molecular response to mycotoxin-induced cytotoxicity. This review summarizes the information connected with claudins, their association with an intestinal barrier, physiological conditions in general, and with gastrointestinal cancers. Moreover, this review also includes information about the changes in claudin expression upon exposition to various mycotoxins.
Collapse
|
95
|
Burman A, Kaji I. Luminal Chemosensory Cells in the Small Intestine. Nutrients 2021; 13:nu13113712. [PMID: 34835968 PMCID: PMC8620795 DOI: 10.3390/nu13113712] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/23/2022] Open
Abstract
In addition to the small intestine's well-known function of nutrient absorption, the small intestine also plays a major role in nutrient sensing. Similar to taste sensors seen on the tongue, GPCR-coupled nutrient sensors are expressed throughout the intestinal epithelium and respond to nutrients found in the lumen. These taste receptors respond to specific ligands, such as digested carbohydrates, fats, and proteins. The activation of nutrient sensors in the intestine allows for the induction of signaling pathways needed for the digestive system to process an influx of nutrients. Such processes include those related to glucose homeostasis and satiety. Defects in intestinal nutrient sensing have been linked to a variety of metabolic disorders, such as type 2 diabetes and obesity. Here, we review recent updates in the mechanisms related to intestinal nutrient sensors, particularly in enteroendocrine cells, and their pathological roles in disease. Additionally, we highlight the emerging nutrient sensing role of tuft cells and recent work using enteroids as a sensory organ model.
Collapse
Affiliation(s)
- Andreanna Burman
- Cell and Developmental Biology and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Izumi Kaji
- Epithelial Biology Center and Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
96
|
Liang W, Lan Y, Chen C, Song M, Xiao J, Huang Q, Cao Y, Ho CT, Lu M. Modulating effects of capsaicin on glucose homeostasis and the underlying mechanism. Crit Rev Food Sci Nutr 2021:1-19. [PMID: 34657531 DOI: 10.1080/10408398.2021.1991883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Abnormal glucose homeostasis is linked to a variety of metabolic syndromes, such as insulin resistance, obesity, type-2 diabetes mellitus, hypertension and cardiovascular diseases. Maintenance of normal glucose homeostasis is important for the body to keep normal biological functions. As the major bioactive ingredient in chili peppers responsible for the pungent flavor, capsaicin has been reported to effectively improve glucose homeostasis with low cytotoxicity. In this review, the modulating effects of capsaicin on glucose homeostasis in cell models, animal models and human trials are summarized through both TRPV1 dependent and TRPV1 independent pathways. The relevant molecular mechanisms underlying its regulatory effects are also evaluated. Understanding the effects and mechanisms of capsaicin on glucose metabolism could provide theoretical evidence for its application in the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Wanxia Liang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Yaqi Lan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Chengyu Chen
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou, China
| | - Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Qingrong Huang
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
97
|
Exogenous Enzymes Influenced Eimeria-Induced Changes in Cecal Fermentation Profile and Gene Expression of Nutrient Transporters in Broiler Chickens. Animals (Basel) 2021; 11:ani11092698. [PMID: 34573663 PMCID: PMC8470256 DOI: 10.3390/ani11092698] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/17/2022] Open
Abstract
Two 21-day experiments were conducted to investigate the effects of exogenous enzymes on growth performance, tight junctions, and nutrient transporters, jejunal oligosaccharides and cecal short-chain fatty acids (SCFA) of broiler chickens challenged with mixed Eimeria. Two different basal diets: high fiber-adequate protein (HFAP; Expt. 1) or low fiber-low protein (LFLP; Expt. 2) were used in the two experiments. In each experiment, birds were allocated to four treatments in a 2 × 2 factorial arrangement (with or without protease and xylanase combination; with or without Eimeria challenge). In Expt. 1, with HFAP diets, Eimeria upregulated (p < 0.05) the expression of claudin-1, but downregulated (p < 0.05) glucose transporters GLUT2/GLUT5. On the contrary, enzymes downregulated (p < 0.05) claudin-1 and alleviated the Eimeria-depressed GLUT2/GLUT5 expression. In both experiments, Eimeria decreased (p < 0.05) cecal saccharolytic SCFA and increased (p < 0.05) cecal branched-chain fatty acids. The challenge × enzyme interaction (p < 0.05) showed that enzymes reversed the Eimeria effects on fermentation pattern shift. In conclusion, Eimeria altered tight junctions and nutrient transporters expression promoted cecal proteolytic fermentation and inhibited saccharolytic fermentation. Exogenous enzymes showed the potential of alleviating the Eimeria-induced intestinal gene expression changes and reversing the unfavorable cecal fermentation pattern.
Collapse
|
98
|
Guo Z, DeLoid GM, Cao X, Bitounis D, Sampathkumar K, Woei Ng K, Joachim Loo SC, Philip D. Effects of ingested nanocellulose and nanochitosan materials on carbohydrate digestion and absorption in an in vitro small intestinal epithelium model. ENVIRONMENTAL SCIENCE. NANO 2021; 8:2554-2568. [PMID: 34840801 PMCID: PMC8622715 DOI: 10.1039/d1en00233c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Nanoscale materials derived from natural biopolymers like cellulose and chitosan have many potentially useful agri-food and oral drug delivery applications. Because of their large and potentially bioactive surface areas and other unique physico-chemical properties, it is essential when evaluating their toxicological impact to assess potential effects on the digestion and absorption of co-ingested nutrients. Here, the effects of cellulose nanofibers (CNF), cellulose nanocrystals (CNC), and chitosan nanoparticles (Chnp) on the digestion and absorption of carbohydrates were studied. Starch digestion was assessed by measuring maltose released during simulated digestion of starch solutions. Glucose absorption was assessed by measuring translocation from the resulting digestas across an in vitro transwell tri-culture model of the small intestinal epithelium and calculating the area under the curve increase in absorbed glucose, analogous to the glycemic index. At 1% w/w, CNF and Chnp had small but significant effects (11% decrease and 14% increase, respectively) and CNC had no effect on starch hydrolysis during simulated digestion of a 1% w/w rice starch solution. In addition, at 2% w/w CNC had no effect on amylolysis in 1% solutions of either rice, corn, or wheat starch. Similarly, absorption of glucose from digestas of starch solutions (i.e., from maltose), was unaffected by 1% w/w CNF or CNC, but was slightly increased (10%, p<0.05) by 1% Chnp, possibly due to the slightly higher maltose concentration in the Chnp-containing digestas. In contrast, all of the test materials caused sharp increases (~1.2, 1.5, and 1.6 fold for CNC, CNF, and Chnp, respectively) in absorption of glucose from starch-free digestas spiked with free glucose at a concentration corresponding to complete hydrolysis of 1% w/w starch. The potential for ingested cellulose and chitosan nanomaterials to increase glucose absorption could have important health implications. Further studies are needed to elucidate the mechanisms underlying the observed increases and to evaluate the potential glycemic effects in an intact in vivo system.
Collapse
Affiliation(s)
- Zhongyuan Guo
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Glen M DeLoid
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Xiaoqiong Cao
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Dimitrios Bitounis
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Kaarunya Sampathkumar
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798, Singapore, Singapore
| | - Kee Woei Ng
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798, Singapore, Singapore
- Skin Research Institute of Singapore, 8A Biomedical Grove, #06-06 Immunos, 138648, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, 1 Cleantech Loop, CleanTech One, Singapore 637141
| | - Say Chye Joachim Loo
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798, Singapore, Singapore
| | - Demokritou Philip
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798, Singapore, Singapore
| |
Collapse
|
99
|
Ontawong A, Duangjai A, Srimaroeng C. Coffea arabica bean extract inhibits glucose transport and disaccharidase activity in Caco-2 cells. Biomed Rep 2021; 15:73. [PMID: 34405045 PMCID: PMC8329997 DOI: 10.3892/br.2021.1449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/14/2021] [Indexed: 11/06/2022] Open
Abstract
The major constituents of Coffea arabica (coffee), including caffeine, chlorogenic acid and caffeic acid, exhibit antihyperglycemic properties in in vitro and in vivo models. However, whether Coffea arabica bean extract (CBE) regulates glucose uptake activity and the underlying mechanisms involved remain unclear. The aim of the present study was to examine the effects of CBE on glucose absorption and identify the mechanisms involved using an in vitro model. The uptake of a fluorescent glucose analog into Caco-2 colorectal adenocarcinoma cells was determined. The expression levels of sodium glucose co-transporter 1 (SGLT1) and glucose transporter 2 (GLUT2) were evaluated. In addition, glycoside hydrolase enzyme activity was investigated. It was observed that CBE inhibited disaccharidase enzyme activity. Furthermore, CBE exerted an inhibitory effect on intestinal glucose absorption by downregulating SGLT1- and GLUT2-mediated 5' AMP-activated protein kinase phosphorylation and suppressing hepatocyte nuclear factor 1α expression. These data suggest that CBE may attenuate glucose absorption and may have potentially beneficial antihyperglycemic effects in the body; however, the mechanisms underlying the effects of CBE must be elucidated through further investigation.
Collapse
Affiliation(s)
- Atcharaporn Ontawong
- Division of Physiology, School of Medical Sciences, University of Phayao, Muang Phayao, Phayao 56000, Thailand
| | - Acharaporn Duangjai
- Division of Physiology, School of Medical Sciences, University of Phayao, Muang Phayao, Phayao 56000, Thailand
| | - Chutima Srimaroeng
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Nong Khai 52000, Thailand
| |
Collapse
|
100
|
Zietek T, Boomgaarden WAD, Rath E. Drug Screening, Oral Bioavailability and Regulatory Aspects: A Need for Human Organoids. Pharmaceutics 2021; 13:1280. [PMID: 34452240 PMCID: PMC8399541 DOI: 10.3390/pharmaceutics13081280] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/31/2022] Open
Abstract
The intestinal epithelium critically contributes to oral bioavailability of drugs by constituting an important site for drug absorption and metabolism. In particular, intestinal epithelial cells (IEC) actively serve as gatekeepers of drug and nutrient availability. IECs' transport processes and metabolism are interrelated to the whole-body metabolic state and represent potential points of origin as well as therapeutic targets for a variety of diseases. Human intestinal organoids represent a superior model of the intestinal epithelium, overcoming limitations of currently used in vitro models. Caco-2 cells or rodent explant models face drawbacks such as their cancer and non-human origin, respectively, but are commonly used to study intestinal nutrient absorption, enterocyte metabolism and oral drug bioavailability, despite poorly correlative data. In contrast, intestinal organoids allow investigating distinct aspects of bioavailability including spatial resolution of transport, inter-individual differences and high-throughput screenings. As several countries have already developed strategic roadmaps to phase out animal experiments for regulatory purposes, intestinal organoid culture and organ-on-a-chip technology in combination with in silico approaches are roads to go in the preclinical and regulatory setup and will aid implementing the 3Rs (reduction, refinement and replacement) principle in basic science.
Collapse
Affiliation(s)
- Tamara Zietek
- Doctors against Animal Experiments, 51143 Köln, Germany
| | | | - Eva Rath
- Chair of Nutrition and Immunology, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|