1
|
Feng S, Wang J, Peng Q, Zhang P, Jiang Y, Zhang H, Song X, Li Y, Huang W, Zhang D, Deng C. Schisandra sphenanthera extract modulates sweet taste receptor pathway, IRS/PI3K, AMPK/mTOR pathway and endogenous metabolites against T2DM. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156348. [PMID: 39740377 DOI: 10.1016/j.phymed.2024.156348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/20/2024] [Accepted: 12/24/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Southern Schisandra is the dried and matured fruit of Schisandra sphenanthera Rehd. et Wils. in the family of Magnoliaceae; Traditional medicine reports that Schisandra sphenanthera has astringent and astringent properties, benefiting qi and promoting the production of body fluid, tranquilising the heart and calming the mind; it is clinically utilized for prolonged cough, thirst due to injury of the body fluid, internal heat and thirst, palpitation and insomnia, etc., and thirst belongs to the category of diabetes mellitus; the literature reports and the preliminary study of our team showed that Schisandra sphenanthera can be used to prevent and control diabetes mellitus. PURPOSE In the research, we investigated the mechanism of action of SDP against T2DM by integrating pharmacodynamics, endogenous metabolite assays and signalling pathways. MATERIALS AND METHODS UPLC-MS/MS was used to identify the chemical constituents. HPLC was utilized to determine the content of eight lignan-like components in SDP. A T2DM rat model was established by the combined induction of high-fat and high-sugar feed and STZ, and the mechanism of action of SDP on T2DM was investigated by using biochemical indices, Western blot analysis of protein expression, mRNA expression, immunohistochemistry and endogenous metabolites. RESULTS The chemical components in SDP were determined by UPLC-MS/MS and HPLC, and biochemical indicators determined that SDP has the effects of lowering blood glucose, anti-glycolipid metabolism, and anti-oxidative stress, and is able to restore pathological damage in the liver and pancreas, activate the PI3K/AKT, AMPK/mTOR, and sweetness receptor signalling pathways, restore the sweetness receptor mRNAs, and modulate the urinary compounds such as malic acid, γ-aminobutyric acid, leucine, N-acetylaspartic acid and other compounds thereby achieving the therapeutic effect of T2DM. CONCLUSION SDP can ameliorate diabetes-induced symptoms related to elevated blood glucose, dyslipidaemia, elevated fasting insulin levels and impaired glucose tolerance in rats; the anti-T2DM of SDP may be through the regulation of the sweet taste receptor pathway, the PI3K/AKT/mTOR and the AMPK/mTOR signalling pathway, which leads to the development of a normal level and exerts an antidiabetic effect.
Collapse
Affiliation(s)
- Shibo Feng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Jiaojiao Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Qin Peng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Panpan Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Yi Jiang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Huawei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Xiaomei Song
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Yuze Li
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Wenli Huang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Dongdong Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Chong Deng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China.
| |
Collapse
|
2
|
Yoshida R, Ninomiya Y. Mechanisms and Functions of Sweet Reception in Oral and Extraoral Organs. Int J Mol Sci 2024; 25:7398. [PMID: 39000505 PMCID: PMC11242429 DOI: 10.3390/ijms25137398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The oral detection of sugars relies on two types of receptor systems. The first is the G-protein-coupled receptor TAS1R2/TAS1R3. When activated, this receptor triggers a downstream signaling cascade involving gustducin, phospholipase Cβ2 (PLCβ2), and transient receptor potential channel M5 (TRPM5). The second type of receptor is the glucose transporter. When glucose enters the cell via this transporter, it is metabolized to produce ATP. This ATP inhibits the opening of KATP channels, leading to cell depolarization. Beside these receptor systems, sweet-sensitive taste cells have mechanisms to regulate their sensitivity to sweet substances based on internal and external states of the body. Sweet taste receptors are not limited to the oral cavity; they are also present in extraoral organs such as the gastrointestinal tract, pancreas, and brain. These extraoral sweet receptors are involved in various functions, including glucose absorption, insulin release, sugar preference, and food intake, contributing to the maintenance of energy homeostasis. Additionally, sweet receptors may have unique roles in certain organs like the trachea and bone. This review summarizes past and recent studies on sweet receptor systems, exploring the molecular mechanisms and physiological functions of sweet (sugar) detection in both oral and extraoral organs.
Collapse
Affiliation(s)
- Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Yuzo Ninomiya
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
- Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Zhu S, Li J, Li Z, Wang Z, Wei Q, Shi F. Effects of non-nutritive sweeteners on growth and intestinal health by regulating hypothalamic RNA profile and ileum microbiota in guinea pigs. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:4342-4353. [PMID: 38328855 DOI: 10.1002/jsfa.13320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Non-nutritive sweeteners (NNS) are commonly used in sweetened foods and beverages; however their role in metabolic regulation is still not clear. In this experiment, we used guinea pigs as an animal model to study the effect of NNS on body growth and intestinal health by modifying gut microbiota and hypothalamus-related proteins. RESULTS For a 28-day feeding experiment a total of 40 guinea pigs were randomly divided into four groups, one control (CN) group and three treatments, in which three NNS were added to the diet: rebaudioside A (RA, 330 mg kg-1), sodium saccharin (SS, 800 mg kg-1), and sucralose (TGS, 167 mg kg-1), respectively. The TGS group exhibited significantly reduced food consumption in comparison with the CN group (P < 0.05) whereas the RA group showed increased food consumption in comparison with the CN group (P < 0.05). Notably, Taste receptor type 1 subunit 2 (T1R2) expression in the hypothalamus was significantly higher in the RA group than in the CN group (P < 0.05). The mRNA expressions of appetite-stimulated genes arouti-related neuropeptide (AGRP), neuropeptide Y (NPY), and thyroid stimulating hormone (TSHB) were significantly higher than those in the CN group (P < 0.05) but mRNA expressions of appetite-suppressed genes tryptophan hydroxylase 2(THP2) were significantly lower in the TGS group (P < 0.05). Furthermore, NNS in the guinea pig diets (RA, SS, TGS) significantly increased the relative abundance of Muribaculaceae but decreased the relative abundance of Clostridia_vadin BB60 in comparison with the CN group (P < 0.05). We also found that dietary supplementation with RA also significantly altered the relative abundance of Lactobacillus. CONCLUSION Our finding confirmed that dietary supplementation with RA and TGS affected body growth and intestinal health by modulating hypothalamic RNA profiles and ileum microbiota, suggesting that NNS should be included in guinea-pig feeding. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shanli Zhu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- College of Agriculture, Jinhua Polytechnic, Jinhua, China
| | - Junrong Li
- College of Agriculture, Jinhua Polytechnic, Jinhua, China
| | - Ziqing Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhe Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Quanwei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Fangxiong Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
4
|
Posta E, Fekete I, Gyarmati E, Stündl L, Zold E, Barta Z. The Effects of Artificial Sweeteners on Intestinal Nutrient-Sensing Receptors: Dr. Jekyll or Mr. Hyde? Life (Basel) 2023; 14:10. [PMID: 38276259 PMCID: PMC10817473 DOI: 10.3390/life14010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
The consumption of artificial and low-calorie sweeteners (ASs, LCSs) is an important component of the Western diet. ASs play a role in the pathogenesis of metabolic syndrome, dysbiosis, inflammatory bowel diseases (IBDs), and various inflammatory conditions. Intestinal nutrient-sensing receptors act as a crosstalk between dietary components, the gut microbiota, and the regulation of immune, endocrinological, and neurological responses. This narrative review aimed to summarize the possible effects of ASs and LCSs on intestinal nutrient-sensing receptors and their related functions. Based on the findings of various studies, long-term AS consumption has effects on the gut microbiota and intestinal nutrient-sensing receptors in modulating incretin hormones, antimicrobial peptides, and cytokine secretion. These effects contribute to the regulation of glucose metabolism, ion transport, gut permeability, and inflammation and modulate the gut-brain, and gut-kidney axes. Based on the conflicting findings of several in vitro, in vivo, and randomized and controlled studies, artificial sweeteners may have a role in the pathogenesis of IBDs, functional bowel diseases, metabolic syndrome, and cancers via the modulation of nutrient-sensing receptors. Further studies are needed to explore the exact mechanisms underlying their effects to decide the risk/benefit ratio of sugar intake reduction via AS and LCS consumption.
Collapse
Affiliation(s)
- Edit Posta
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (E.G.); (Z.B.)
| | - Istvan Fekete
- Institute of Food Technology, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi út 138, 4032 Debrecen, Hungary; (I.F.); (L.S.)
| | - Eva Gyarmati
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (E.G.); (Z.B.)
- Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei Blvd. 98, 4032 Debrecen, Hungary
| | - László Stündl
- Institute of Food Technology, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi út 138, 4032 Debrecen, Hungary; (I.F.); (L.S.)
| | - Eva Zold
- Department of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, Móricz Zsigmond Str. 22, 4032 Debrecen, Hungary;
| | - Zsolt Barta
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (E.G.); (Z.B.)
| |
Collapse
|
5
|
Song JW, Lee KH, Seong H, Shin DM, Shon WJ. Taste receptor type 1 member 3 enables western diet-induced anxiety in mice. BMC Biol 2023; 21:243. [PMID: 37926812 PMCID: PMC10626698 DOI: 10.1186/s12915-023-01723-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Accumulating evidence supports that the Western diet (WD), a diet high in saturated fat and sugary drinks, contributes to the pathogenesis of anxiety disorders, which are the most prevalent mental disorders worldwide. However, the underlying mechanisms by which WD causes anxiety remain unclear. Abundant expression of taste receptor type 1 member 3 (TAS1R3) has been identified in the hypothalamus, a key brain area involved in sensing peripheral nutritional signals and regulating anxiety. Thus, we investigated the influence of excessive WD intake on anxiety and mechanisms by which WD intake affects anxiety development using wild-type (WT) and Tas1r3 deficient (Tas1r3-/-) mice fed a normal diet (ND) or WD for 12 weeks. RESULTS WD increased anxiety in male WT mice, whereas male Tas1r3-/- mice were protected from WD-induced anxiety, as assessed by open field (OF), elevated plus maze (EPM), light-dark box (LDB), and novelty-suppressed feeding (NSF) tests. Analyzing the hypothalamic transcriptome of WD-fed WT and Tas1r3-/- mice, we found 1,432 genes significantly up- or down-regulated as a result of Tas1r3 deficiency. Furthermore, bioinformatic analysis revealed that the CREB/BDNF signaling-mediated maintenance of neuronal regeneration, which can prevent anxiety development, was enhanced in WD-fed Tas1r3-/- mice compared with WD-fed WT mice. Additionally, in vitro studies further confirmed that Tas1r3 knockdown prevents the suppression of Creb1 and of CREB-mediated BDNF expression caused by high levels of glucose, fructose, and palmitic acid in hypothalamic neuronal cells. CONCLUSIONS Our results imply that TAS1R3 may play a key role in WD-induced alterations in hypothalamic functions, and that inhibition of TAS1R3 overactivation in the hypothalamus could offer therapeutic targets to alleviate the effects of WD on anxiety.
Collapse
Affiliation(s)
- Jae Won Song
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Keon-Hee Lee
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Hobin Seong
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| | - Woo-Jeong Shon
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
6
|
Favalier N, Roy J, Dias K, Maunas P, Turonnet N, Conde-Sieira M, Panserat S, Soengas JL, Marandel L. Sex dimorphism of glucosensing parameters and appetite-regulating peptides in the hypothalamus of rainbow trout broodstocks. Comp Biochem Physiol A Mol Integr Physiol 2023; 281:111436. [PMID: 37085140 DOI: 10.1016/j.cbpa.2023.111436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023]
Abstract
Rainbow trout (Oncorhynchus mykiss) is traditionally considered as a poor user of digestible carbohydrates harbouring persistent postprandial hyperglycaemia and decreased growth performances when fed a diet containing more than 20% of digestible carbohydrates. While this glucose-intolerant phenotype is well-described in juveniles, evidence points to a particular regulation of glucose metabolism in rainbow trout broodstrocks. By detecting changes in glucose levels and triggering a specific metabolic response, the hypothalamus plays a key role in the regulation of peripheral glucose metabolism. Therefore, our objective was to assess, for the first time in fish, the short-term consequences in hypothalamus, the glucose sensing and feed intake regulation of feeding mature female and male, and neomale rainbow trout with a diet containing either no or a 33% carbohydrate. The hypothalamic glucosensing capacity was assessed through mRNA levels of glucosensing related-genes and feed intake regulation through appetite-regulating peptides. Our data indicate that a brief period of carbohydrate intake (5 meals at 8 °C) did not induce specific changes in glucosensing capacity and appetite-regulating peptides in the hypothalamus of rainbow trout broodstock. Our results did however demonstrate, for the first time in fish, the existence of sex dimorphism of glucosensing-related genes and appetite-regulating peptides.
Collapse
Affiliation(s)
- Nathan Favalier
- INRAE, Université de Pau et des Pays de l'Adour, e2s, St-Pee-sur-Nivelle, France
| | - Jérôme Roy
- INRAE, Université de Pau et des Pays de l'Adour, e2s, St-Pee-sur-Nivelle, France
| | - Karine Dias
- INRAE, Université de Pau et des Pays de l'Adour, e2s, St-Pee-sur-Nivelle, France
| | - Patrick Maunas
- INRAE, Université de Pau et des Pays de l'Adour, e2s, St-Pee-sur-Nivelle, France
| | - Nicolas Turonnet
- INRAE, Université de Pau et des Pays de l'Adour, e2s, St-Pee-sur-Nivelle, France
| | - Marta Conde-Sieira
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, E-36310 Vigo, Spain
| | - Stephane Panserat
- INRAE, Université de Pau et des Pays de l'Adour, e2s, St-Pee-sur-Nivelle, France
| | - José Luis Soengas
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, E-36310 Vigo, Spain
| | - Lucie Marandel
- INRAE, Université de Pau et des Pays de l'Adour, e2s, St-Pee-sur-Nivelle, France.
| |
Collapse
|
7
|
Hu R, Jiang X, Yang H, Liu G. Selection signature analysis reveals RDH5 performed key function in vision during sheep domestication process. Arch Anim Breed 2023. [DOI: 10.5194/aab-66-81-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Abstract. As one of the most successful domesticated animals in the Neolithic age,
sheep gradually migrated all over the world with human activities. During the
domestication process, remarkable changes have taken place in morphology,
physiology, and behavior, resulting in different breeds with different
characters via artificial and natural selection. However, the genetic
background responsible for these phenotypic variations remains largely
unclear. Here, we used whole genome resequencing technology to compare and
analyze the genome differences between Asiatic mouflon wild sheep (Ovis orientalis) and Hu
sheep (Ovis aries). A total of 755 genes were positively selected in the process of
domestication and selection, and the genes related to sensory perception had
directional evolution in the autosomal region, such as OPRL1, LEF1, TAS1R3, ATF6, VSX2, MYO1A, RDH5, and some novel
genes. A missense mutation of c.T722C/p.M241T in exon 4 of RDH5 existing in sheep
were found, and the T allele was completely fixed in Hu sheep. In addition, the
mutation with the C allele reduced the retinol dehydrogenase activity encoding
by RDH5, which can impair retinoic acid metabolism and further influenced the visual
cycle. Overall, our results showed significant enrichment for positively
selected genes involved in sensory perception development during sheep
domestication; RDH5 and its variants may be related to the retinal degeneration
in sheep. We infer that the wild sheep ancestors with weaker visual sensitivity
were weeded out by humans, and the mutation was selective, swept by the dual
pressures of natural and artificial selection.
Collapse
|
8
|
Lu MY, Liu TW, Liang PC, Huang CI, Tsai YS, Tsai PC, Ko YM, Wang WH, Lin CC, Chen KY, Wang SC, Wei YJ, Hsu PY, Jang TY, Hsieh MY, Wang CW, Yeh ML, Lin ZY, Huang CF, Huang JF, Dai CY, Chuang WL, Yu ML. Decision tree algorithm predicts hepatocellular carcinoma among chronic hepatitis C patients following viral eradication. Am J Cancer Res 2023; 13:190-203. [PMID: 36777503 PMCID: PMC9906075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/03/2023] [Indexed: 02/14/2023] Open
Abstract
Successful eradication of the hepatitis C virus (HCV) cannot eliminate the risk of hepatocellular carcinoma (HCC). Next-generation RNA sequencing provides comprehensive genomic insights into the pathogenesis of HCC. Artificial intelligence has opened a new era in precision medicine. This study integrated clinical features and genetic biomarkers to establish a machine learning-based HCC model following viral eradication. A prospective cohort of 55 HCV patients with advanced fibrosis, who achieved a sustained virologic response after antiviral therapy, was enrolled. The primary outcome was the occurrence of HCC. The genomic signatures of peripheral blood mononuclear cells (PBMC) were determined by RNA sequencing at baseline and 24 weeks after end-of-treatment. Machine learning algorithms were implemented to extract the predictors of HCC. HCC occurred in 8 of the 55 patients, with an annual incidence of 2.7%. Pretreatment PBMC DEFA1B, HBG2, ADCY4, and posttreatment TAS1R3, ABCA3, and FOSL1 genes were significantly downregulated, while the pretreatment ANGPTL6 gene was significantly upregulated in the HCC group compared to that in the non-HCC group. A gene score derived from the result of the decision tree algorithm can identify HCC with an accuracy of 95.7%. Gene score = TAS1R3 (≥0.63 FPKM, yes/no = 0/1) + FOSL1 (≥0.27 FPKM, yes/no = 0/1) + ABCA3 (≥2.40 FPKM, yes/no = 0/1). Multivariate Cox regression analysis showed that this gene score was the most important predictor of HCC (hazard ratio = 2.38, 95% confidence interval [CI] = 1.06-5.36, P = 0.036). Combining the gene score and fibrosis-4 index, a nomogram was constructed to predict the probability of HCC with an area under the receiver operating characteristic curve up to 0.950 (95% CI = 0.888-1.000, P = 7.0 × 10-5). Decision curve analysis revealed that the nomogram had a net benefit in HCC detection. The calibration curve showed that the nomogram had optimal concordance between the predicted and actual HCC probabilities. In conclusion, down-regulated posttreatment PBMC TAS1R3, ABCA3, and FOSL1 expression were significantly correlated with HCC development after HCV eradication. Decision-tree-based algorithms can refine the assessment of HCC risk for personalized HCC surveillance.
Collapse
Affiliation(s)
- Ming-Ying Lu
- School of Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-Sen UniversityKaohsiung, Taiwan
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Ta-Wei Liu
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Po-Cheng Liang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Ching-I Huang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Yi-Shan Tsai
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Pei-Chien Tsai
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Yu-Min Ko
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Wen-Hsuan Wang
- Department of Nursing, Kaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Ching-Chih Lin
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Kuan-Yu Chen
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Yu-Ju Wei
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Po-Yao Hsu
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Tyng-Yuan Jang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Ming-Yen Hsieh
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Chih-Wen Wang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Ming-Lun Yeh
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Zu-Yau Lin
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Chung-Feng Huang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Jee-Fu Huang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Chia-Yen Dai
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Wan-Long Chuang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Ming-Lung Yu
- School of Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-Sen UniversityKaohsiung, Taiwan
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial HospitalKaohsiung, Taiwan
- National Pingtung University of Science and TechnologyPingtung, Taiwan
| |
Collapse
|
9
|
Yanagisawa Y. How dietary amino acids and high protein diets influence insulin secretion. Physiol Rep 2023; 11:e15577. [PMID: 36695783 PMCID: PMC9875820 DOI: 10.14814/phy2.15577] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/26/2022] [Accepted: 12/31/2022] [Indexed: 01/26/2023] Open
Abstract
Glucose homeostasis is the maintenance and regulation of blood glucose concentration within a tight physiological range, essential for the functioning of most tissues and organs. This is primarily achieved by pancreatic secretion of insulin and glucagon. Deficient pancreatic endocrine function, coupled with or without peripheral insulin resistance leads to prolonged hyperglycemia with chronic impairment of glucose homeostasis, most commonly seen in diabetes mellitus. High protein diets (HPDs) are thought to modulate glucose homeostasis through various metabolic pathways. Insulin secretion can be directly modulated by the amino acid products of protein digestion, which activate nutrient receptors and nutrient transporters expressed by the endocrine pancreas. Insulin secretion can also be modulated indirectly, through incretin release from enteroendocrine cells, and via vagal neuronal pathways. Additionally, glucose homeostasis can be promoted by the satiating effects of anorectic hormones released following HPD consumption. This review summarizes the insulinotropic mechanisms by which amino acids and HPDs may influence glucose homeostasis, with a particular focus on their applicability in the management of Type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yuuki Yanagisawa
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| |
Collapse
|
10
|
Wu HT, Lin CH, Pai HL, Chen YC, Cheng KP, Kuo HY, Li CH, Ou HY. Sucralose, a Non-nutritive Artificial Sweetener Exacerbates High Fat Diet-Induced Hepatic Steatosis Through Taste Receptor Type 1 Member 3. Front Nutr 2022; 9:823723. [PMID: 35685876 PMCID: PMC9171434 DOI: 10.3389/fnut.2022.823723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease globally, and it is strongly associated with obesity. To combat obesity, artificial sweeteners are often used to replace natural sugars, and sucralose is one of the most extensively used sweeteners. It was known that sucralose exerted effects on lipid metabolism dysregulation, and hepatic inflammation; however, the effects of sucralose on hepatic steatosis were still obscure. In this study, we found that supplements of sucralose enhanced high-fat-diet (HFD)-induced hepatic steatosis. In addition, treatment of sucralose increased reactive oxygen species (ROS) generation and induced endoplasmic reticulum (ER) stress in HepG2 cells. Pretreatment of ROS or ER stress inhibitors reversed the effects of sucralose on lipogenesis. Furthermore, pretreatment of taste receptor type 1 membrane 3 (T1R3) inhibitor or T1R3 knockdown reversed sucralose-induced lipogenesis in HepG2 cells. Taken together, sucralose might activate T1R3 to generate ROS and promote ER stress and lipogenesis, and further accelerate to the development of hepatic steatosis.
Collapse
Affiliation(s)
- Hung-Tsung Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Ching-Han Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Hsiu-Ling Pai
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei City, Taiwan
| | - Yi-Cheng Chen
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Kai-Pi Cheng
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Hsin-Yu Kuo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chung-Hao Li
- Department of Family Medicine, Tainan Municipal An-Nan Hospital, China Medical University, Tainan City, Taiwan
| | - Horng-Yih Ou
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
- *Correspondence: Horng-Yih Ou,
| |
Collapse
|
11
|
Dong H, Liu J, Zhu J, Zhou Z, Tizzano M, Peng X, Zhou X, Xu X, Zheng X. Oral Microbiota-Host Interaction Mediated by Taste Receptors. Front Cell Infect Microbiol 2022; 12:802504. [PMID: 35425718 PMCID: PMC9004699 DOI: 10.3389/fcimb.2022.802504] [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: 10/26/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Taste receptors, originally identified in taste buds, function as the periphery receptors for taste stimuli and play an important role in food choice. Cohort studies have revealed that single nucleotide polymorphisms of taste receptors such as T1R1, T1R2, T2R38 are associated with susceptibility to oral diseases like dental caries. Recent studies have demonstrated the wide expression of taste receptors in various tissues, including intestinal epithelia, respiratory tract, and gingiva, with an emerging role of participating in the interaction between mucosa surface and microorganisms via monitoring a wide range of metabolites. On the one hand, individuals with different oral microbiomes exhibited varied taste sensitivity, suggesting a potential impact of the oral microbiota composition on taste receptor function. On the other hand, animal studies and in vitro studies have uncovered that a variety of oral cells expressing taste receptors such as gingival solitary chemosensory cells, gingival epithelial cells (GECs), and gingival fibroblasts can detect bacterial signals through bitter taste receptors to trigger host innate immune responses, thus regulating oral microbial homeostasis. This review focuses on how taste receptors, particularly bitter and sweet taste receptors, mediate the oral microbiota-host interaction as well as impact the occurrence and development of oral diseases. Further studies delineating the role of taste receptors in mediating oral microbiota-host interaction will advance our knowledge in oral ecological homeostasis establishment, providing a novel paradigm and treatment target for the better management of dental infectious diseases.
Collapse
Affiliation(s)
- Hao Dong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiaxin Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianhui Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Zhiyan Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Marco Tizzano
- Basic and Translation Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xian Peng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Xin Zheng, ; Xin Xu,
| | - Xin Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Xin Zheng, ; Xin Xu,
| |
Collapse
|
12
|
Ekechukwu ON, Christian M. Metabolic responses of light and taste receptors - unexpected actions of GPCRs in adipocytes. Rev Endocr Metab Disord 2022; 23:111-120. [PMID: 34195966 PMCID: PMC8873064 DOI: 10.1007/s11154-021-09667-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2021] [Indexed: 11/30/2022]
Abstract
The G-protein-coupled receptor (GPCR) superfamily includes sensory receptors that can detect and respond to taste and light. Recent investigations have identified key metabolic roles for such receptors in tissues considered 'non-sensory' such as adipose tissue. The major functions of white and brown adipose tissues include energy storage/release and thermogenesis, respectively. These processes are tightly controlled by GPCR pathways that serve to maintain energy homeostasis. Opsins 3 and 4 are GPCRs activated by blue light and in adipocytes control lipolysis as well as affect brown adipocyte activity. Furthermore, Opsin 3 signals to regulate the conversion of white to thermogenic beige/BRITE (Brown-in-white) adipocytes. Taste receptors that respond to fatty acids, sweet and bitter are expressed in adipocytes as well as in taste buds. Ffar2 and the long chain fatty acid receptor GPR120 are highly expressed in white adipocytes and the human tongue. In adipose tissue Ffar2 mediates the metabolic effects of butyrate and propionate produced by the gut microbiome. GPR120 is highly expressed in brown adipose tissue and regulates fatty acid oxidation and mitochondrial function. The type I taste receptor Tas1r3 senses sweet and umami, is expressed in adipocytes and on obesogenic diets Tas1r3 global gene knockout protects from metabolic dysfunction. Type II taste receptors that sense bitter are expressed by adipocytes and bitter agonists have been found to modulate adipocyte differentiation and lipid storage levels. This review explores recent unexpected findings of light and taste receptors in adipocytes and examines effects of their signaling in the control of adipose tissue biology.
Collapse
Affiliation(s)
- Onyinye Nuella Ekechukwu
- School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, UK
| | - Mark Christian
- School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, UK.
| |
Collapse
|
13
|
Lin C, Tordoff MG, Li X, Bosak NP, Inoue M, Ishiwatari Y, Chen L, Beauchamp GK, Bachmanov AA, Reed DR. Genetic controls of Tas1r3-independent sucrose consumption in mice. Mamm Genome 2021; 32:70-93. [PMID: 33710367 DOI: 10.1007/s00335-021-09860-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/11/2021] [Indexed: 10/21/2022]
Abstract
We have previously used crosses between C57BL/6ByJ (B6) and 129P3/J (129) inbred strains to map a quantitative trait locus (QTL) on mouse chromosome (Chr) 4 that affects behavioral and neural responses to sucrose. We have named it the sucrose consumption QTL 2 (Scon2), and shown that it corresponds to the Tas1r3 gene, which encodes a sweet taste receptor subunit TAS1R3. To discover other sucrose consumption QTLs, we have intercrossed B6 inbred and 129.B6-Tas1r3 congenic mice to produce F2 hybrids, in which Scon2 (Tas1r3) does not segregate, and hence does not contribute to phenotypical variation. Chromosome mapping using this F2 intercross identified two main-effect QTLs, Scon3 (Chr9) and Scon10 (Chr14), and an epistatically interacting QTL pair Scon3 (Chr9)-Scon4 (Chr1). Using serial backcrosses, congenic and consomic strains, we conducted high-resolution mapping of Scon3 and Scon4 and analyzed their epistatic interactions. We used mice with different Scon3 or Scon4 genotypes to understand whether these two QTLs influence sucrose intake via gustatory or postoral mechanisms. These studies found no evidence for involvement of the taste mechanisms, but suggested involvement of energy metabolism. Mice with the B6 Scon4 genotype drank less sucrose in two-bottle tests, and also had a higher respiratory exchange ratio and lower energy expenditure under basal conditions (when they had only chow and water available). Our results provide evidence that Scon3 and Scon4 influence mouse-to-mouse variation in sucrose intake and that both likely act through a common postoral mechanism.
Collapse
Affiliation(s)
- Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | | | - Xia Li
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Sonora Quest Laboratories, Phoenix, AZ, USA
| | | | - Masashi Inoue
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Laboratory of Cellular Neurobiology, School of Life Science, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan
| | - Yutaka Ishiwatari
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Ajinomoto Co. Inc, Tokyo, Japan
| | - Longhui Chen
- Monell Chemical Senses Center, Philadelphia, PA, USA.,Tannbach Capital, Hong Kong, China
| | | | - Alexander A Bachmanov
- Monell Chemical Senses Center, Philadelphia, PA, USA.,GlaxoSmithKline, Collegeville, PA, USA
| | | |
Collapse
|
14
|
Loss of the nutrient receptor Tas1R3 reduces atherosclerotic plaque accumulation and hepatic steatosis in ApoE -/- mice. J Physiol Biochem 2020; 76:623-636. [PMID: 33033981 DOI: 10.1007/s13105-020-00768-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 09/25/2020] [Indexed: 10/23/2022]
Abstract
The taste receptor type I (Tas1R) family consists of three G protein-coupled receptors (T1R1, T1R2, and T1R3) that form heterodimers recognizing sweet compounds (T1R2/T1R3) or amino acids (T1R1/T1R3). These receptors are nutrient sensors that facilitate appropriate physiological responses with nutrient availability. However, their contribution to the development of pathologies associated with overnutrition (e.g., atherosclerosis) is unclear. The aim of the present study was to determine if T1R3 deletion would reduce atherosclerotic plaque development in mice. We generated atherosclerotic mice with whole-body deletion of T1R3 by crossing T1R3-/- mice with ApoE-/- mice. T1R3+/+ ApoE-/- and T1R3-/- ApoE-/- mice were maintained on an atherogenic high-fat diet for 8 weeks. Weight gain and food consumption were measured during the 8-week diet. Atherosclerotic lesion development and size were assessed by en face analysis of intact aortas and microscopic analysis of aortic roots. Our results indicate that T1R3 deletion in male and female ApoE-/- mice reduces aortic atherosclerotic plaque accumulation. Hepatic triglyceride accumulation, which was measured by quantification of oil red O staining, was also reduced in T1R3-/- mice. While the ablation of T1R3 reduced the final body weight of both males and females by approximately 12%, serum lipids, insulin, and glucose were either unchanged or slightly reduced. Immunoblot analysis of the phosphorylation of p70S6K, an effector of mTORC1, suggests T1R3 ablation reduces mTORC1 activity by approximately 50% in the male livers. Collectively, these findings suggest that the whole-body deletion of T1R3 reduces atherosclerosis and hepatic steatosis in a manner largely independent of the measured effects on whole-body glucose and lipid homeostasis.
Collapse
|
15
|
Aydin MD, Aydin A, Caglar O, Aydin ME, Karadeniz E, Nalci KA, Demirtas R. New description of vagal nerve commanted intrapancreatic taste buds and blood glucose level: An experimental analysis. ACTA ACUST UNITED AC 2020; 11:181-185. [PMID: 34336606 PMCID: PMC8314032 DOI: 10.34172/bi.2021.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/19/2020] [Accepted: 07/04/2020] [Indexed: 11/17/2022]
Abstract
![]()
Introduction: There have been thousands of neurochemical mechanism about blood glucose level regulation, but intrapancreatic taste buds and their roles in blood glucose level has not been described. We aimed to investigate if there are taste buds cored neural networks in the pancreas, and there is any relationship between blood glucose levels. Methods: This examination was done on 32 chosen rats with their glucose levels. Animals are divided into owned blood glucose levels. If mean glucose levels were equal to 105 ± 10 mg/dL accepted as euglycemic (G-I; n = 14), 142 ± 18 mg/dL values accepted as hyperglycemic (G-II; n = 9) and 89 ± 9 mg/dL accepted as hypoglycemic (G-III; n = 9). After the experiment, animals were sacrificed under general anesthesia. Their pancreatic tissues were examined histological methods and numbers of newly described taste bud networks analyzed by Stereological methods. Results compared with Mann-Whitney U test P < 0.005 considered as significant. Results: The mean normal blood glucose level (mg/dL) and taste bud network densities of per cm3 were: 105 ± 10 mg/dL; 156±21 in G-I; 142 ± 18 mg/dL and 95 ± 14 in G-II and 89 ± 9 mg/dL and 232 ± 34 in G-III. P values as follows: P < 0.001 of G-II/G-I; P < 0.005 of G-III/G-I and P < 0.0001 of G-III/G-II. We detected periarterial located taste buds like cell clusters and peripherally located ganglia connected with Langerhans cells via thin nerve fibers. There was an inverse relationship between the number of taste buds networks and blood glucose level. Conclusion: Newly described intrapancreatic taste buds may have an important role in the regulation of blood glucose level.
Collapse
Affiliation(s)
- Mehmet Dumlu Aydin
- Department of Neurosurgery, Medical Faculty of Ataturk University, Erzurum, Turkey
| | - Aybike Aydin
- Medical Faculty of Cerrapasa, Istanbul University, Istanbul, Turkey
| | - Ozgur Caglar
- Department of Pediatric Surgery, Medical Faculty of Ataturk University, Erzurum, Turkey
| | - Muhammed Enes Aydin
- Department of Anesthesiology and Reanimation, Medical Faculty of Ataturk University, Erzurum, Turkey
| | - Erdem Karadeniz
- Department of General Surgery, Medical Faculty of Ataturk University, Erzurum, Turkey
| | - Kemal Alp Nalci
- Department of Pharmacology, Medical Faculty of Ataturk University, Erzurum, Turkey
| | - Rabia Demirtas
- Department of Pathology, Medical Faculty of Ataturk University, Erzurum, Turkey
| |
Collapse
|
16
|
Allelic variation of the Tas1r3 taste receptor gene affects sweet taste responsiveness and metabolism of glucose in F1 mouse hybrids. PLoS One 2020; 15:e0235913. [PMID: 32673349 PMCID: PMC7365461 DOI: 10.1371/journal.pone.0235913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/25/2020] [Indexed: 11/25/2022] Open
Abstract
In mammals, inter- and intraspecies differences in consumption of sweeteners largely depend on allelic variation of the Tas1r3 gene (locus Sac) encoding the T1R3 protein, a sweet taste receptor subunit. To assess the influence of Tas1r3 polymorphisms on feeding behavior and metabolism, we examined the phenotype of F1 male hybrids obtained from crosses between the following inbred mouse strains: females from 129SvPasCrl (129S2) bearing the recessive Tas1r3 allele and males from either C57BL/6J (B6), carrying the dominant allele, or the Tas1r3-gene knockout strain C57BL/6J-Tas1r3tm1Rfm (B6-Tas1r3-/-). The hybrids 129S2B6F1 and 129S2B6-Tas1r3-/-F1 had identical background genotypes and different sets of Tas1r3 alleles. The effect of Tas1r3 hemizygosity was analyzed by comparing the parental strain B6 (Tas1r3 homozygote) and hemizygous F1 hybrids B6 × B6-Tas1r3-/-. Data showed that, in 129S2B6-Tas1r3-/-F1 hybrids, the reduction of glucose tolerance, along with lower consumption of and lower preference for sweeteners during the initial licking responses, is due to expression of the recessive Tas1r3 allele. Hemizygosity of Tas1r3 did not influence these behavioral and metabolic traits. However, the loss of the functional Tas1r3 allele was associated with a small decline in the long-term intake and preference for sweeteners and reduction of plasma insulin and body, liver, and fat mass.
Collapse
|
17
|
Park S, Liu M, Song MY. Mental stress and physical activity interact with the genetic risk scores of the genetic variants related to sweetness preference in high sucrose-containing food and glucose tolerance. Food Sci Nutr 2020; 8:3492-3503. [PMID: 32724612 PMCID: PMC7382188 DOI: 10.1002/fsn3.1632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 11/20/2022] Open
Abstract
We hypothesized that subjects with genetic variants that increase sweet taste preference would consume more sucrose-containing foods and have altered energy and glucose metabolisms, which would have interactions with lifestyles. Korean genome and epidemiology study (KoGES) was conducted to determine genetic variants and lifestyles including nutrient intakes by the Korean Center for Disease and Control during 2004-2013. Subjects were 8,842 adults aged 40-69 years in Ansan/Ansung cohorts in Korea. The associations between genetic risk scores(GRS) selected for influencing higher sweet preference and energy and glucose metabolism were examined using logistic regression after adjusting for covariates. GRS included 8 SNPs, TAS1R2_rs61761364, SLC2A5_rs11121306, SLC2A7_ rs769902, SLC2A5_rs765618, TRPM5_rs1965606, TRPV1_rs224495, TRPV1_ rs8065080, and TRPV1_rs8078502. Sweet taste preference was higher by 1.30-folds in high GRS than in low GRS (p < .0001). Consistent with sweet taste preference, carriers with high GRS had a higher intake of sucrose-containing foods by 1.25 (1.08-1.46)-fold than those with low GRS after adjusting age, gender, BMI, and energy intake. However, glucose intolerance risk was rather lower by 0.861 (0.76-0.98)-fold in high GRS than low GRS (p < .05). GRS tended to interact with mental stress to affect sucrose intake (p = .048). Only in low mental stress levels, sucrose-containing food intake was higher in high GRS than low GRS. There was an interaction of GRS with physical activity to influence glucose intolerance. Serum glucose concentrations were lower by 0.808-folds in high GRS than low GRS only in a high physical activity state. In conclusion, adults with genetically high sweet taste preference had a positive association with high sucrose-containing food intakes and improved glucose tolerance. The genetic impact on sweetness preference was associated with offset by high mental stress and lack of physical activity.
Collapse
Affiliation(s)
- Sunmin Park
- Department of Food and NutritionObesity/Diabetes Research CenterHoseo UniversityAsanSouth Korea
| | - Meiling Liu
- Department of Food and NutritionObesity/Diabetes Research CenterHoseo UniversityAsanSouth Korea
| | - Mi Young Song
- Department of Food Science and NutritionWoo Song UniversityDaejeonSouth Korea
| |
Collapse
|
18
|
Sclafani A, Zukerman S, Ackroff K. Residual Glucose Taste in T1R3 Knockout but not TRPM5 Knockout Mice. Physiol Behav 2020; 222:112945. [PMID: 32417232 DOI: 10.1016/j.physbeh.2020.112945] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/19/2022]
Abstract
Knockout (KO) mice missing the sweet taste receptor subunit T1R3 or the signaling protein TRPM5 have greatly attenuated sweetener preferences. Yet both types of KO mice develop preferences for glucose but not fructose in 24-h tests, which has been attributed to the postoral reinforcing actions of glucose. Here we probed for residual sugar taste sensitivity in KO mice. Unlike wildtype (WT) mice, food-restricted T1R3 KO and TRPM5 KO mice displayed little attraction for 8% glucose and 8% fructose in 1-min, two-bottle choice tests. However, in 1-h tests about half of the T1R3 KO mice displayed a significant preference for glucose over fructose (78-84%), while WT mice showed either no or weak preferences (41-56%) for glucose. Following one-bottle training sessions, WT mice display greater glucose preferences although still weaker than those observed in T1R3 KO mice. In contrast, TRPM5 KO mice were indifferent to sugars in 1-h tests but developed a strong preference for glucose over fructose in 24-h tests. T1R3 taste cells contain the sodium glucose cotransporter 1 (SGLT1) and the ATP-gated K+ (KATP) metabolic sensor, which may mediate the unlearned glucose preference displayed by T1R3 KO mice. Unlike WT mice, many T1R3 KO mice strongly preferred glucose to a non-metabolizable glucose analog (α-methyl-D-glucopyranoside, MDG) in initial 1-h choice tests. Glucose and MDG are both ligands for SGLT1 which indicates that SGLT1 sensing does not mediate the glucose preference of T1R3 KO mice. Instead, KATP sensing and/or other oral sensors are implicated. The MDG findings also argue against postoral sensing as the primary source of the initial glucose preference displayed by T1R3 KO mice. Why only half of the T1R3 KO mice showed this preference in 1-h tests remains to be determined. All T1R3 KO mice preferred glucose to fructose in 24-h tests, which appears to be due to both oral and postoral glucose sensing.
Collapse
Affiliation(s)
- Anthony Sclafani
- Department of Psychology, Brooklyn College of City University of New York, Brooklyn, New York 11210, USA.
| | - Steven Zukerman
- Department of Psychology, Brooklyn College of City University of New York, Brooklyn, New York 11210, USA
| | - Karen Ackroff
- Department of Psychology, Brooklyn College of City University of New York, Brooklyn, New York 11210, USA
| |
Collapse
|
19
|
Gendron CM, Chakraborty TS, Chung BY, Harvanek ZM, Holme KJ, Johnson JC, Lyu Y, Munneke AS, Pletcher SD. Neuronal Mechanisms that Drive Organismal Aging Through the Lens of Perception. Annu Rev Physiol 2019; 82:227-249. [PMID: 31635526 DOI: 10.1146/annurev-physiol-021119-034440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Sensory neurons provide organisms with data about the world in which they live, for the purpose of successfully exploiting their environment. The consequences of sensory perception are not simply limited to decision-making behaviors; evidence suggests that sensory perception directly influences physiology and aging, a phenomenon that has been observed in animals across taxa. Therefore, understanding the neural mechanisms by which sensory input influences aging may uncover novel therapeutic targets for aging-related physiologies. In this review, we examine different perceptive experiences that have been most clearly linked to aging or age-related disease: food perception, social perception, time perception, and threat perception. For each, the sensory cues, receptors, and/or pathways that influence aging as well as the individual or groups of neurons involved, if known, are discussed. We conclude with general thoughts about the potential impact of this line of research on human health and aging.
Collapse
Affiliation(s)
- Christi M Gendron
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Tuhin S Chakraborty
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Brian Y Chung
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Zachary M Harvanek
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Kristina J Holme
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Jacob C Johnson
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Yang Lyu
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Allyson S Munneke
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Scott D Pletcher
- Department of Molecular and Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, Michigan 48109, USA; .,Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
20
|
Sarnelli G, Annunziata G, Magno S, Oriolo C, Savastano S, Colao A. Taste and the Gastrointestinal tract: from physiology to potential therapeutic target for obesity. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2019; 9:1-9. [PMID: 31391920 DOI: 10.1038/s41367-019-0012-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Flavor is the combination of gustatory, olfactory and trigeminal sensations, representing the three main sensory pathways that allow detecting environmental chemical substances. Taste, in particular, is a complex chemosensory path that allows identification of substances present in ingested foods and beverages. In this manuscript, we propose a conceptual roadmap from aspects related to the evolution and the physiological role of taste, up to the current knowledge about its implication in the modulation of a healthy state, or obesity. More specifically, we focused on the role of stimulation of taste receptors in releasing gut hormones (also known as enterohormones), and their effects on the regulation of food intake, by inducing satiety, either by locally acting (in the gastrointestinal tract), or centrally (in the brain). Recent evidence demonstrated that some enterohormones are able to modulate gastrointestinal motility, thus affecting an orexigenic responses in the central nervous system. In keeping with this, we discuss the ability of the gustatory system to be a final checkpoint control for food intake regulation, and we speculate about taste perception manipulation in the management of obesity.
Collapse
Affiliation(s)
- Giovanni Sarnelli
- 1Department of Clinical Medicine and Surgery, Division of Gastroenterology, University of Naples Federico II, Naples, Italy
| | | | - Silvia Magno
- Obesity Center at the Endocrinology Unit, Department of Clinical and Experimental Medicine, Pisa, Italy
| | - Claudia Oriolo
- 4Endocrinology Unit, Medical Department of Care Continuity and Disability, University of Bologna, Bologna, Italy
| | - Silvia Savastano
- 5Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Napoli, Italy
| | - Annamaria Colao
- 5Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Napoli, Italy
| | | |
Collapse
|
21
|
Murovets VO, Sozontov EA, Zachepilo TG. The Effect of the Taste Receptor Protein T1R3 on the Development of Islet Tissue of the Murine Pancreas. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2019; 484:1-4. [PMID: 31016494 DOI: 10.1134/s0012496619010010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Indexed: 01/06/2023]
Abstract
T1R3 protein, the main subunit of the sweet taste receptor and receptor of amino acid taste, is expressed in the epithelium of the tongue and gastrointestinal tract, in β cells of the pancreas, hypothalamus, and numerous other organs. Recently, convincing evidences on the involvement of T1R3 in the control of carbohydrate and lipid metabolism, and the control of incretin and insulin production were obtained. In the study on Tas1r3-gene knockout mouse strain and parent C57BL/6J strain as a control, the data on the effect of T1R3 on morphological characteristics of Langerhans islets in the pancreas were obtained. In Tas1r3 knockout animals, we found a reduction in the size of islets and their density in pancreatic tissue as compared to the parent strain. In addition, a decrease in the expression of active caspase-3 in the islets of gene-knockout mice was demonstrated. The data obtained indicate that the lack of functioning gene encoding sweet taste receptor protein causes a dystrophy of the islet tissue and is associated with the development of pathological changes in the pancreas specific to type 2 diabetes mellitus and obesity in humans.
Collapse
Affiliation(s)
- V O Murovets
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034, St. Petersburg, Russia.
| | - E A Sozontov
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034, St. Petersburg, Russia.,St. Petersburg State University, 199034, St. Petersburg, Russia
| | - T G Zachepilo
- Pavlov Institute of Physiology, Russian Academy of Sciences, 199034, St. Petersburg, Russia
| |
Collapse
|
22
|
Wang F, Song X, Zhou L, Liang G, Huang F, Jiang G, Zhang L. The downregulation of sweet taste receptor signaling in enteroendocrine L-cells mediates 3-deoxyglucosone-induced attenuation of high glucose-stimulated GLP-1 secretion. Arch Physiol Biochem 2018; 124:430-435. [PMID: 29277113 DOI: 10.1080/13813455.2017.1419366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CONTEXT Sweet taste receptors (STRs) involve in regulating the release of glucose-stimulated glucagon-like peptide-1 (GLP-1). Our in vivo and in vitro studies found that 3-deoxyglucosone (3DG) inhibited glucose-stimulated GLP-1 secretion. OBJECTIVE This study investigated the role of STRs in 3DG-induced inhibition of high glucose-stimulated GLP-1 secretion. METHODS STC-1 cells were incubated with lactisole or 3DG for 1 h under 25 mM glucose conditions. Western blotting was used to study the expression of STRs signaling molecules and ELISA was used to analyse GLP-1 and cyclic adenosine monophosphate (cAMP) levels. RESULTS Lactisole inhibited GLP-1 secretion. Exposure to 25 mM glucose increased the expressions of STRs subunits when compared with 5.6 mM glucose. 3DG decreased GLP-1 secretion and STRs subunits expressions, with affecting other components of STRs pathway, including the downregulation of transient receptor potential cation channel subfamily M member 5 (TRPM5) expression and the reduction of intracellular cAMP levels. CONCLUSION 3DG attenuates high glucose-stimulated GLP-1 secretion by reducing STR subunit expression and downstream signaling components.
Collapse
Affiliation(s)
- Fei Wang
- a Suzhou Academy of Wumen Chinese Medicine , Suzhou Hospital of Traditional Chinese Medicine , Suzhou , P. R. China
| | - Xiudao Song
- a Suzhou Academy of Wumen Chinese Medicine , Suzhou Hospital of Traditional Chinese Medicine , Suzhou , P. R. China
| | - Liang Zhou
- a Suzhou Academy of Wumen Chinese Medicine , Suzhou Hospital of Traditional Chinese Medicine , Suzhou , P. R. China
| | - Guoqiang Liang
- a Suzhou Academy of Wumen Chinese Medicine , Suzhou Hospital of Traditional Chinese Medicine , Suzhou , P. R. China
| | - Fei Huang
- a Suzhou Academy of Wumen Chinese Medicine , Suzhou Hospital of Traditional Chinese Medicine , Suzhou , P. R. China
| | - Guorong Jiang
- a Suzhou Academy of Wumen Chinese Medicine , Suzhou Hospital of Traditional Chinese Medicine , Suzhou , P. R. China
| | - Lurong Zhang
- a Suzhou Academy of Wumen Chinese Medicine , Suzhou Hospital of Traditional Chinese Medicine , Suzhou , P. R. China
| |
Collapse
|
23
|
Murovets VO, Lukina EA, Zolotarev VA. The Effect of Tas1r3 Gene Polymorphism on Preference and Consumption of Sucrose and Low-Calorie Sweeteners in Interstrain Hybrid Mice of the First Filial Generation. J EVOL BIOCHEM PHYS+ 2018. [DOI: 10.1134/s0022093018030079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Emerging Concepts in Brain Glucose Metabolic Functions: From Glucose Sensing to How the Sweet Taste of Glucose Regulates Its Own Metabolism in Astrocytes and Neurons. Neuromolecular Med 2018; 20:281-300. [DOI: 10.1007/s12017-018-8503-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/13/2018] [Indexed: 12/16/2022]
|
25
|
Feng R, Qian C, Liu Q, Jin Y, Liu L, Li S, Liao Y, Zhou H, Liu W, Rayner CK, Ma J. Expression of sweet taste receptor and gut hormone secretion in modelled type 2 diabetes. Gen Comp Endocrinol 2017; 252:142-149. [PMID: 28782537 DOI: 10.1016/j.ygcen.2017.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/29/2017] [Accepted: 08/03/2017] [Indexed: 01/05/2023]
Abstract
Sweet taste receptors (STRs) are expressed in L cells which secret glucagon-like peptide-1 (GLP-1) in the gut. The STR blocker lactisole reduces GLP-1 secretion and increases blood glucose levels. Therefore, we investigated the expression of sweet taste molecules in the proximal and distal small intestine, and gut hormone secretion, in healthy control and type 2 diabetic rats. Two groups of rats (Sprague Dawley (SD), and Zucker diabetic fatty (ZDF)) were involved in the study. Each group (n=10) received an intragastric glucose infusion (50% glucose solution, 2g/kg body weight). Blood samples were taken for measurement of blood glucose, plasma insulin, and GLP-1 concentrations. One week later, we obtained small intestinal tissue and detected the expression of STRs and glucose transporters (GTs) by real time polymerase chain reaction (Real Time-PCR). Sweet taste molecules of T1R2, T1R3, α-gustducin and TRPM5 in ileum were dramatically higher than those in duodenum (P<0.01 for each). T1R3, α-gustducin and TRPM5 expression were less in the ileum of ZDF than those in SD (P<0.05 for each), while expression of glucose transporter 2 (GLUT-2) in ileum was significantly higher in ZDF rats. Plasma GLP-1 levels were higher in ZDF rats than SD rats at t=0, 15, 30, 60 and 120min (P<0.01). In conclusion, transcript levels of ileal T1R3 and GLUT-2 are disordered in ZDF rats suggesting that intestinal sweet taste receptor expression is associated with altered glucose metabolism. The mechanism needs further investigation, but might provide a potential therapy in the treatment of type 2 diabetes.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/metabolism
- Disease Models, Animal
- Duodenum/metabolism
- Glucagon-Like Peptide 1/blood
- Glucagon-Like Peptide 1/metabolism
- Glucose Transporter Type 2/genetics
- Glucose Transporter Type 2/metabolism
- Ileum/metabolism
- Insulin/blood
- Intestinal Mucosa/metabolism
- Male
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Rats, Zucker
- Receptors, G-Protein-Coupled/metabolism
- Taste
- Transducin/metabolism
Collapse
Affiliation(s)
- Rilu Feng
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Cheng Qian
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qianjing Liu
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yunqiu Jin
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lianyong Liu
- Department of Endocrinology and Metabolism, Shanghai Punan Hospital, Shanghai, China
| | - Shengxian Li
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu Liao
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huan Zhou
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Liu
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chris K Rayner
- Discipline of Medicine, University of Adelaide, Adelaide 5000, Australia; Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Jing Ma
- Division of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
26
|
Kohno D. Sweet taste receptor in the hypothalamus: a potential new player in glucose sensing in the hypothalamus. J Physiol Sci 2017; 67:459-465. [PMID: 28378265 PMCID: PMC10717116 DOI: 10.1007/s12576-017-0535-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023]
Abstract
The hypothalamic feeding center plays an important role in energy homeostasis. The feeding center senses the systemic energy status by detecting hormone and nutrient levels for homeostatic regulation, resulting in the control of food intake, heat production, and glucose production and uptake. The concentration of glucose is sensed by two types of glucose-sensing neurons in the feeding center: glucose-excited neurons and glucose-inhibited neurons. Previous studies have mainly focused on glucose metabolism as the mechanism underlying glucose sensing. Recent studies have indicated that receptor-mediated pathways also play a role in glucose sensing. This review describes sweet taste receptors in the hypothalamus and explores the role of sweet taste receptors in energy homeostasis.
Collapse
Affiliation(s)
- Daisuke Kohno
- Advanced Scientific Research Leaders Development Unit, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan.
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan.
| |
Collapse
|
27
|
Extraoral Taste Receptor Discovery: New Light on Ayurvedic Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017. [PMID: 28642799 PMCID: PMC5469997 DOI: 10.1155/2017/5435831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
More and more research studies are revealing unexpectedly important roles of taste for health and pathogenesis of various diseases. Only recently it has been shown that taste receptors have many extraoral locations (e.g., stomach, intestines, liver, pancreas, respiratory system, heart, brain, kidney, urinary bladder, pancreas, adipose tissue, testis, and ovary), being part of a large diffuse chemosensory system. The functional implications of these taste receptors widely dispersed in various organs or tissues shed a new light on several concepts used in ayurvedic pharmacology (dravyaguna vijnana), such as taste (rasa), postdigestive effect (vipaka), qualities (guna), and energetic nature (virya). This review summarizes the significance of extraoral taste receptors and transient receptor potential (TRP) channels for ayurvedic pharmacology, as well as the biological activities of various types of phytochemical tastants from an ayurvedic perspective. The relative importance of taste (rasa), postdigestive effect (vipaka), and energetic nature (virya) as ethnopharmacological descriptors within Ayurveda boundaries will also be discussed.
Collapse
|
28
|
Conde-Sieira M, Soengas JL. Nutrient Sensing Systems in Fish: Impact on Food Intake Regulation and Energy Homeostasis. Front Neurosci 2017; 10:603. [PMID: 28111540 PMCID: PMC5216673 DOI: 10.3389/fnins.2016.00603] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/19/2016] [Indexed: 12/27/2022] Open
Abstract
Evidence obtained in recent years in a few species, especially rainbow trout, supports the presence in fish of nutrient sensing mechanisms. Glucosensing capacity is present in central (hypothalamus and hindbrain) and peripheral [liver, Brockmann bodies (BB, main accumulation of pancreatic endocrine cells in several fish species), and intestine] locations whereas fatty acid sensors seem to be present in hypothalamus, liver and BB. Glucose and fatty acid sensing capacities relate to food intake regulation and metabolism in fish. Hypothalamus is as a signaling integratory center in a way that detection of increased levels of nutrients result in food intake inhibition through changes in the expression of anorexigenic and orexigenic neuropeptides. Moreover, central nutrient sensing modulates functions in the periphery since they elicit changes in hepatic metabolism as well as in hormone secretion to counter-regulate changes in nutrient levels detected in the CNS. At peripheral level, the direct nutrient detection in liver has a crucial role in homeostatic control of glucose and fatty acid whereas in BB and intestine nutrient sensing is probably involved in regulation of hormone secretion from endocrine cells.
Collapse
Affiliation(s)
- Marta Conde-Sieira
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo Vigo, Spain
| | - José L Soengas
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo Vigo, Spain
| |
Collapse
|
29
|
In vitro evidence in rainbow trout supporting glucosensing mediated by sweet taste receptor, LXR, and mitochondrial activity in Brockmann bodies, and sweet taste receptor in liver. Comp Biochem Physiol B Biochem Mol Biol 2016; 200:6-16. [DOI: 10.1016/j.cbpb.2016.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/25/2016] [Accepted: 04/27/2016] [Indexed: 12/31/2022]
|
30
|
Abstract
The T1R2 (taste type 1 receptor, member 2)/T1R3 (taste type 1 receptor, member 3) sweet taste receptor is expressed in taste buds on the tongue, where it allows the detection of energy-rich carbohydrates of food. This single receptor responds to all compounds perceived as sweet by humans, including natural sugars and natural and artificial sweeteners. Importantly, the T1R2/T1R3 sweet taste receptor is also expressed in extra-oral tissues, including the stomach, pancreas, gut, liver, and brain. Although its physiological role remains to be established in numerous organs, T1R2/T1R3 is suspected to be involved in the regulation of metabolic processes, such as sugar sensing, glucose homeostasis, and satiety hormone release. In this review, the physiological role of the sweet taste receptor in taste perception and metabolic regulation is discussed by focusing on dysfunctions leading to diabetes. Current knowledge of T1R2/T1R3 inhibitors making this receptor a promising therapeutic target for the treatment of type 2 diabetes is also summarized and discussed.
Collapse
Affiliation(s)
- Fabrice Neiers
- Centre des Sciences du Goût et de l'Alimentation, INRA, CNRS, Université de Bourgogne - Franche-Comté, Dijon, 21000, France
| | - Marie-Chantal Canivenc-Lavier
- Centre des Sciences du Goût et de l'Alimentation, INRA, CNRS, Université de Bourgogne - Franche-Comté, Dijon, 21000, France
| | - Loïc Briand
- Centre des Sciences du Goût et de l'Alimentation, INRA, CNRS, Université de Bourgogne - Franche-Comté, Dijon, 21000, France.
| |
Collapse
|
31
|
Nakagawa Y, Nagasawa M, Medina J, Kojima I. Glucose Evokes Rapid Ca2+ and Cyclic AMP Signals by Activating the Cell-Surface Glucose-Sensing Receptor in Pancreatic β-Cells. PLoS One 2015; 10:e0144053. [PMID: 26630567 PMCID: PMC4667910 DOI: 10.1371/journal.pone.0144053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
Glucose is a primary stimulator of insulin secretion in pancreatic β-cells. High concentration of glucose has been thought to exert its action solely through its metabolism. In this regard, we have recently reported that glucose also activates a cell-surface glucose-sensing receptor and facilitates its own metabolism. In the present study, we investigated whether glucose activates the glucose-sensing receptor and elicits receptor-mediated rapid actions. In MIN6 cells and isolated mouse β-cells, glucose induced triphasic changes in cytoplasmic Ca(2+) concentration ([Ca(2+)]c); glucose evoked an immediate elevation of [Ca(2+)]c, which was followed by a decrease in [Ca(2+)]c, and after a certain lag period it induced large oscillatory elevations of [Ca(2+)]c. Initial rapid peak and subsequent reduction of [Ca(2+)]c were independent of glucose metabolism and reproduced by a nonmetabolizable glucose analogue. These signals were also blocked by an inhibitor of T1R3, a subunit of the glucose-sensing receptor, and by deletion of the T1R3 gene. Besides Ca(2+), glucose also induced an immediate and sustained elevation of intracellular cAMP ([cAMP]c). The elevation of [cAMP]c was blocked by transduction of the dominant-negative Gs, and deletion of the T1R3 gene. These results indicate that glucose induces rapid changes in [Ca(2+)]c and [cAMP]c by activating the cell-surface glucose-sensing receptor. Hence, glucose generates rapid intracellular signals by activating the cell-surface receptor.
Collapse
Affiliation(s)
- Yuko Nakagawa
- Department of Cell Biology, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Japan
| | - Masahiro Nagasawa
- Department of Cell Biology, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Japan
| | - Johan Medina
- Department of Cell Biology, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Japan
| | - Itaru Kojima
- Department of Cell Biology, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Japan
- * E-mail:
| |
Collapse
|
32
|
Glendinning JI, Stano S, Holter M, Azenkot T, Goldman O, Margolskee RF, Vasselli JR, Sclafani A. Sugar-induced cephalic-phase insulin release is mediated by a T1r2+T1r3-independent taste transduction pathway in mice. Am J Physiol Regul Integr Comp Physiol 2015; 309:R552-60. [PMID: 26157055 PMCID: PMC4591378 DOI: 10.1152/ajpregu.00056.2015] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/06/2015] [Indexed: 12/11/2022]
Abstract
Sensory stimulation from foods elicits cephalic phase responses, which facilitate digestion and nutrient assimilation. One such response, cephalic-phase insulin release (CPIR), enhances glucose tolerance. Little is known about the chemosensory mechanisms that activate CPIR. We studied the contribution of the sweet taste receptor (T1r2+T1r3) to sugar-induced CPIR in C57BL/6 (B6) and T1r3 knockout (KO) mice. First, we measured insulin release and glucose tolerance following oral (i.e., normal ingestion) or intragastric (IG) administration of 2.8 M glucose. Both groups of mice exhibited a CPIR following oral but not IG administration, and this CPIR improved glucose tolerance. Second, we examined the specificity of CPIR. Both mouse groups exhibited a CPIR following oral administration of 1 M glucose and 1 M sucrose but not 1 M fructose or water alone. Third, we studied behavioral attraction to the same three sugar solutions in short-term acceptability tests. B6 mice licked more avidly for the sugar solutions than for water, whereas T1r3 KO mice licked no more for the sugar solutions than for water. Finally, we examined chorda tympani (CT) nerve responses to each of the sugars. Both mouse groups exhibited CT nerve responses to the sugars, although those of B6 mice were stronger. We propose that mice possess two taste transduction pathways for sugars. One mediates behavioral attraction to sugars and requires an intact T1r2+T1r3. The other mediates CPIR but does not require an intact T1r2+T1r3. If the latter taste transduction pathway exists in humans, it should provide opportunities for the development of new treatments for controlling blood sugar.
Collapse
Affiliation(s)
- John I Glendinning
- Department of Biology, Barnard College, Columbia University, New York, New York;
| | - Sarah Stano
- Department of Biology, Barnard College, Columbia University, New York, New York
| | - Marlena Holter
- Department of Biology, Barnard College, Columbia University, New York, New York
| | - Tali Azenkot
- Department of Biology, Barnard College, Columbia University, New York, New York
| | - Olivia Goldman
- Department of Biology, Barnard College, Columbia University, New York, New York
| | | | - Joseph R Vasselli
- Obesity Research Center, Department of Medicine, Columbia University, New York, New York; and
| | - Anthony Sclafani
- Department of Psychology, Brooklyn College of City University of New York, Brooklyn, New York
| |
Collapse
|