1
|
Jiang J, Chen D, Yu B, He J, Yu J, Mao X, Huang Z, Luo Y, Luo J, Zheng P. Lactic Acid and Glutamine Have Positive Synergistic Effects on Growth Performance, Intestinal Function, and Microflora of Weaning Piglets. Animals (Basel) 2024; 14:3532. [PMID: 39682497 DOI: 10.3390/ani14233532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
The objective of this study was to evaluate the effects of dietary addition of lactic acid and glutamine, and their interactions, on growth performance, nutrient digestibility, digestive enzyme activity, intestinal barrier functions, microflora, and expressions of intestinal development-related genes of weaning piglets. Ninety-six 24-day-old weaning piglets (Duroc × Landrace × Yorkshire, weaned at 21 ± 1 d and fed the basal diet for a 3 d adaptation period) with initial body weight of 7.24 ± 0.09 kg were randomly assigned to one of four dietary treatments with six replicates per treatment and four pigs per replicate in a 2 × 2 factorial treatment arrangements: (1) CON (a 2-period basal diet; control), (2) LS (supplemented with 2% lactic acid), (3) GS (supplemented with 1% glutamine), and (4) LGS (supplemented with 2% lactic acid and 1% glutamine). The study lasted for 28 d. On days 25-28, fresh fecal samples were collected to evaluate apparent total tract digestibility (ATTD) of nutrients. After 28 d, one weaning pig per pen was euthanized, and physiological samples obtained. Results showed that the supplementation of lactic acid improved the ADFI of the pigs (p < 0.05), while the pigs fed the glutamine diet had a greater ADFI and higher G/F (p < 0.05), and there were significant interactive effects between lactic acid and glutamine on the ADFI and G/F of the pigs (p < 0.05). The ATTD of CP and ash for pigs fed with lactic acid was significantly enhanced, and pigs fed the glutamine diet had greater ATTD of CP and ash (p < 0.05), while there were significant interactive effects between lactic acid and glutamine on the ATTD of CP and ash of the pigs (p < 0.05). Pigs fed with lactic acid exhibited greater activity of α-amylase and lipase (p < 0.05); moreover, the activity of lipase in the pigs showed a significant interactive effect between lactic acid and glutamine (p < 0.05). There was a greater villus height and villus height to crypt depth ratio in pigs fed with lactic acid (p < 0.05), and the villus height to crypt depth ratio of pigs fed with glutamine was greater (p < 0.05). There were greater GLUT2, IGF-1, TGF-β2, OCLN, and ZO-1 mRNA levels in pigs fed with lactic acid (p < 0.05), and the supplementation of glutamine increased SGLT1, GLUT2, PepT1, IGF-1, IGF-1R, TGFβ-2, GLP-2, and OCLN mRNA levels (p < 0.05), Additionally, expressions of SGLT1, GLUT2, PepT1, IGF-1, IGF-1R, TGFβ-2, GLP-2, CLDN-2, OCLN, and ZO-1 mRNA levels of pigs showed a positive interactive effect between lactic acid and glutamine (p < 0.05). Supplementation of lactic acid significantly increased the populations of Bifidobacterium in cecal digesta, Lactobacillus in colonic digesta, and the content of butyric acid in colonic digesta (p < 0.05). In addition, there were significant interactive effects between lactic acid and glutamine on populations of Bifidobacterium in cecal digesta, Lactobacillus in colonic digesta, and the content of acetic acid, butyric acid, and total VFAs in cecal digesta of the pigs (p < 0.05). Collectively, the current results indicate that dietary supplementation with lactic acid and glutamine had a positive synergistic effect on weaning pigs, which could improve growth performance through promoting the development of the small intestine, increasing digestive and barrier function, and regulating the balance of microflora in pigs, and which might be a potential feeding additive ensemble to enhance the health and growth of weaning piglets in the post-antibiotic era.
Collapse
Affiliation(s)
- Junjie Jiang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiangbing Mao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
2
|
Ma S, Zuo J, Chen B, Fu Z, Lin X, Wu J, Zheng B, Lu X. Structural, properties and digestion in vitro changes of starch subjected to high pressure homogenization: An update review. Int J Biol Macromol 2024; 282:137118. [PMID: 39489250 DOI: 10.1016/j.ijbiomac.2024.137118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
High pressure homogenization (HPH) is considered as a promising method for improving the ideal metabolic reaction of starch-based foods in the body, but there is still no comprehensive understanding of the structure-property relationship of starch treated with HPH. This study reviews the advantages and limitations of HPH in starch-based foods processing in recent years. It also elaborates the bidirectional regulation of HPH on starch structure-property and its potential in improving nutritional quality, which includes the regular modification effects of HPH on the multi-scale structure, physicochemical properties, and digestion characteristics of starch. It was found that HPH could lead to the degradation of amylopectin, destruction of amorphous structure, and homogenization of fine particles, promoting gelatinization and ultimately endowing starch with good solubility and digestibility. Moreover, it could reorganize and reorder the internal starch chains, or cause the particles to disintegrate into an amorphous state, thereby enhancing the anti-digestibility of starch. The interaction of starch with different nutrients during the HPH process could be further investigated in future studies and explored with other techniques for structure-property modifications, which would help expand the development of personalized starch foods to meet growing consumer demands.
Collapse
Affiliation(s)
- Shuang Ma
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China; China-Ireland International Cooperation Centre for Food Material Science and Structure Design, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jiaxin Zuo
- Centre of Excellence in Agri-food Technologies, National Centre for Food Manufacturing, College of Health and Science, University of Lincoln, Holbeach, Spalding, UK
| | - Bingbing Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zhaoxia Fu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xu Lin
- College of Jinshan, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jiaqi Wu
- College of Jinshan, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Baodong Zheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China; China-Ireland International Cooperation Centre for Food Material Science and Structure Design, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xu Lu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China; China-Ireland International Cooperation Centre for Food Material Science and Structure Design, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
3
|
Liu XY, Chi YF, Wu YS, Chai JK. Research progress and considerations on oral rehydration therapy for the prevention and treatment of severe burn shock: A narrative review. Burns 2024; 50:107160. [PMID: 39322503 DOI: 10.1016/j.burns.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/17/2024] [Accepted: 04/29/2024] [Indexed: 09/27/2024]
Abstract
Severe burns are a significant cause of life-threatening conditions in both peacetime and wartime. Shock is a critical complication during the early stages of burn injury, contributing substantially to mortality and long-term disability. Effective fluid resuscitation is crucial for preventing and treating shock, with prompt administration being vital. However, timely intravenous fluid resuscitation is often challenging, and errors in resuscitation significantly contribute to mortality. Therefore, exploring a more rapid and effective non-invasive method of fluid resuscitation is necessary. Oral rehydration therapy (ORT) has shown considerable potential in this regard. This paper reviews ORT's historical development and current research progress, discussing its application in early anti-shock treatment for burns. While ORT is generally safe, potential complications like diarrhoea, vomiting, and abdominal discomfort must be noted, particularly if the rehydration rate is too rapid or if gastrointestinal issues exist. Careful patient assessment and monitoring are essential during ORT administration. Based on a comprehensive review of relevant research, we present provisional guidelines for ORT in burn patients. These guidelines aim to inform clinical practice but should be applied cautiously due to limited clinical evidence. Implementation must be tailored to the patient's condition under healthcare supervision, with adjustments according to evolving circumstances: ① Initiation timing: Start as soon as possible, and the ideal start time is usually within 6 h after injury. ② Rate of application: Employing a fractional administration approach, wherein small quantities of approximately 150-250 millilitres are provided for each instance and the initial fluid rate of oral rehydration can be simplified to 100 mL/kg/24 h. ③ Composition combination: In addition to essential salts and glucose, the oral rehydration solution can incorporate various anti-inflammatory and cellular protection constituents.
Collapse
Affiliation(s)
- Xiang-Yu Liu
- Graduate School, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing 100853, China; Senior Department of Burns & Plastic Surgery, Institute of Burn in the Fourth Medical Centre, Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing 100048, China
| | - Yun-Fei Chi
- Senior Department of Burns & Plastic Surgery, Institute of Burn in the Fourth Medical Centre, Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing 100048, China
| | - Yu-Shou Wu
- Graduate School, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing 100853, China; Senior Department of Burns & Plastic Surgery, Institute of Burn in the Fourth Medical Centre, Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing 100048, China
| | - Jia-Ke Chai
- Senior Department of Burns & Plastic Surgery, Institute of Burn in the Fourth Medical Centre, Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing 100048, China.
| |
Collapse
|
4
|
Novoselsky R, Harnik Y, Yakubovsky O, Katina C, Levin Y, Bahar Halpern K, Pencovich N, Nachmany I, Itzkovitz S. Intracellular polarization of RNAs and proteins in the human small intestinal epithelium. PLoS Biol 2024; 22:e3002942. [PMID: 39621797 PMCID: PMC11637431 DOI: 10.1371/journal.pbio.3002942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 12/12/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
The intestinal epithelium is a polarized monolayer of cells, with an apical side facing the lumen and a basal side facing the blood stream. In mice, both proteins and mRNAs have been shown to exhibit global basal-apical polarization; however, polarization in the human intestine has not been systematically explored. Here, we employed laser-capture microdissection to isolate apical and basal epithelial segments from intestinal tissues of 8 individuals and performed RNA sequencing and mass-spectrometry proteomics. We find a substantial polarization of mRNA molecules that largely overlaps polarization patterns observed in mice. This mRNA polarization remains consistent across different zones of the intestinal villi and is generally correlated with the polarization of proteins. Our protein analysis exposes streamlined intracellular nutrient transport and processing and reveals that mitochondria and ribosomes are less polarized in humans compared to mice. Our study provides a resource for understanding human intestinal epithelial biology.
Collapse
Affiliation(s)
- Roy Novoselsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yotam Harnik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oran Yakubovsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Corine Katina
- The De Botton Protein Profiling, The Nancy and Stephen Grand Israel and Health Sciences National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- The De Botton Protein Profiling, The Nancy and Stephen Grand Israel and Health Sciences National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Niv Pencovich
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ido Nachmany
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
5
|
Nwako JG, McCauley HA. Enteroendocrine cells regulate intestinal homeostasis and epithelial function. Mol Cell Endocrinol 2024; 593:112339. [PMID: 39111616 PMCID: PMC11401774 DOI: 10.1016/j.mce.2024.112339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/11/2024]
Abstract
Enteroendocrine cells (EECs) are well-known for their systemic hormonal effects, especially in the regulation of appetite and glycemia. Much less is known about how the products made by EECs regulate their local environment within the intestine. Here, we focus on paracrine interactions between EECs and other intestinal cells as they regulate three essential aspects of intestinal homeostasis and physiology: 1) intestinal stem cell function and proliferation; 2) nutrient absorption; and 3) mucosal barrier function. We also discuss the ability of EECs to express multiple hormones, describe in vitro and in vivo models to study EECs, and consider how EECs are altered in GI disease.
Collapse
Affiliation(s)
- Jennifer G Nwako
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Molecular Biology Research Building 5341C, Chapel Hill, NC 27599, USA
| | - Heather A McCauley
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Molecular Biology Research Building 5341C, Chapel Hill, NC 27599, USA.
| |
Collapse
|
6
|
Beck DL, Gilbert ER, Cline MA. Embryonic thermal challenge is associated with increased stressor resiliency later in life: Molecular and morphological mechanisms in the small intestine. Comp Biochem Physiol A Mol Integr Physiol 2024; 297:111724. [PMID: 39111617 DOI: 10.1016/j.cbpa.2024.111724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024]
Abstract
Developing chick embryos that are subjected to increased incubation temperature are more stressor-resilient later in life, but the underlying process is poorly understood. The potential mechanism may involve changes in small intestine function. In this study, we determined behavioral, morphological, and molecular effects of increased embryonic incubation temperatures and post-hatch heat challenge in order to understand how embryonic heat conditioning (EHC) affects gut function. At 4 days post-hatch, duodenum, jejunum, and ileum samples were collected at 0, 2, and 12 h relative to the start of heat challenge. In EHC chicks, we found that markers of heat and oxidative stress were generally lower while those of nutrient transport and antioxidants were higher. Temporally, gene expression changes in response to the heat challenge were similar in control and EHC chicks for markers of heat and oxidative stress. Crypt depth was greater in control than EHC chicks at 2 h post-challenge, and the villus height to crypt depth ratio increased from 2 to 12 h in both control and EHC chicks. Collectively, these results suggest that EHC chicks might be more energetically efficient at coping with thermal challenge, preferentially allocating nutrients to other tissues while protecting the mucosal layer from oxidative damage. These results provide targets for future studies aimed at understanding the molecular mechanisms underlying effects of embryonic heat exposure on intestinal function and stressor resiliency later in life.
Collapse
Affiliation(s)
- David L Beck
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Elizabeth R Gilbert
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Mark A Cline
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
7
|
Kiriyama Y, Tokumaru H, Sadamoto H, Kobayashi S, Nochi H. Effects of Phenolic Acids Produced from Food-Derived Flavonoids and Amino Acids by the Gut Microbiota on Health and Disease. Molecules 2024; 29:5102. [PMID: 39519743 PMCID: PMC11548037 DOI: 10.3390/molecules29215102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The gut microbiota metabolizes flavonoids, amino acids, dietary fiber, and other components of foods to produce a variety of gut microbiota-derived metabolites. Flavonoids are the largest group of polyphenols, and approximately 7000 flavonoids have been identified. A variety of phenolic acids are produced from flavonoids and amino acids through metabolic processes by the gut microbiota. Furthermore, these phenolic acids are easily absorbed. Phenolic acids generally represent phenolic compounds with one carboxylic acid group. Gut microbiota-derived phenolic acids have antiviral effects against several viruses, such as SARS-CoV-2 and influenza. Furthermore, phenolic acids influence the immune system by inhibiting the secretion of proinflammatory cytokines, such as interleukin-1β and tumor necrosis factor-α. In the nervous systems, phenolic acids may have protective effects against neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. Moreover, phenolic acids can improve levels of blood glucose, cholesterols, and triglycerides. Phenolic acids also improve cardiovascular functions, such as blood pressure and atherosclerotic lesions. This review focuses on the current knowledge of the effects of phenolic acids produced from food-derived flavonoids and amino acids by the gut microbiota on health and disease.
Collapse
Affiliation(s)
- Yoshimitsu Kiriyama
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido 1314-1, Sanuki 769-2193, Kagawa, Japan (H.S.); (S.K.); (H.N.)
- Institute of Neuroscience, Tokushima Bunri University, Shido 1314-1, Sanuki 769-2193, Kagawa, Japan
| | - Hiroshi Tokumaru
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido 1314-1, Sanuki 769-2193, Kagawa, Japan (H.S.); (S.K.); (H.N.)
| | - Hisayo Sadamoto
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido 1314-1, Sanuki 769-2193, Kagawa, Japan (H.S.); (S.K.); (H.N.)
| | - Suguru Kobayashi
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido 1314-1, Sanuki 769-2193, Kagawa, Japan (H.S.); (S.K.); (H.N.)
- Institute of Neuroscience, Tokushima Bunri University, Shido 1314-1, Sanuki 769-2193, Kagawa, Japan
| | - Hiromi Nochi
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido 1314-1, Sanuki 769-2193, Kagawa, Japan (H.S.); (S.K.); (H.N.)
| |
Collapse
|
8
|
Moloi S, Halász T, Csivincsik Á, Nagy G. Suitability of red fox ( Vulpes vulpes) and golden jackal ( Canis aureus) as hosts of Echinococcus multilocularis based on egg production characteristics and literature data on the intestinal ecosystems of carnivores. CURRENT RESEARCH IN PARASITOLOGY & VECTOR-BORNE DISEASES 2024; 6:100225. [PMID: 39554486 PMCID: PMC11567931 DOI: 10.1016/j.crpvbd.2024.100225] [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: 09/02/2024] [Revised: 10/19/2024] [Accepted: 10/27/2024] [Indexed: 11/19/2024]
Abstract
Echinococcus multilocularis is the most important food-borne parasite in Europe. Its natural definitive host is the red fox (Vulpes vulpes) while other canid species play a secondary role in the maintenance of its endemics. However, recent studies call attention to the potential of golden jackal (Canis aureus) as a suitable definitive host for E. multilocularis. Our study aimed to evaluate the quantitative and qualitative egg production traits of adult E. multilocularis in different hosts as an indicator of reproductive success. Investigation of 111 and 82 parasites from 33 red foxes and 29 golden jackals, respectively, we ascertained that the proportion of worms with mature eggs was significantly lower in golden jackals than in red foxes. Those worms, which produced mature eggs in golden jackal hosts, originated from less crowded infrapopulations than their fox-originated counterparts. Other characteristics of the parasite's reproductive ability, such as the proportion of fertile worms, and mean egg production were similar in the two hosts. Comparing these findings to evolutionary data on different canid taxa, we hypothesised that the mutual presence of red fox and a differently evolved host of E. multilocularis might contribute to the formation of stable parasite circulation in these multi-host systems.
Collapse
Affiliation(s)
- Sibusiso Moloi
- Department of Animal Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, 40 Guba S. Str., Kaposvár, 7400, Hungary
| | - Tibor Halász
- Department of Animal Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, 40 Guba S. Str., Kaposvár, 7400, Hungary
| | - Ágnes Csivincsik
- Department of Animal Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, 40 Guba S. Str., Kaposvár, 7400, Hungary
| | - Gábor Nagy
- Department of Animal Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Kaposvár Campus, 40 Guba S. Str., Kaposvár, 7400, Hungary
| |
Collapse
|
9
|
D’Orazio G, La Ferla B. Synthesis of a Small Library of Glycoderivative Putative Ligands of SGLT1 and Preliminary Biological Evaluation. Molecules 2024; 29:5067. [PMID: 39519708 PMCID: PMC11547630 DOI: 10.3390/molecules29215067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Sodium-glucose co-transporter 1 (SGLT1) is primarily expressed on the membrane of enterocytes, a type of epithelial cell found in the intestines, where it mediates the unidirectional absorption of glucose and galactose. Beyond its well-established role in nutrient absorption, SGLT1 also plays a protective role in maintaining the integrity of the intestinal barrier. Specifically, the natural ligand of SGLT1 (d-glucose) and a synthetic C-glucoside developed by our group can induce a protective anti-inflammatory effect on the intestinal epithelium. In this paper, we report the creation of a small library of C-glycoside, putative ligands for SGLT1, to gain further insights into its unclear mechanism of action. Preliminary biological experiments performed on an in vitro model of doxorubicin-induced mucositis, a severe intestinal inflammatory condition, indicate that the aromatic moiety present in all the compounds of the library is crucial for biological activity, while the sugar component appears to have less influence. These findings will be exploited to develop new, more potent anti-inflammatory compounds and to better understand and rationalize the protective mechanism of action.
Collapse
Affiliation(s)
- Giuseppe D’Orazio
- Department of Chemistry, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy
| | - Barbara La Ferla
- Department of Earth and Environmental Sciences DISAT, Università degli Studi di Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| |
Collapse
|
10
|
Westerbeke FHM, Attaye I, Rios-Morales M, Nieuwdorp M. Glycaemic sugar metabolism and the gut microbiota: past, present and future. FEBS J 2024. [PMID: 39359099 DOI: 10.1111/febs.17293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 08/02/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Non-communicable diseases (NCDs), such as type 2 diabetes (T2D) and metabolic dysfunction-associated fatty liver disease, have reached epidemic proportions worldwide. The global increase in dietary sugar consumption, which is largely attributed to the production and widespread use of cheap alternatives such as high-fructose corn syrup, is a major driving factor of NCDs. Therefore, a comprehensive understanding of sugar metabolism and its impact on host health is imperative to rise to the challenge of reducing NCDs. Notably, fructose appears to exert more pronounced deleterious effects than glucose, as hepatic fructose metabolism induces de novo lipogenesis and insulin resistance through distinct mechanisms. Furthermore, recent studies have demonstrated an intricate relationship between sugar metabolism and the small intestinal microbiota (SIM). In contrast to the beneficial role of colonic microbiota in complex carbohydrate metabolism, sugar metabolism by the SIM appears to be less beneficial to the host as it can generate toxic metabolites. These fermentation products can serve as a substrate for fatty acid synthesis, imposing negative health effects on the host. Nevertheless, due to the challenging accessibility of the small intestine, our knowledge of the SIM and its involvement in sugar metabolism remains limited. This review presents an overview of the current knowledge in this field along with implications for future research, ultimately offering potential therapeutic avenues for addressing NCDs.
Collapse
Affiliation(s)
- Florine H M Westerbeke
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, location AMC, The Netherlands
| | - Ilias Attaye
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, location AMC, The Netherlands
| | - Melany Rios-Morales
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, location AMC, The Netherlands
| | - Max Nieuwdorp
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, location AMC, The Netherlands
| |
Collapse
|
11
|
Haynes J, Palaniappan B, Crutchley JM, Sundaram U. Regulation of Enterocyte Brush Border Membrane Primary Na-Absorptive Transporters in Human Intestinal Organoid-Derived Monolayers. Cells 2024; 13:1623. [PMID: 39404387 PMCID: PMC11482628 DOI: 10.3390/cells13191623] [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: 08/26/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
In the small intestine, sodium (Na) absorption occurs primarily via two apical transporters, Na-hydrogen exchanger 3 (NHE3) and Na-glucose cotransporter 1 (SGLT1). The two primary Na-absorptive pathways were previously shown to compensatorily regulate each other in rabbit and rat intestinal epithelial cells. However, whether NHE3 and SGLT1 regulate one another in normal human enterocytes is unknown, mainly due to a lack of appropriate experimental models. To investigate this, we generated 2D enterocyte monolayers from human jejunal 3D organoids and used small interfering RNAs (siRNAs) to knock down NHE3 or SGLT1. Molecular and uptake studies were performed to determine the effects on NHE3 and SGLT1 expression and activity. Knockdown of NHE3 by siRNA in enterocyte monolayers was verified by qPCR and Western blot analysis and resulted in reduced NHE3 activity. However, in NHE3 siRNA-transfected cells, SGLT1 activity was significantly increased. siRNA knockdown of SGLT1 was confirmed by qPCR and Western blot analysis and resulted in reduced SGLT1 activity. However, in SGLT1 siRNA-transfected cells, NHE3 activity was significantly increased. These results demonstrate for the first time the functionality of siRNA in patient-derived organoid monolayers. Furthermore, they show that the two primary Na absorptive pathways in human enterocytes reciprocally regulate one another.
Collapse
Affiliation(s)
| | | | | | - Uma Sundaram
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA
| |
Collapse
|
12
|
Socha-Banasiak A, Sakowicz A, Gaj Z, Kolejwa M, Gach A, Czkwianianc E. Intestinal fructose transporters GLUT5 and GLUT2 in children and adolescents with obesity and metabolic disorders. Adv Med Sci 2024; 69:349-355. [PMID: 39059468 DOI: 10.1016/j.advms.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/19/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE The excessive fructose intake including high-fructose corn syrup (HFCS) may be responsible for increase of obesity occurrence. This study was designed to find potential differences in duodenal fructose transporters on mRNA and protein levels between obese and normal weight children and adolescents. MATERIALS/METHODS We performed a cross-sectional study on a group of 106 hospitalized patients aged 12 to 18. Glucose transporter 2 (GLUT2) and glucose transporter 5 (GLUT5) mRNA as well as protein levels (ELISA and Western blot methods) were assessed in duodenal mucosa biopsies of the patients categorized as obese or normal weight. Additionally, the expression of the aforementioned transporters was analyzed in patients based on the presence of insulin resistance (IR) and metabolic syndrome (MS). RESULTS In children with obesity, increased duodenal protein levels of GLUT5 (Relative protein GLUT5 expression/ACTB) (0.027 ± 0.009 vs. 0.011 ± 0.006, p < 0.05) but not GLUT2 as compared with the normal weight group, were revealed. No significant differences in duodenal relative GLUT2 and GLUT5 genes expression between the studied groups were found. There was no relationship between the presence of IR or MS and intestinal mRNA GLUT2 and GLUT5 as well as GLUT2 protein expression. CONCLUSION The upregulation of the duodenal GLUT5 may contribute to obesity occurrence in children and adolescents.
Collapse
Affiliation(s)
- Anna Socha-Banasiak
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland.
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | - Zuzanna Gaj
- Center of Medical Laboratory Diagnostics and Screening, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | - Michał Kolejwa
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | - Agnieszka Gach
- Department of Genetics, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | - Elżbieta Czkwianianc
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| |
Collapse
|
13
|
Sæterstad S, Østvik AE, Hansen MD, Bruland T, van Beelen Granlund A. The effect of rs2910686 on ERAP2 expression in IBD and epithelial inflammatory response. J Transl Med 2024; 22:750. [PMID: 39123229 PMCID: PMC11316291 DOI: 10.1186/s12967-024-05532-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND ERAP2 is an aminopeptidase involved in antigen processing and presentation, and harbor genetic variants linked to several inflammatory diseases such as Inflammatory Bowel Disease (IBD). The lack of an ERAP2 gene homologue in mice has hampered functional studies, and most human studies have focused on cells of hematopoietic origin. Using an IBD biobank as vantage point, this study explores how genetic variation in ERAP2 affects gene expression in human-derived epithelial organoids upon proinflammatory stimulation. METHODS An IBD patient cohort was genotyped with regards to two single nucleotide polymorphisms (SNP) (rs2910686/rs2248374) associated with ERAP2 expression levels, and we examined the correlation between colon gene expression and genotype, specifically aiming to establish a relationship with ERAP2 expression proficiency. Human-derived colon organoids (colonoids) with known ERAP2 genotype were established and used to explore differences in whole genome gene expression between ERAP2-deficient (n = 4) and -proficient (n = 4) donors upon pro-inflammatory encounter. RESULTS When taking rs2910686 genotype into account, ERAP2 gene expression is upregulated in the inflamed colon of IBD patients. Colonoids upregulate ERAP2 upon IFNɣ stimulation, and ERAP2 expression proficiency is dependent on rs2910686 genotype. Colonoid genotyping confirms that mechanisms independent of the frequently studied SNP rs2248374 can cause ERAP2-deficiency. A total of 586 genes involved in various molecular mechanisms are differentially expressed between ERAP2 proficient- and deficient colonoids upon proinflammatory stimulation, including genes encoding proteins with the following molecular function: catalytic activity (AOC1, CPE, ANPEP and MEP1A), regulator activity (TNFSF9, MDK, GDF15, ILR6A, LGALS3 and FLNA), transmembrane transporter activity (SLC40A1 and SLC5A1), and extracellular matrix structural constituents (FGL2, HMCN2, and MUC17). CONCLUSIONS ERAP2 is upregulated in the inflamed IBD colon mucosa, and expression proficiency is highly correlated with genotype of rs2910686. While the SNP rs2248374 is commonly used to determine ERAP2 expressional proficiency, our data confirms that mechanisms independent of this SNP can lead to ERAP2 deficiency. Our data demonstrates that epithelial ERAP2 presence affects the inflammatory response in colonoids, suggesting a pleiotropic role of ERAP2 beyond MHC class I antigen processing.
Collapse
Affiliation(s)
- Siri Sæterstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann Elisabeth Østvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Marianne Doré Hansen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Atle van Beelen Granlund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway.
- Department of Pathology, St. Olav's University Hospital, Trondheim, Norway.
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
14
|
Lee SM, Park SY, Kim JY. Comparative evaluation of the antihyperglycemic effects of three extracts of sea mustard (Undaria pinnatifida): In vitro and in vivo studies. Food Res Int 2024; 190:114623. [PMID: 38945577 DOI: 10.1016/j.foodres.2024.114623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 07/02/2024]
Abstract
Undaria pinnatifida (UP) contains multiple bioactive substances, such as polyphenols, polysaccharides, and amino acids, which are associated with various biological properties. This study aimed to evaluate the antihyperglycemic effects of three extracts obtained from UP. UP was extracted under three different conditions: a low-temperature water extract at 50 °C (UPLW), a high-temperature water extract at 90 °C (UPHW), and a 70 % ethanol extract (UPE). Nontargeted chemical profiling using high-performance liquid chromatography-triple/time-of-flight mass spectrometry (HPLC-Triple TOF-MS/MS) was conducted on the three UP extracts. Subsequently, α-glucosidase inhibitory (AGI) activity, glucose uptake, and the mRNA expression of sodium/glucose cotransporter 1 (SGLT1) and glucose transporter 2 (GLUT2) were evaluated in Caco-2 cell monolayers. Furthermore, an oral carbohydrate tolerance test was performed on C57BL/6 mice. The mice were orally administered UP at 300 mg/kg body weight (B.W.), and the blood glucose level and area under the curve (AUC) were measured. Compared with glucose, UPLW, UPHW and UPE significantly inhibited both glucose uptake and the mRNA expression of SGLT1 and GLUT2 in Caco-2 cell monolayers. After glucose, maltose, and sucrose loading, the blood glucose levels and AUC of the UPLW group were significantly lower than those of the control group. These findings suggest that UPLW has antihyperglycemic effects by regulating glucose uptake through glucose transporters and can be expected to alleviate postprandial hyperglycemia. Therefore, UPLW may have potential as a functional food ingredient for alleviating postprandial hyperglycemia.
Collapse
Affiliation(s)
- Sung Min Lee
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232, Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea
| | - Soo-Yeon Park
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232, Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea
| | - Ji Yeon Kim
- Department of Food Science and Biotechnology, Seoul National University of Science and Technology, 232, Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea.
| |
Collapse
|
15
|
Quero J, Paesa M, Morales C, Mendoza G, Osada J, Teixeira JA, Ferreira-Santos P, Rodríguez-Yoldi MJ. Biological Properties of Boletus edulis Extract on Caco-2 Cells: Antioxidant, Anticancer, and Anti-Inflammatory Effects. Antioxidants (Basel) 2024; 13:908. [PMID: 39199154 PMCID: PMC11352050 DOI: 10.3390/antiox13080908] [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: 06/12/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Boletus edulis (BE) is a mushroom well known for its taste, nutritional value, and medicinal properties. The objective of this work was to study the biological effects of BE extracts on human colon carcinoma cells (Caco-2), evaluating parameters related to oxidative stress and inflammation. In this study, a hydroethanolic extract of BE was obtained by ohmic heating green technology. The obtained BE extracts are mainly composed of sugars (mainly trehalose), phenolic compounds (taxifolin, rutin, and ellagic acid), and minerals (K, P, Mg, Na, Ca, Zn, Se, etc.). The results showed that BE extracts were able to reduce cancer cell proliferation by the induction of cell cycle arrest at the G0/G1 stage, as well as cell death by autophagy and apoptosis, the alteration of mitochondrial membrane potential, and caspase-3 activation. The extracts modified the redox balance of the cell by increasing the ROS levels associated with a decrease in the thioredoxin reductase activity. Similarly, BE extracts attenuated Caco-2 inflammation by reducing both iNOS and COX-2 mRNA expression and COX-2 protein expression. In addition, BE extracts protected the intestine from the oxidative stress induced by H2O2. Therefore, this study provides information on the potential use of BE bioactive compounds as anticancer therapeutic agents and as functional ingredients to prevent oxidative stress in the intestinal barrier.
Collapse
Affiliation(s)
- Javier Quero
- Department of Pharmacology and Physiology, Forensic and Legal Medicine, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain; (J.Q.); (C.M.); (G.M.)
| | - Mónica Paesa
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain;
- Institute of Nanoscience and Materials of Aragon (INMA), CSIC-University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain
| | - Carmen Morales
- Department of Pharmacology and Physiology, Forensic and Legal Medicine, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain; (J.Q.); (C.M.); (G.M.)
| | - Gracia Mendoza
- Department of Pharmacology and Physiology, Forensic and Legal Medicine, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain; (J.Q.); (C.M.); (G.M.)
- Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain
| | - Jesús Osada
- Department of Biochemistry and Molecular Cell Biology, Veterinary Faculty, University of Zaragoza, 50009 Zaragoza, Spain;
- CIBERobn, ISCIII, IIS Aragón, IA2, 50009 Zaragoza, Spain
| | - José António Teixeira
- CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- LABBELS—Associate Laboratory, Braga/Guimarães, 4710-057 Braga, Portugal
| | - Pedro Ferreira-Santos
- Department of Chemical Engineering, Faculty of Science, University of Vigo, 32004 Ourense, Spain
- IAA—Instituto de Agroecoloxía e Alimentación, University of Vigo (Campus Auga), As Lagoas, 32004 Ourense, Spain
| | - María Jesús Rodríguez-Yoldi
- Department of Pharmacology and Physiology, Forensic and Legal Medicine, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain; (J.Q.); (C.M.); (G.M.)
- CIBERobn, ISCIII, IIS Aragón, IA2, 50009 Zaragoza, Spain
| |
Collapse
|
16
|
Ritter MJ, Amano I, van der Spek AH, Gower AC, Undeutsch HJ, Rodrigues VAP, Daniel HE, Hollenberg AN. Nuclear Receptor Corepressors NCOR1 and SMRT Regulate Metabolism via Intestinal Regulation of Carbohydrate Transport. Endocrinology 2024; 165:bqae100. [PMID: 39106294 PMCID: PMC11337007 DOI: 10.1210/endocr/bqae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/09/2024]
Abstract
Nuclear receptor action is mediated in part by the nuclear receptor corepressor 1 (NCOR1) and the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT). NCOR1 and SMRT regulate metabolic pathways that govern body mass, insulin sensitivity, and energy expenditure, representing an understudied area in the realm of metabolic health and disease. Previously, we found that NCOR1 and SMRT are essential for maintaining metabolic homeostasis and their knockout (KO) leads to rapid weight loss and hypoglycemia, which is not survivable. Because of a potential defect in glucose absorption, we sought to determine the role of NCOR1 and SMRT specifically in intestinal epithelial cells (IECs). We used a postnatal strategy to disrupt NCOR1 and SMRT throughout IECs in adult mice. These mice were characterized metabolically and underwent metabolic phenotyping, body composition analysis, and glucose tolerance testing. Jejunal IECs were isolated and profiled by bulk RNA sequencing. We found that the postnatal KO of NCOR1 and SMRT from IECs leads to rapid weight loss and hypoglycemia with a significant reduction in survival. This was accompanied by alterations in glucose metabolism and activation of fatty acid oxidation in IECs. Metabolic phenotyping confirmed a reduction in body mass driven by a loss of body fat without altered food intake. This appeared to be mediated by a reduction of key intestinal carbohydrate transporters, including SGLT1, GLUT2, and GLUT5. Intestinal NCOR1 and SMRT act in tandem to regulate glucose levels and body weight. This in part may be mediated by regulation of intestinal carbohydrate transporters.
Collapse
Affiliation(s)
- Megan J Ritter
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, New York, NY 10021, USA
| | - Izuki Amano
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, New York, NY 10021, USA
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Anne H van der Spek
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, New York, NY 10021, USA
- Department of Endocrinology, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam UMC, 1105 AZ Amsterdam, the Netherlands
| | - Adam C Gower
- Boston University Clinical and Translational Science Institute, Boston, MA 02118, USA
| | - Hendrik J Undeutsch
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, New York, NY 10021, USA
| | - Victor A P Rodrigues
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Hanix E Daniel
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, New York, NY 10021, USA
| | - Anthony N Hollenberg
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, New York, NY 10021, USA
| |
Collapse
|
17
|
Ribeiro KS, Karmakar E, Park C, Garg R, Kung GP, Kadakia I, Gopianand JS, Arun T, Kisselev O, Gnana-Prakasam JP. Iron Regulates Cellular Proliferation by Enhancing the Expression of Glucose Transporter GLUT3 in the Liver. Cells 2024; 13:1147. [PMID: 38994998 PMCID: PMC11240476 DOI: 10.3390/cells13131147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
Iron is often accumulated in the liver during pathological conditions such as cirrhosis and cancer. Elevated expression of glucose transporters GLUT1 and GLUT3 is associated with reduced overall survival in patients with hepatocellular carcinoma. However, it is not known whether iron can regulate glucose transporters and contribute to tumor proliferation. In the present study, we found that treatment of human liver cell line HepG2 with ferric ammonium citrate (FAC) resulted in a significant upregulation of GLUT3 mRNA and protein in a dose-dependent manner. Similarly, iron accumulation in mice fed with high dietary iron as well as in mice injected intraperitoneally with iron dextran enhanced the GLUT3 expression drastically in the liver. We demonstrated that iron-induced hepatic GLUT3 upregulation is mediated by the LKB1/AMPK/CREB1 pathway, and this activation was reversed when treated with iron chelator deferiprone. In addition, inhibition of GLUT3 using siRNA prevented iron-mediated increase in the expression of cell cycle markers and cellular hyperproliferation. Furthermore, exogenous sodium beta-hydroxybutyrate treatment prevented iron-mediated hepatic GLUT3 activation both in vitro and in vivo. Together, these results underscore the importance of iron, AMPK, CREB1 and GLUT3 pathways in cell proliferation and highlight the therapeutic potential of sodium beta-hydroxybutyrate in hepatocellular carcinoma with high GLUT3 expression.
Collapse
Affiliation(s)
- Kleber S Ribeiro
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - Eshani Karmakar
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - Christine Park
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - Richa Garg
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - George P Kung
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - Isha Kadakia
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | | | - Tejas Arun
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - Oleg Kisselev
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | | |
Collapse
|
18
|
Rud CL, Hvistendahl MK, Langdahl B, Kraglund F, Baunwall SMD, Lal S, Jeppesen PB, Hvas CL. Protein-based oral rehydration solutions for patients with an ileostomy: A randomised, double-blinded crossover study. Clin Nutr 2024; 43:1747-1758. [PMID: 38850996 DOI: 10.1016/j.clnu.2024.05.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND & AIM Patients with an ileostomy are at increased risk of dehydration and sodium depletion. Treatments recommended may include oral rehydration solutions (ORS). We aimed to investigate if protein type or protein hydrolysation affects absorption from iso-osmolar ORS in patients with an ileostomy. METHODS This was a randomised, double-blinded, active comparator-controlled 3 × 3 crossover intervention study. We developed three protein-based ORS with whey protein isolate, caseinate or whey protein hydrolysate. The solutions contained 40-48 g protein/L, 34-45 mmol sodium/L and had an osmolality of 248-270 mOsm/kg. The patients ingested 500 mL/d. The study consisted of three 4-week periods with a >2-week washout between each intervention. The primary outcome was wet-weight ileostomy output. Ileostomy output and urine were collected for a 24-h period before and after each intervention. Additionally, blood sampling, dietary records, muscle-strength tests, bioimpedance analyses, questionnaires and psychometric tests were conducted. RESULTS We included 14 patients, of whom 13 completed at least one intervention. Ten patients completed all three interventions. Wet-weight ileostomy output did not change following either of the three interventions and did not differ between interventions (p = 0.38). A cluster of statistically significant improvements related to absorption was observed following the intake of whey protein isolate ORS, including decreased faecal losses of energy (-365 kJ/d, 95% confidence interval (CI), -643 to -87, p = 0.012), potassium (-7.8 mmol/L, 95%CI, -12.0 to -3.6, p = 0.001), magnesium (-4.0 mmol/L, 95%CI, -7.4 to -0.7, p = 0.020), improved plasma aldosterone (-4674 pmol/L 95%CI, -8536 to -812, p = 0.019), estimated glomerular filtration rate (eGFR) (2.8 mL/min/1.73 m2, 95%CI, 0.3 to 5.4, p = 0.03) and CO2 (1.7 mmol/L 95%CI, 0.1 to 3.3, p = 0.04). CONCLUSION Ingestion of 500 mL/d of iso-osmolar solutions containing either whey protein isolate, caseinate or whey protein hydrolysate for four weeks resulted in unchanged and comparable ileostomy outputs in patients with an ileostomy. Following whey protein isolate ORS, we observed discrete improvements in a series of absorption proxies in both faeces and blood, indicating increased absorption. The protein-based ORS were safe and well-tolerated. Treatments should be tailored to each patient, and future studies are warranted to explore treatment-effect heterogeneity and whether different compositions or doses of ORS can improve absorption and nutritional status in patients with an ileostomy. CLINICALTRIALS GOV STUDY IDENTIFIER NCT04141826.
Collapse
Affiliation(s)
- Charlotte Lock Rud
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark.
| | - Mark Krogh Hvistendahl
- Department of Intestinal Failure and Liver Diseases, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Bente Langdahl
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; Department of Endocrinology and Internal Medicine, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Frederik Kraglund
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Simon Mark Dahl Baunwall
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Simon Lal
- Intestinal Failure Unit, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Palle Bekker Jeppesen
- Department of Intestinal Failure and Liver Diseases, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Christian Lodberg Hvas
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
19
|
Ontawong A, Pengnet S, Thim-Uam A, Vaddhanaphuti CS, Munkong N, Phatsara M, Kuntakhut K, Inchai J, Amornlerdpison D, Rattanaphot T. Red rice bran aqueous extract ameliorate diabetic status by inhibiting intestinal glucose transport in high fat diet/STZ-induced diabetic rats. J Tradit Complement Med 2024; 14:391-402. [PMID: 39035687 PMCID: PMC11259718 DOI: 10.1016/j.jtcme.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/14/2023] [Accepted: 12/24/2023] [Indexed: 07/23/2024] Open
Abstract
Red rice (Oryza sativa L.) consumption has grown recently, partly due to its potential health benefits in several disease prevention. The impact of red rice bran aqueous extract (RRBE) on intestinal glucose uptake and diabetes mellitus (DM) progression has not been thoroughly investigated. This study aimed to evaluate the effect of RRBE on ex vivo intestinal glucose absorption and its potential as an antihyperglycemic compound using a high-fat diet and streptozotocin (STZ)-induced diabetic rats. High-fat diet/STZ-induced diabetic rats were supplemented with either 1000 mg/kg body weight (BW) of RRBE, 70 mg/kg BW of metformin (Met), or a combination of RRBE and Met for 3 months. Plasma parameters, intestinal glucose transport, morphology, liver and soleus muscle glycogen accumulation were assessed. Treatment with RRBE, metformin, or combination markedly reversed hyperglycemia, hypertriglyceridemia, insulin resistance, and pancreatic morphology changes associated with T2DM. Correspondingly, all supplements effectively downregulated glucose transporters, resulting in a reduction of intestinal glucose transport-additionally, liver and soleus muscle glycogen accumulation was reduced in RRBE + Met treated group. Taken together, RRBE potentially suppressed intestinal glucose transporters' function and expression, reducing diabetic status.
Collapse
Affiliation(s)
- Atcharaporn Ontawong
- Division of Physiology, School of Medical Sciences, University of Phayao, 19 Moo 2 Mae-Ka District, Muang, Phayao, 56000, Thailand
| | - Sirinat Pengnet
- Division of Physiology, School of Medical Sciences, University of Phayao, 19 Moo 2 Mae-Ka District, Muang, Phayao, 56000, Thailand
| | - Arthid Thim-Uam
- Division of Biochemistry, School of Medical Sciences, University of Phayao, 19 Moo 2 Mae-Ka District, Muang, Phayao, 56000, Thailand
| | - Chutima S. Vaddhanaphuti
- Faculty of Medicine, Chiang Mai University, 110 Faculty of Medicine, CMU, Inthawarorot Rd., Sri Phum, Muang, Chiang Mai, 50200, Thailand
| | - Narongsuk Munkong
- Department of Pathology, School of Medicine, University of Phayao, 19 Moo 2 Mae-Ka District, Muang, Phayao, 56000, Thailand
| | - Manussaborn Phatsara
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 52000, Thailand
| | - Kullanat Kuntakhut
- Center of Excellence in Agricultural Innovation for Graduate Entrepreneur, Maejo University, 63, Sansai-Phrao Street, Sansai, Chiang Mai, 50290, Thailand
| | - Jakkapong Inchai
- Faculty of Medicine, Chiang Mai University, 110 Faculty of Medicine, CMU, Inthawarorot Rd., Sri Phum, Muang, Chiang Mai, 50200, Thailand
| | - Doungporn Amornlerdpison
- Center of Excellence in Agricultural Innovation for Graduate Entrepreneur, Maejo University, 63, Sansai-Phrao Street, Sansai, Chiang Mai, 50290, Thailand
- Faculty of Fisheries Technology and Aquatic Resources, Maejo University, Chiang Mai, 50290, Thailand
| | - Teerawat Rattanaphot
- Center of Excellence in Agricultural Innovation for Graduate Entrepreneur, Maejo University, 63, Sansai-Phrao Street, Sansai, Chiang Mai, 50290, Thailand
| |
Collapse
|
20
|
Bauer I, Rimbach G, Cordeiro S, Bosy-Westphal A, Weghuber J, Ipharraguerre IR, Lüersen K. A comprehensive in-vitro/ in-vivo screening toolbox for the elucidation of glucose homeostasis modulating properties of plant extracts (from roots) and its bioactives. Front Pharmacol 2024; 15:1396292. [PMID: 38989154 PMCID: PMC11233739 DOI: 10.3389/fphar.2024.1396292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Plant extracts are increasingly recognized for their potential in modulating (postprandial) blood glucose levels. In this context, root extracts are of particular interest due to their high concentrations and often unique spectrum of plant bioactives. To identify new plant species with potential glucose-lowering activity, simple and robust methodologies are often required. For this narrative review, literature was sourced from scientific databases (primarily PubMed) in the period from June 2022 to January 2024. The regulatory targets of glucose homeostasis that could be modulated by bioactive plant compounds were used as search terms, either alone or in combination with the keyword "root extract". As a result, we present a comprehensive methodological toolbox for studying the glucose homeostasis modulating properties of plant extracts and its constituents. The described assays encompass in-vitro investigations involving enzyme inhibition (α-amylase, α-glucosidase, dipeptidyl peptidase 4), assessment of sodium-dependent glucose transporter 1 activity, and evaluation of glucose transporter 4 translocation. Furthermore, we describe a patch-clamp technique to assess the impact of extracts on KATP channels. While validating in-vitro findings in living organisms is imperative, we introduce two screenable in-vivo models (the hen's egg test and Drosophila melanogaster). Given that evaluation of the bioactivity of plant extracts in rodents and humans represents the current gold standard, we include approaches addressing this aspect. In summary, this review offers a systematic guide for screening plant extracts regarding their influence on key regulatory elements of glucose homeostasis, culminating in the assessment of their potential efficacy in-vivo. Moreover, application of the presented toolbox might contribute to further close the knowledge gap on the precise mechanisms of action of plant-derived compounds.
Collapse
Affiliation(s)
- Ilka Bauer
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Gerald Rimbach
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Sönke Cordeiro
- Institute of Physiology, University of Kiel, Kiel, Germany
| | - Anja Bosy-Westphal
- Division of Human Nutrition, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Julian Weghuber
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria
- FFoQSI—Austrian Competence Centre for Feed and Food Quality, Safety & Innovation, Tulln, Austria
| | - Ignacio R. Ipharraguerre
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Kai Lüersen
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
21
|
Dagbasi A, Byrne C, Blunt D, Serrano-Contreras JI, Becker GF, Blanco JM, Camuzeaux S, Chambers E, Danckert N, Edwards C, Bernal A, Garcia MV, Hanyaloglu A, Holmes E, Ma Y, Marchesi J, Martinez-Gili L, Mendoza L, Tashkova M, Perez-Moral N, Garcia-Perez I, Robles AC, Sands C, Wist J, Murphy KG, Frost G. Diet shapes the metabolite profile in the intact human ileum, which affects PYY release. Sci Transl Med 2024; 16:eadm8132. [PMID: 38896603 DOI: 10.1126/scitranslmed.adm8132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The human ileum contains a high density of enteroendocrine L-cells, which release the appetite-suppressing hormones glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) in response to food intake. Recent evidence highlighted the potential role of food structures in PYY release, but the link between food structures, ileal metabolites, and appetite hormone release remains unclear owing to limited access to intact human ileum. In a randomized crossover trial (ISRCTN11327221; isrctn.com), we investigated the role of human ileum in GLP-1 and PYY release by giving healthy volunteers diets differing in fiber and food structure: high-fiber (intact or disrupted food structures) or low-fiber disrupted food structures. We used nasoenteric tubes to sample chyme from the intact distal ileum lumina of humans in the fasted state and every 60 min for 480 min postprandially. We demonstrate the highly dynamic, wide-ranging molecular environment of the ileum over time, with a substantial decrease in ileum bacterial numbers and bacterial metabolites after food intake. We also show that high-fiber diets, independent of food structure, increased PYY release compared with a low-fiber diet during 0 to 240 min postprandially. High-fiber diets also increased ileal stachyose, and a disrupted high-fiber diet increased certain ileal amino acids. Treatment of human ileal organoids with ileal fluids or an amino acid and stachyose mixture stimulated PYY expression in a similar profile to blood PYY concentrations, confirming the role of ileal metabolites in PYY release. Our study demonstrates the diet-induced changes over time in the metabolite environment of intact human ileum, which play a role in PYY release.
Collapse
Affiliation(s)
- Aygul Dagbasi
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Claire Byrne
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Dominic Blunt
- Department of Imaging, Charing Cross Hospital, Imperial NHS Trust, London W6 8RF, UK
| | - Jose Ivan Serrano-Contreras
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Georgia Franco Becker
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Jesus Miguens Blanco
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Stephane Camuzeaux
- National Phenome Centre, Imperial College London, Hammersmith Hospital Campus, London W12 0HS, UK
| | - Edward Chambers
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Nathan Danckert
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | | | - Andres Bernal
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Maria Valdivia Garcia
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Aylin Hanyaloglu
- Institute of Reproductive and Development Biology (IRDB), Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Hammersmith Hospital, London W12 0NN, UK
| | - Elaine Holmes
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Yue Ma
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Julian Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Laura Martinez-Gili
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London W12 0NN, UK
| | - Lilian Mendoza
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Martina Tashkova
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | | | - Isabel Garcia-Perez
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Andres Castillo Robles
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Caroline Sands
- National Phenome Centre, Imperial College London, Hammersmith Hospital Campus, London W12 0HS, UK
| | - Julien Wist
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
- Centre for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Kevin G Murphy
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| | - Gary Frost
- Section of Nutrition, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
22
|
Riemma MA, Mele E, Donniacuo M, Telesca M, Bellocchio G, Castaldo G, Rossi F, De Angelis A, Cappetta D, Urbanek K, Berrino L. Glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors, anti-diabetic drugs in heart failure and cognitive impairment: potential mechanisms of the protective effects. Front Pharmacol 2024; 15:1422740. [PMID: 38948473 PMCID: PMC11212466 DOI: 10.3389/fphar.2024.1422740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024] Open
Abstract
Heart failure and cognitive impairment emerge as public health problems that need to be addressed due to the aging global population. The conditions that often coexist are strongly related to advancing age and multimorbidity. Epidemiological evidence indicates that cardiovascular disease and neurodegenerative processes shares similar aspects, in term of prevalence, age distribution, and mortality. Type 2 diabetes increasingly represents a risk factor associated not only to cardiometabolic pathologies but also to neurological conditions. The pathophysiological features of type 2 diabetes and its metabolic complications (hyperglycemia, hyperinsulinemia, and insulin resistance) play a crucial role in the development and progression of both heart failure and cognitive dysfunction. This connection has opened to a potential new strategy, in which new classes of anti-diabetic medications, such as glucagon-like peptide-1 receptor (GLP-1R) agonists and sodium-glucose cotransporter 2 (SGLT2) inhibitors, are able to reduce the overall risk of cardiovascular events and neuronal damage, showing additional protective effects beyond glycemic control. The pleiotropic effects of GLP-1R agonists and SGLT2 inhibitors have been extensively investigated. They exert direct and indirect cardioprotective and neuroprotective actions, by reducing inflammation, oxidative stress, ions overload, and restoring insulin signaling. Nonetheless, the specificity of pathways and their contribution has not been fully elucidated, and this underlines the urgency for more comprehensive research.
Collapse
Affiliation(s)
- Maria Antonietta Riemma
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Elena Mele
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Maria Donniacuo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Marialucia Telesca
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Gabriella Bellocchio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giuseppe Castaldo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- CEINGE-Advanced Biotechnologies, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Donato Cappetta
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- CEINGE-Advanced Biotechnologies, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
23
|
Cho H, Huh KM, Cho HJ, Kim B, Shim MS, Cho YY, Lee JY, Lee HS, Kwon YJ, Kang HC. Beyond nanoparticle-based oral drug delivery: transporter-mediated absorption and disease targeting. Biomater Sci 2024; 12:3045-3067. [PMID: 38712883 DOI: 10.1039/d4bm00313f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Various strategies at the microscale/nanoscale have been developed to improve oral absorption of therapeutics. Among them, gastrointestinal (GI)-transporter/receptor-mediated nanosized drug delivery systems (NDDSs) have drawn attention due to their many benefits, such as improved water solubility, improved chemical/physical stability, improved oral absorption, and improved targetability of their payloads. Their therapeutic potential in disease animal models (e.g., solid tumors, virus-infected lungs, metastasis, diabetes, and so on) has been investigated, and could be expanded to disease targeting after systemic/lymphatic circulation, although the detailed paths and mechanisms of endocytosis, endosomal escape, intracellular trafficking, and exocytosis through the epithelial cell lining in the GI tract are still unclear. Thus, this review summarizes and discusses potential GI transporters/receptors, their absorption and distribution, in vivo studies, and potential sequential targeting (e.g., oral absorption and disease targeting in organs/tissues).
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| | - Kang Moo Huh
- Department of Polymer Science and Engineering & Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyun Ji Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| | - Bogeon Kim
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
- Regulated Cell Death (RCD) Control Material Research Institute, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
- Regulated Cell Death (RCD) Control Material Research Institute, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
- Regulated Cell Death (RCD) Control Material Research Institute, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
- Regulated Cell Death (RCD) Control Material Research Institute, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| |
Collapse
|
24
|
Franco-Pérez J. Mechanisms Underlying Memory Impairment Induced by Fructose. Neuroscience 2024; 548:27-38. [PMID: 38679409 DOI: 10.1016/j.neuroscience.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/04/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Fructose consumption has increased over the years, especially in adolescents living in urban areas. Growing evidence indicates that daily fructose consumption leads to some pathological conditions, including memory impairment. This review summarizes relevant data describing cognitive deficits after fructose intake and analyzes the underlying neurobiological mechanisms. Preclinical experiments show sex-related deficits in spatial memory; that is, while males exhibit significant imbalances in spatial processing, females seem unaffected by dietary supplementation with fructose. Recognition memory has also been evaluated; however, only female rodents show a significant decline in the novel object recognition test performance. According to mechanistic evidence, fructose intake induces neuroinflammation, mitochondrial dysfunction, and oxidative stress in the short term. Subsequently, these mechanisms can trigger other long-term effects, such as inhibition of neurogenesis, downregulation of trophic factors and receptors, weakening of synaptic plasticity, and long-term potentiation decay. Integrating all these neurobiological mechanisms will help us understand the cellular and molecular processes that trigger the memory impairment induced by fructose.
Collapse
Affiliation(s)
- Javier Franco-Pérez
- Laboratorio Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, CDMX, México, Mexico.
| |
Collapse
|
25
|
Xu S, Chen Y, Gong Y. Improvement of Theaflavins on Glucose and Lipid Metabolism in Diabetes Mellitus. Foods 2024; 13:1763. [PMID: 38890991 PMCID: PMC11171799 DOI: 10.3390/foods13111763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
In diabetes mellitus, disordered glucose and lipid metabolisms precipitate diverse complications, including nonalcoholic fatty liver disease, contributing to a rising global mortality rate. Theaflavins (TFs) can improve disorders of glycolipid metabolism in diabetic patients and reduce various types of damage, including glucotoxicity, lipotoxicity, and other associated secondary adverse effects. TFs exert effects to lower blood glucose and lipids levels, partly by regulating digestive enzyme activities, activation of OATP-MCT pathway and increasing secretion of incretins such as GIP. By the Ca2+-CaMKK ꞵ-AMPK and PI3K-AKT pathway, TFs promote glucose utilization and inhibit endogenous glucose production. Along with the regulation of energy metabolism by AMPK-SIRT1 pathway, TFs enhance fatty acids oxidation and reduce de novo lipogenesis. As such, the administration of TFs holds significant promise for both the prevention and amelioration of diabetes mellitus.
Collapse
Affiliation(s)
- Shiyu Xu
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China;
- Key Laboratory of Tea Science of Ministry of Education, Changsha 410128, China
| | - Ying Chen
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China;
- Key Laboratory of Tea Science of Ministry of Education, Changsha 410128, China
| | - Yushun Gong
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
26
|
Chen Z, Chen J, Wang L, Wang W, Zheng J, Wu S, Sun Y, Pan Y, Li S, Liu M, Cai Z. Effects of Three Kinds of Carbohydrate Pharmaceutical Excipients-Fructose, Lactose and Arabic Gum on Intestinal Absorption of Gastrodin through Glucose Transport Pathway in Rats. Pharm Res 2024; 41:1201-1216. [PMID: 38834905 DOI: 10.1007/s11095-024-03720-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Some glucoside drugs can be transported via intestinal glucose transporters (IGTs), and the presence of carbohydrate excipients in pharmaceutical formulations may influence the absorption of them. This study, using gastrodin as probe drug, aimed to explore the effects of fructose, lactose, and arabic gum on intestinal drug absorption mediated by the glucose transport pathway. METHODS The influence of fructose, lactose, and arabic gum on gastrodin absorption was assessed via pharmacokinetic experiments and single-pass intestinal perfusion. The expression of sodium-dependent glucose transporter 1 (SGLT1) and sodium-independent glucose transporter 2 (GLUT2) was quantified via RT‒qPCR and western blotting. Alterations in rat intestinal permeability were evaluated through H&E staining, RT‒qPCR, and immunohistochemistry. RESULTS Fructose reduced the area under the curve (AUC) and peak concentration (Cmax) of gastrodin by 42.7% and 63.71%, respectively (P < 0.05), and decreased the effective permeability coefficient (Peff) in the duodenum and jejunum by 58.1% and 49.2%, respectively (P < 0.05). SGLT1 and GLUT2 expression and intestinal permeability remained unchanged. Lactose enhanced the AUC and Cmax of gastrodin by 31.5% and 65.8%, respectively (P < 0.05), and increased the Peff in the duodenum and jejunum by 33.7% and 26.1%, respectively (P < 0.05). SGLT1 and GLUT2 levels did not significantly differ, intestinal permeability increased. Arabic gum had no notable effect on pharmacokinetic parameters, SGLT1 or GLUT2 expression, or intestinal permeability. CONCLUSION Fructose, lactose, and arabic gum differentially affect intestinal drug absorption through the glucose transport pathway. Fructose competitively inhibited drug absorption, while lactose may enhance absorption by increasing intestinal permeability. Arabic gum had no significant influence.
Collapse
Affiliation(s)
- Zhenzhen Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiasheng Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Liyang Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wentao Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiaqi Zheng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shiqiong Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yinzhu Sun
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yuru Pan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sai Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Menghua Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Zheng Cai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China.
| |
Collapse
|
27
|
Ma Y, Wei X, Peng J, Wei F, Wen Y, Liu M, Song B, Wang Y, Zhang Y, Peng T. Ephedra sinica polysaccharide regulate the anti-inflammatory immunity of intestinal microecology and bacterial metabolites in rheumatoid arthritis. Front Pharmacol 2024; 15:1414675. [PMID: 38846095 PMCID: PMC11153800 DOI: 10.3389/fphar.2024.1414675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Ephedra sinica polysaccharide (ESP) exerts substantial therapeutic effects on rheumatoid arthritis (RA). However, the mechanism through which ESP intervenes in RA remains unclear. A close correlation has been observed between enzymes and derivatives in the gut microbiota and the inflammatory immune response in RA. Methods A type II collagen-induced arthritis (CIA) mice model was treated with Ephedra sinica polysaccharide. The therapeutic effect of ESP on collagen-induced arthritis mice was evaluated. The anti-inflammatory and cartilage-protective effects of ESP were also evaluated. Additionally, metagenomic sequencing was performed to identify changes in carbohydrate-active enzymes and resistance genes in the gut microbiota of the ESP-treated CIA mice. Liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry were performed to observe the levels of serum metabolites and short-chain fatty acids in the gut. Spearman's correlational analysis revealed a correlation among the gut microbiota, antibiotic-resistance genes, and microbiota-derived metabolites. Results ESP treatment significantly reduced inflammation levels and cartilage damage in the CIA mice. It also decreased the levels of pro-inflammatory cytokines interleukin (IL)-6, and IL-1-β and protected the intestinal mucosal epithelial barrier, inhibiting inflammatory cell infiltration and mucosal damage. Here, ESP reduced the TLR4, MyD88, and TRAF6 levels in the synovium, inhibited the p65 expression and pp65 phosphorylation in the NF-κB signaling pathway, and blocked histone deacetylase (HDAC1 and HDAC2) signals. ESP influenced the gut microbiota structure, microbial carbohydrate-active enzymes, and microbial resistance related to resistance genes. ESP increased the serum levels of L-tyrosine, sn-glycero-3-phosphocholine, octadecanoic acid, N-oleoyl taurine, and decreased N-palmitoyl taurine in the CIA mice. Conclusion ESP exhibited an inhibitory effect on RA. Its action mechanism may be related to the ability of ESP to effectively reduce pro-inflammatory cytokines levels, protect the intestinal barrier, and regulate the interaction between mucosal immune systems and abnormal local microbiota. Accordingly, immune homeostasis was maintained and the inhibition of fibroblast-like synoviocyte (FLS) proliferation through the HDAC/TLR4/NF-κB pathway was mediated, thereby contributing to its anti-inflammatory and immune-modulating effects.
Collapse
Affiliation(s)
- Yanmiao Ma
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xiuhong Wei
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jiehao Peng
- Department of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Fuxia Wei
- Department of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Ya Wen
- Department of First Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Mingran Liu
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Bo Song
- Department of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yonghui Wang
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yumin Zhang
- Department of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Tao Peng
- Famous Chinese Medicine Studio, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine, Taiyuan, China
- Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine Taiyuan, Taiyuan, China
| |
Collapse
|
28
|
Watabe E, Kawanabe A, Kamitori K, Ichihara S, Fujiwara Y. Sugar binding of sodium-glucose cotransporters analyzed by voltage-clamp fluorometry. J Biol Chem 2024; 300:107215. [PMID: 38522518 PMCID: PMC11061222 DOI: 10.1016/j.jbc.2024.107215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/19/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
Sugar absorption is crucial for life and relies on glucose transporters, including sodium-glucose cotransporters (SGLTs). Although the structure of SGLTs has been resolved, the substrate selectivity of SGLTs across diverse isoforms has not been determined owing to the complex substrate-recognition processes and limited analysis methods. Therefore, this study used voltage-clamp fluorometry (VCF) to explore the substrate-binding affinities of human SGLT1 in Xenopus oocytes. VCF analysis revealed high-affinity binding of D-glucose and D-galactose, which are known transported substrates. D-fructose, which is not a transported substrate, also bound to SGLT1, suggesting potential recognition despite the lack of transport activity. VCF analysis using the T287N mutant of the substrate-binding pocket, which has reduced D-glucose transport capacity, showed that its D-galactose-binding affinity exceeded its D-glucose-binding affinity. This suggests that the change in the VCF signal was due to substrate binding to the binding pocket. Both D-fructose and L-sorbose showed similar binding affinities, indicating that SGLT1 preferentially binds to pyranose-form sugars, including D-fructopyranose. Electrophysiological analysis confirmed that D-fructose binding did not affect the SGLT1 transport function. The significance of the VCF assay lies in its ability to measure sugar-protein interactions in living cells, thereby bridging the gap between structural analyses and functional characterizations of sugar transporters. Our findings also provide insights into SGLT substrate selectivity and the potential for developing medicines with reduced side effects by targeting non-glucose sugars with low bioreactivity.
Collapse
Affiliation(s)
- Erika Watabe
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| | - Akira Kawanabe
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| | - Kazuyo Kamitori
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan; International Institute of Rare Sugar Research and Education, Kagawa University, Miki-cho, Kagawa, Japan
| | - Satoko Ichihara
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| | - Yuichiro Fujiwara
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan; International Institute of Rare Sugar Research and Education, Kagawa University, Miki-cho, Kagawa, Japan; Laboratory of Physiology and Biophysics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima City, Hiroshima, Japan.
| |
Collapse
|
29
|
Ran R, Muñoz Briones J, Jena S, Anderson NL, Olson MR, Green LN, Brubaker DK. Detailed survey of an in vitro intestinal epithelium model by single-cell transcriptomics. iScience 2024; 27:109383. [PMID: 38523788 PMCID: PMC10959667 DOI: 10.1016/j.isci.2024.109383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/01/2023] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
The co-culture of two adult human colorectal cancer cell lines, Caco-2 and HT29, on Transwell is commonly used as an in vitro gut mimic, yet the translatability of insights from such a system to adult human physiological contexts is not fully characterized. Here, we used single-cell RNA sequencing on the co-culture to obtain a detailed survey of cell type heterogeneity in the system and conducted a holistic comparison with human physiology. We identified the intestinal stem cell-, transit amplifying-, enterocyte-, goblet cell-, and enteroendocrine-like cells in the system. In general, the co-culture was fetal intestine-like, with less variety of gene expression compared to the adult human gut. Transporters for major types of nutrients were found in the majority of the enterocytes-like cells in the system. TLR 4 was not expressed in the sample, indicating that the co-culture model is incapable of mimicking the innate immune aspect of the human epithelium.
Collapse
Affiliation(s)
- Ran Ran
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Javier Muñoz Briones
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Purdue Interdisciplinary Life Science Program, West Lafayette, IN, USA
| | - Smrutiti Jena
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Nicole L. Anderson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Matthew R. Olson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Leopold N. Green
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Douglas K. Brubaker
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH, USA
| |
Collapse
|
30
|
Mondal S, Bandyopadhyay A. Glucose transporters (GLUTs): Underreported yet crucial molecules in unraveling testicular toxicity. Biochimie 2024; 219:55-62. [PMID: 37967737 DOI: 10.1016/j.biochi.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Glucose transporters (GLUTs) are crucial in maintaining glucose homeostasis and supporting energy production in various tissues, including the testes. This review article delves into the distribution and function of GLUTs in distinct testicular cell types, namely Leydig cells, Sertoli cells, germ cells, and spermatozoa, shedding light on their significance in the context of male reproductive health-an issue of mounting global concern. Furthermore, this article examines the implications of GLUT dysregulation in testicular dysfunction. Altered GLUT expression has been associated with impaired steroidogenesis, spermatogenesis, sperm count, and motility in various animal models. Lastly, the article underscores the potential therapeutic implications of targeting GLUTs concerning testicular toxicity. Insights gleaned from studies in diabetes and cancer suggest that modulating GLUT expression and translocation could present novel strategies for mitigating testicular dysfunction and safeguarding male fertility. In summary, the intricate interplay between GLUTs, glucose metabolism, and testicular health underscores the significance of sustaining testicular glucose homeostasis for male reproductive health. Manipulating GLUTs presents an innovative avenue to address testicular dysfunction, potentially revolutionizing therapeutic strategies to restore male fertility and overall reproductive well-being. Future research in this field holds great promise for advancing male fertility treatments and reproductive health interventions.
Collapse
Affiliation(s)
- Shirsha Mondal
- Department of Zoology, Govt. College Dhimarkheda (Rani Durgavati Vishwavidyalaya), Katni, 483 332, Madhya Pradesh, India.
| | - Arindam Bandyopadhyay
- Department of Zoology, University of Allahabad, Prayagraj, 211 002, Uttar Pradesh, India.
| |
Collapse
|
31
|
Paul S, Pallavi A, Gandasi NR. Exploring the potential of pheophorbide A, a chlorophyll-derived compound in modulating GLUT for maintaining glucose homeostasis. Front Endocrinol (Lausanne) 2024; 15:1330058. [PMID: 38529398 PMCID: PMC10961331 DOI: 10.3389/fendo.2024.1330058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Pheophorbide A, a chlorophyll-breakdown product, is primarily investigated for its anti-oxidant and anti-inflammatory activity. Recent reports on pheophorbide A have shown its potential in lowering blood glucose levels, thus leading to the exploration of its use in diabetes management. Literature has also shown its effect on enhanced insulin secretion, whereas its mechanism on glucose stimulated insulin secretion (GSIS) in pancreatic β cells remains unexplored. Methods In-silico and in-vitro investigations were used to explore the effect of pheophorbide A on class I glucose transporters (GLUTs). In-silico studies include - Molecular docking studies and stability assessment using GROMACS. In-vitro studies include - MTT assay, Glucose uptake assay, Live-cell imaging and tracking of GLUTs in presence of Pheophorbide A compared to control. Results Molecular docking studies revealed better binding affinity of pheophorbide A with GLUT4 (-11.2 Kcal/mol) and GLUT1 (-10.7 Kcal/mol) when compared with metformin (-5.0 Kcal/mol and -4.9 Kcal/mol, respectively). Glucose levels are largely regulated by GLUTs where GLUT1 is one of the transporters that is ubiquitously present in human β cells. Thus, we confirmed the stability of the complex, that is, pheophorbide A-GLUT1 using GROMACS for 100 ns. We further assessed its effect on a pancreatic β cell line (INS-1) for its viability using an MTT assay. Pheophorbide A (0.1-1 µM) showed a dose-dependent response on cell viability and was comparable to standard metformin. To assess how pheophorbide A mechanistically acts on GLUT1 in pancreatic β cell, we transfected INS-1 cells with GLUT1-enhanced green fluorescent protein and checked how the treatment of pheophorbide A (0.50 µM) modulates GLUT1 trafficking using live-cell imaging. We observed a significant increase in GLUT1 density when treated with pheophorbide A (0.442 ± 0.01 µm-2) at 20 mM glucose concentration when compared to GLUT1 control (0.234 ± 0.01 µm-2) and metformin (0.296 ± 0.02 µm-2). The average speed and distance travelled by GLUT1 puncta were observed to decrease when treated with pheophorbide A. The present study also demonstrated the potential of pheophorbide A to enhance glucose uptake in β cells. Conclusion The current study's findings were validated by in-silico and cellular analyses, suggesting that pheophorbide A may regulate GLUT1 and might be regarded as a potential lead for boosting the GSIS pathway, thus maintaining glucose homeostasis.
Collapse
Affiliation(s)
- Saptadipa Paul
- Cell Metabolism Lab (GA-08), Department of Developmental Biology and Genetics (DBG), Indian Institute of Science (IISc), Bengaluru, India
| | - Anuma Pallavi
- Cell Metabolism Lab (GA-08), Department of Developmental Biology and Genetics (DBG), Indian Institute of Science (IISc), Bengaluru, India
| | - Nikhil R. Gandasi
- Cell Metabolism Lab (GA-08), Department of Developmental Biology and Genetics (DBG), Indian Institute of Science (IISc), Bengaluru, India
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
32
|
Sakizli U, Takano T, Yoo SK. GALDAR: A genetically encoded galactose sensor for visualizing sugar metabolism in vivo. PLoS Biol 2024; 22:e3002549. [PMID: 38502638 PMCID: PMC10950222 DOI: 10.1371/journal.pbio.3002549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Sugar metabolism plays a pivotal role in sustaining life. Its dynamics within organisms is less understood compared to its intracellular metabolism. Galactose, a hexose stereoisomer of glucose, is a monosaccharide transported via the same transporters with glucose. Galactose feeds into glycolysis and regulates protein glycosylation. Defects in galactose metabolism are lethal for animals. Here, by transgenically implementing the yeast galactose sensing system into Drosophila, we developed a genetically encoded sensor, GALDAR, which detects galactose in vivo. Using this heterologous system, we revealed dynamics of galactose metabolism in various tissues. Notably, we discovered that intestinal stem cells do not uptake detectable levels of galactose or glucose. GALDAR elucidates the role for galactokinase in metabolism of galactose and a transition of galactose metabolism during the larval period. This work provides a new system that enables analyses of in vivo sugar metabolism.
Collapse
Affiliation(s)
- Uğurcan Sakizli
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
- Division of Developmental Biology and Regenerative Medicine, Kobe University, Kobe, Japan
| | - Tomomi Takano
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
| | - Sa Kan Yoo
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
- Division of Developmental Biology and Regenerative Medicine, Kobe University, Kobe, Japan
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
| |
Collapse
|
33
|
Zhao BC, Wang TH, Chen J, Qiu BH, Xu YR, Li JL. Essential oils improve nursery pigs' performance and appetite via modulation of intestinal health and microbiota. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:174-188. [PMID: 38357573 PMCID: PMC10864218 DOI: 10.1016/j.aninu.2023.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/22/2023] [Accepted: 10/12/2023] [Indexed: 02/16/2024]
Abstract
Optimal intestinal health and functionality are essential for animal health and performance, and simultaneously intestinal nutrient transporters and intestinal peptides are also involved in appetite and feed intake control mechanisms. Given the potential of essential oil (EO) in improving animal performance and improving feed palatability, we hypothesized that dietary supplementation of cinnamaldehyde and carvacrol could improve performance and appetite of nursery pigs by modulating intestinal health and microbiota. Cinnamaldehyde (100 mg/kg), carvacrol (100 mg/kg), and their mixtures (including 50 mg/kg cinnamaldehyde and 50 mg/kg carvacrol) were supplemented into the diets of 240 nursery pigs for 42 d, and data related to performance were measured. Thereafter, the influence of EO on intestinal health, appetite and gut microbiota and their correlations were explored. EO supplementation increased (P < 0.05) the body weight, average daily gain (ADG) and average daily feed intake (ADFI) of piglets, and reduced (P < 0.05) diarrhea rates in nursery pigs. Furthermore, EO increased (P < 0.05) the intestinal absorption area and the abundance of tight junction proteins, and decreased (P < 0.05) intestinal permeability and local inflammation. In terms of intestinal development and the mucus barrier, EO promoted intestinal development and increased (P < 0.05) the number of goblet cells. Additionally, we found that piglets in the EO-supplemented group had upregulated (P < 0.05) levels of transporters and digestive enzymes in the intestine, which were significantly associated with daily gain and feed utilization. In addition, EO supplementation somewhat improved appetite in nursery pigs, increased the diversity of the gut microbiome and the abundance of beneficial bacteria, and there was a correlation between altered bacterial structure and appetite-related hormones. These findings indicate that EO is effective in promoting growth performance and nutrient absorption as well as in regulating appetite by improving intestinal health and bacterial structure.
Collapse
Affiliation(s)
- Bi-Chen Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tian-Hao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jian Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bai-Hao Qiu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ya-Ru Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| |
Collapse
|
34
|
López-Mejía L, Guillén-Lopez S, Vela-Amieva M, Santillán-Martínez R, Abreu M, González-Herrra MD, Díaz-Martínez R, Reyes-Magaña JG. Importance of genetic sequencing studies in managing chronic neonatal diarrhea: a case report of a novel variant in the glucose-galactose transporter SLC5A1. Front Pediatr 2024; 12:1284671. [PMID: 38440183 PMCID: PMC10909829 DOI: 10.3389/fped.2024.1284671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/02/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction Congenital glucose-galactose malabsorption (CGGM) is a rare autosomal recessive disorder that primarily causes chronic intractable diarrhea. This study aims to describe the clinical history, laboratory profile, diagnostic workflow, and management of the first patient reported with CGGM in Mexico. Methods The case involves a Mexican female infant with recurrent admissions to the emergency room since birth due to chronic diarrhea. Results The infant was born at term by C-section with a birth weight of 3.120 kg and height of 48 cm for consanguineous parents. She had been breastfed until day 5 of her life when she presented lethargy, diarrhea, abdominal discomfort, and jaundice. During the first evaluation at the emergency room, the significant laboratory finding was blood tyrosine elevation; afterward, amino acid and succinylacetone determinations were obtained, discarding tyrosinemia. When admitted to the hospital, an abdominal ultrasound detected a duplex collecting system. At this time, rice formula was introduced to the patient. She was discharged with jaundice improvement, but diarrhea persisted. Several formula changes had been made from rice to extensively hydrolyzed casein protein to whey-based, with no clinical improvement; the patient still had 10-12 excretions daily. In the second hospitalization, the patient presented anemia, severe dehydration, hyperammonemia, and renal tubular acidosis. A next-generation sequencing panel for inborn errors of metabolism and congenital diarrhea was performed, identifying a homozygous variant in SLC5A1 (c.1667T > C). The diagnosis of CGGM was made at 3 months of age. The infant was initially treated with a modular galactose-glucose-free formula with oil, fructose, casein, minerals, and vitamins until a commercial fructose-based formula was introduced. This led to a complete resolution of diarrhea and improved nutritional status. Discussion Diagnosing CGGM is challenging for clinicians, and next-generation sequencing is a valuable tool for providing appropriate treatment. More detailed information on patients with this condition might lead to possible phenotype-genotype correlations. This case's primary clinical and biochemical findings were chronic diarrhea, anemia, jaundice, renal tubular acidosis, hyperammonemia, and initial hypertyrosinemia. Symptoms were resolved entirely with the fructose-based formula.
Collapse
Affiliation(s)
- Lizbeth López-Mejía
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Sara Guillén-Lopez
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | - Melania Abreu
- Laboratorio de Biología Molecular, Genos Medica, Mexico City, Mexico
- Centro de Cáncer, Centro Médico ABC, Mexico City, Mexico
| | | | - Rubicel Díaz-Martínez
- Servicio de Genetica, Hospital del Niño Dr. Rodolfo Nieto Padrón, Villahermosa, Mexico
| | | |
Collapse
|
35
|
Klip A, De Bock K, Bilan PJ, Richter EA. Transcellular Barriers to Glucose Delivery in the Body. Annu Rev Physiol 2024; 86:149-173. [PMID: 38345907 DOI: 10.1146/annurev-physiol-042022-031657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Glucose is the universal fuel of most mammalian cells, and it is largely replenished through dietary intake. Glucose availability to tissues is paramount for the maintenance of homeostatic energetics and, hence, supply should match demand by the consuming organs. In its journey through the body, glucose encounters cellular barriers for transit at the levels of the absorbing intestinal epithelial wall, the renal epithelium mediating glucose reabsorption, and the tight capillary endothelia (especially in the brain). Glucose transiting through these cellular barriers must escape degradation to ensure optimal glucose delivery to the bloodstream or tissues. The liver, which stores glycogen and generates glucose de novo, must similarly be able to release it intact to the circulation. We present the most up-to-date knowledge on glucose handling by the gut, liver, brain endothelium, and kidney, and discuss underlying molecular mechanisms and open questions. Diseases associated with defects in glucose delivery and homeostasis are also briefly addressed. We propose that the universal problem of sparing glucose from catabolism in favor of translocation across the barriers posed by epithelia and endothelia is resolved through common mechanisms involving glucose transfer to the endoplasmic reticulum, from where glucose exits the cells via unconventional cellular mechanisms.
Collapse
Affiliation(s)
- Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Erik A Richter
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Albaik M, Sheikh Saleh D, Kauther D, Mohammed H, Alfarra S, Alghamdi A, Ghaboura N, Sindi IA. Bridging the gap: glucose transporters, Alzheimer's, and future therapeutic prospects. Front Cell Dev Biol 2024; 12:1344039. [PMID: 38298219 PMCID: PMC10824951 DOI: 10.3389/fcell.2024.1344039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Glucose is the major source of chemical energy for cell functions in living organisms. The aim of this mini-review is to provide a clearer and simpler picture of the fundamentals of glucose transporters as well as the relationship of these transporters to Alzheimer's disease. This study was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). Electronic databases (PubMed and ScienceDirect) were used to search for relevant studies mainly published during the period 2018-2023. This mini-review covers the two main types of glucose transporters, facilitated glucose transporters (GLUTs) and sodium-glucose linked transporters (SGLTs). The main difference between these two types is that the first type works through passive transport across the glucose concentration gradient. The second type works through active co-transportation to transport glucose against its chemical gradient. Fluctuation in glucose transporters translates into a disturbance of normal functioning, such as Alzheimer's disease, which may be caused by a significant downregulation of GLUTs most closely associated with insulin resistance in the brain. The first sign of Alzheimer's is a lack of GLUT4 translocation. The second sign is tau hyperphosphorylation, which is caused by GLUT1 and 3 being strongly upregulated. The current study focuses on the use of glucose transporters in treating diseases because of their proven therapeutic potential. Despite this, studies remain insufficient and inconclusive due to the complex and intertwined nature of glucose transport processes. This study recommends further understanding of the mechanisms related to these vectors for promising future therapies.
Collapse
Affiliation(s)
- Mai Albaik
- Department of Chemistry Preparatory Year Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | | | - Dana Kauther
- Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Hajira Mohammed
- Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Shurouq Alfarra
- Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Adel Alghamdi
- Department of Biology Preparatory Year Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmacy Practice Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
37
|
Liu J, Liu K, Wang Y, Shi Z, Xu R, Zhang Y, Li J, Liu C, Xue B. Death receptor 5 is required for intestinal stem cell activity during intestinal epithelial renewal at homoeostasis. Cell Death Dis 2024; 15:27. [PMID: 38199990 PMCID: PMC10782029 DOI: 10.1038/s41419-023-06409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024]
Abstract
Intestinal epithelial renewal, which depends on the proliferation and differentiation of intestinal stem cells (ISCs), is essential for epithelial homoeostasis. Understanding the mechanism controlling ISC activity is important. We found that death receptor 5 (DR5) gene deletion (DR5-/-) mice had impaired epithelial absorption and barrier function, resulting in delayed weight gain, which might be related to the general reduction of differentiated epithelial cells. In DR5-/- mice, the expression of ISC marker genes, the number of Olfm4+ ISCs, and the number of Ki67+ and BrdU+ cells in crypt were reduced. Furthermore, DR5 deletion inhibited the expression of lineage differentiation genes driving ISC differentiation into enterocytes, goblet cells, enteroendocrine cells, and Paneth cells. Therefore, DR5 gene loss may inhibit the intestinal epithelial renewal by dampening ISC activity. The ability of crypts from DR5-/- mice to form organoids decreased, and selective DR5 activation by Bioymifi promoted organoid growth and the expression of ISC and intestinal epithelial cell marker genes. Silencing of endogenous DR5 ligand TRAIL in organoids down-regulated the expression of ISC and intestinal epithelial cell marker genes. So, DR5 expressed in intestinal crypts was involved in the regulation of ISC activity. DR5 deletion in vivo or activation in organoids inhibited or enhanced the activity of Wnt, Notch, and BMP signalling through regulating the production of Paneth cell-derived ISC niche factors. DR5 gene deletion caused apoptosis and DNA damage in transit amplifying cells by inhibiting ERK1/2 activity in intestinal crypts. Inhibition of ERK1/2 with PD0325901 dampened the ISC activity and epithelial regeneration. In organoids, when Bioymifi's effect in activating ERK1/2 activity was completely blocked by PD0325901, its role in stimulating ISC activity and promoting epithelial regeneration was also eliminated. In summary, DR5 in intestinal crypts is essential for ISC activity during epithelial renewal under homoeostasis.
Collapse
Affiliation(s)
- Jianbo Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kaixuan Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Wang
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ziru Shi
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Runze Xu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yundi Zhang
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingxin Li
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanyong Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Xue
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
38
|
Lam TP, Tran NVN, Pham LHD, Lai NVT, Dang BTN, Truong NLN, Nguyen-Vo SK, Hoang TL, Mai TT, Tran TD. Flavonoids as dual-target inhibitors against α-glucosidase and α-amylase: a systematic review of in vitro studies. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:4. [PMID: 38185713 PMCID: PMC10772047 DOI: 10.1007/s13659-023-00424-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024]
Abstract
Diabetes mellitus remains a major global health issue, and great attention is directed at natural therapeutics. This systematic review aimed to assess the potential of flavonoids as antidiabetic agents by investigating their inhibitory effects on α-glucosidase and α-amylase, two key enzymes involved in starch digestion. Six scientific databases (PubMed, Virtual Health Library, EMBASE, SCOPUS, Web of Science, and WHO Global Index Medicus) were searched until August 21, 2022, for in vitro studies reporting IC50 values of purified flavonoids on α-amylase and α-glucosidase, along with corresponding data for acarbose as a positive control. A total of 339 eligible articles were analyzed, resulting in the retrieval of 1643 flavonoid structures. These structures were rigorously standardized and curated, yielding 974 unique compounds, among which 177 flavonoids exhibited inhibition of both α-glucosidase and α-amylase are presented. Quality assessment utilizing a modified CONSORT checklist and structure-activity relationship (SAR) analysis were performed, revealing crucial features for the simultaneous inhibition of flavonoids against both enzymes. Moreover, the review also addressed several limitations in the current research landscape and proposed potential solutions. The curated datasets are available online at https://github.com/MedChemUMP/FDIGA .
Collapse
Affiliation(s)
- Thua-Phong Lam
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Uppsala University, 75105, Uppsala, Sweden
| | - Ngoc-Vi Nguyen Tran
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Uppsala University, 75105, Uppsala, Sweden
| | - Long-Hung Dinh Pham
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam
- Department of Chemistry, Imperial College London, London, W12 0BZ, UK
| | - Nghia Vo-Trong Lai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam
| | - Bao-Tran Ngoc Dang
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam
| | - Ngoc-Lam Nguyen Truong
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam
| | - Song-Ky Nguyen-Vo
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam
| | - Thuy-Linh Hoang
- California Northstate University College of Pharmacy, California, 95757, USA
| | - Tan Thanh Mai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam.
| | - Thanh-Dao Tran
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000, Ho Chi Minh City, Vietnam.
| |
Collapse
|
39
|
Yamamoto K, Harada N, Yasuda T, Hatoko T, Wada N, Lu X, Seno Y, Kurihara T, Yamane S, Inagaki N. Intestinal Morphology and Glucose Transporter Gene Expression under a Chronic Intake of High Sucrose. Nutrients 2024; 16:196. [PMID: 38257088 PMCID: PMC10820040 DOI: 10.3390/nu16020196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/30/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Sucrose is a disaccharide that is degraded into fructose and glucose in the small intestine. High-sucrose and high-fructose diets have been reported, using two-dimensional imaging, to alter the intestinal morphology and the expression of genes associated with sugar transport, such as sodium glucose co-transporter 1 (SGLT1), glucose transporter 2 (GLUT2), and glucose transporter 5 (GLUT5). However, it remains unclear how high-fructose and high-sucrose diets affect the expression of sugar transporters and the intestinal morphology in the whole intestine. We investigate the influence of a chronic high-sucrose diet on the expression of the genes associated with sugar transport as well as its effects on the intestinal morphology using 3D imaging. High sucrose was found to increase GLUT2 and GLUT5 mRNA levels without significant changes in the intestinal morphology using 3D imaging. On the other hand, the delay in sucrose absorption by an α-glucosidase inhibitor significantly improved the intestinal morphology and the expression levels of SGLT1, GLUT2, and GLUT5 mRNA in the distal small intestine to levels similar to those in the proximal small intestine, thereby improving glycemic control after both glucose and sucrose loading. These results reveal the effects of chronic high-sugar exposure on glucose absorption and changes in the intestinal morphology.
Collapse
Affiliation(s)
- Kana Yamamoto
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Norio Harada
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takuma Yasuda
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Tomonobu Hatoko
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Naoki Wada
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Xuejing Lu
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Youhei Seno
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takashi Kurihara
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shunsuke Yamane
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Nobuya Inagaki
- P.I.I.F. Tazuke-Kofukai Medical Research Institute, Kitano Hospital, Osaka 530-8480, Japan
| |
Collapse
|
40
|
Tang X, Zeng Y, Xiong K, Li M. The inflammatory injury of porcine small intestinal epithelial cells induced by deoxynivalenol is related to the decrease in glucose transport. J Anim Sci 2024; 102:skae107. [PMID: 38619320 PMCID: PMC11069187 DOI: 10.1093/jas/skae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/13/2024] [Indexed: 04/16/2024] Open
Abstract
The present study aimed to investigate the effects of deoxynivalenol (DON) stimulation on inflammatory injury and the expression of the glucose transporters sodium-dependent glucose transporter 1 (SGLT1) and glucose transporter protein 2 (GLU2) in porcine small intestinal epithelial cells (IPEC-J2). Additionally, the study aimed to provide initial insights into the connection between the expression of glucose transporters and the inflammatory injury of IPEC-J2 cells. DON concentration and DON treatment time were determined using the CCK‑8 assay. Accordingly, 1.0 µg/mL DON and treatment for 24 h were chosen for subsequent experiments. Then IPEC-J2 cells were treated without DON (CON, N = 6) or with 1 μg/mL DON (DON, N = 6). Lactate dehydrogenase (LDH) content, apoptosis rate, and proinflammatory cytokines including interleukin (IL)-1β, Il-6, and tumor necrosis factor α (TNF-α) were measured. Additionally, the expression of AMP-activated protein kinase α1 (AMPK-α1), the content of glucose, intestinal alkaline phosphatase (AKP), and sodium/potassium-transporting adenosine triphosphatase (Na+/K+-ATPase) activity, and the expression of SGLT1 and GLU2 of IPEC-J2 cells were also analyzed. The results showed that DON exposure significantly increased LDH release and apoptosis rate of IPEC-J2 cells. Stimulation with DON resulted in significant cellular inflammatory damage, as evidenced by a significant increase in proinflammatory cytokines (IL-1β, IL-6, and TNF-α). Additionally, DON caused damage to the glucose absorption capacity of IPEC-J2 cells, indicated by decreased levels of glucose content, AKP activity, Na+/K+-ATPase activity, AMPK-α1 protein expression, and SGLT1 expression. Correlation analysis revealed that glucose absorption capacity was negatively correlated with cell inflammatory cytokines. Based on the findings of this study, it can be preliminarily concluded that the cell inflammatory damage caused by DON may be associated with decreased glucose absorption.
Collapse
Affiliation(s)
- Xiaopeng Tang
- State Engineering Technology Institute for Karst Desertfication Control, School of Karst Science, Guizhou Normal University, Guiyang 5500025, China
| | - Yan Zeng
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550025, China
| | - Kangning Xiong
- State Engineering Technology Institute for Karst Desertfication Control, School of Karst Science, Guizhou Normal University, Guiyang 5500025, China
| | - Meijun Li
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha 410127, China
| |
Collapse
|
41
|
Zhao Q, Yang J, Li J, Zhang L, Yan X, Yue T, Yuan Y. Hypoglycemic effect and intestinal transport of phenolics-rich extract from digested mulberry leaves in Caco-2/insulin-resistant HepG2 co-culture model. Food Res Int 2024; 175:113689. [PMID: 38129030 DOI: 10.1016/j.foodres.2023.113689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Phenolics of mulberry (Morus alba L.) leaves (MLs) have potential anti-diabetic effects, but they may be chemically modified during gastrointestinal digestion so affect their biological activity. In this study, an in vitro digestion model coupled with Caco-2 monolayer and Caco-2/insulin-resistant HepG2 coculture model were used to study the transport and hypoglycemic effects of phenolics in raw MLs (U-MLs) and solid-fermented MLs (F-MLs). The results of LC-MS/MS analysis showed that the Papp (apparent permeability coefficient, 10-6cm/s) of phenolics in digested MLs ranged from 0.002 ± 0.00 (quercetin 3-O-glucoside) to 60.19 ± 0.67 (ferulic acid), indicating higher phenolic acids absorbability and poor flavonoids absorbability. The Papp values of phenolic extracts of F-MLs in Caco-2 monolayer were significantly higher (p > 0.05) than that of U-MLs. Digested phenolic extracts inhibited the activities of sucrase (60.13 ± 2.03 %) and maltase (82.35 ± 0.78 %) and decreased 9.28 ± 0.84 % of glucose uptake in Caco-2 monolayer. Furthermore, a decrease in the mRNA expression of glucose transporters SGLT1 (0.64 ± 0.18), GLUT2 (0.14 ± 0.02) and the sucrase-isomaltase (0.59 ± 0.00) was observed. In Caco-2/insulin-resistant HepG2 co-culture model, phenolic extracts regulated glucose metabolism by up-regulating the mRNA expressions of IRS1 (9.32-fold), Akt (17.07-fold) and GYS2 (1.5-fold), and down-regulating the GSK-3β (0.22-fold), PEPCK (0.49-fold) and FOXO1 (0.10-fold) mRNA levels. Both U-MLs and F-MLs could improve glucose metabolism, and the partial least squares (PLS) analysis showed that luteoforol and p-coumaric acid were the primary phenolics that strongly correlated with the hypoglycemic ability of MLs. Results suggested that phenolics of MLs can be used as dietary supplements to regulate glucose metabolism.
Collapse
Affiliation(s)
- Qiannan Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Jinyi Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Jiahui Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Lei Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xiaohai Yan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Tianli Yue
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; College of Food Science and Techonology, Northwest University, Xi'an 710069, China.
| | - Yahong Yuan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; College of Food Science and Techonology, Northwest University, Xi'an 710069, China.
| |
Collapse
|
42
|
Numata S, Oishee MJ, McDermott J, Koepsell H, Vallon V, Blanco G. Deletion of the Sodium Glucose Cotransporter 1 (Sglt-1) impairs mouse sperm movement. Mol Reprod Dev 2024; 91:e23723. [PMID: 38282316 DOI: 10.1002/mrd.23723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 01/30/2024]
Abstract
The Sodium Glucose Cotransporter Isoform 1 (Sglt-1) is a symporter that moves Na+ and glucose into the cell. While most studies have focused on the role of Sglt-1 in the small intestine and kidney, little is known about this transporter's expression and function in other tissues. We have previously shown that Sglt-1 is expressed in the mouse sperm flagellum and that its inhibition interferes with sperm metabolism and function. Here, we further investigated the importance of Sglt-1 in sperm, using a Sglt-1 knockout mouse (Sglt-1 KO). RNA, immunocytochemistry, and glucose uptake analysis confirmed the ablation of Sglt-1 in sperm. Sglt-1 KO male mice are fertile and exhibit normal sperm counts and morphology. However, Sglt-1 null sperm displayed a significant reduction in total, progressive and other parameters of sperm motility compared to wild type (WT) sperm. The reduction in motility was exacerbated when sperm were challenged to swim in media with higher viscosity. Parameters of capacitation, namely protein tyrosine phosphorylation and acrosomal reaction, were similar in Sglt-1 KO and WT sperm. However, Sglt-1 KO sperm displayed a significant decrease in hyperactivation. The impaired motility of Sglt-1 null sperm was observed in media containing glucose as the only energy substrate. Interestingly, the addition of pyruvate and lactate to the media partially recovered sperm motility of Sglt-1 KO sperm, both in the low and high viscosity media. Altogether, these results support an important role for Sglt-1 in sperm energetics and function, providing sperm with a higher capacity for glucose uptake.
Collapse
Affiliation(s)
- September Numata
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mumtarin Jannat Oishee
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey McDermott
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hermann Koepsell
- Institute for Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Gustavo Blanco
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
43
|
Al-Busaidi A, Alabri O, Alomairi J, ElSharaawy A, Al Lawati A, Al Lawati H, Das S. Gut Microbiota and Insulin Resistance: Understanding the Mechanism of Better Treatment of Type 2 Diabetes Mellitus. Curr Diabetes Rev 2024; 21:e170124225723. [PMID: 38243954 DOI: 10.2174/0115733998281910231231051814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Gut microbiota refers to the population of trillions of microorganisms present in the human intestine. The gut microbiota in the gastrointestinal system is important for an individual's good health and well-being. The possibility of an intrauterine colonization of the placenta further suggests that the fetal environment before birth may also affect early microbiome development. Various factors influence the gut microbiota. Dysbiosis of microbiota may be associated with various diseases. Insulin regulates blood glucose levels, and disruption of the insulin signaling pathway results in insulin resistance. Insulin resistance or hyperinsulinemia is a pathological state in which the insulin-responsive cells have a diminished response to the hormone compared to normal physiological responses, resulting in reduced glucose uptake by the tissue cells. Insulin resistance is an important cause of type 2 diabetes mellitus. While there are various factors responsible for the etiology of insulin resistance, dysbiosis of gut microbiota may be an important contributing cause for metabolic disturbances. We discuss the mechanisms in skeletal muscles, adipose tissue, liver, and intestine by which insulin resistance can occur due to gut microbiota's metabolites. A better understanding of gut microbiota may help in the effective treatment of type 2 diabetes mellitus and metabolic syndrome.
Collapse
Affiliation(s)
- Alsalt Al-Busaidi
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Omer Alabri
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Jaifar Alomairi
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | | | | | - Hanan Al Lawati
- Pharmacy Program, Department of Pharmaceutics, Oman College of Health Sciences, Muscat 113, Oman
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
44
|
Cottam A, Cottam D, Roslin M, Surve A. Exploring Bariatric Surgery's Impact on Weight Loss and Diabetes: Sodium and Glucose Receptor Modulation. JSLS 2024; 28:e2023.00051. [PMID: 38562948 PMCID: PMC10984375 DOI: 10.4293/jsls.2023.00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
Sodium-glucose cotransporters (SGLT) and glucose transporters (GLUT) have been shown to influence diabetes management by modulating glucose uptake by the intestine. Therefore, alterations in gastrointestinal anatomy during bariatric surgery can change SGLT and GLUT receptor activity. These changes offer an additional mechanism for weight loss and may explain the differential impact of the various bariatric surgical procedures. This review examines the current literature on SGLT and GLUT receptors and their effects on weight loss through genetic studies, pharmacologic inhibition, and how SGLT/GLUT receptors impact surgical physiologic modulation. A better understanding of Type I sodium-glucose cotransport receptors (SGLT-1), GLUT-2, and GLUT-5 could provide insight for improved procedures and allow us to determine the best method to tailor operations to a patient's individual needs.
Collapse
Affiliation(s)
- Austin Cottam
- Bariatric Medicine Institute, Salt Lake City, Utah, USA. (Drs A. Cottam, D. Cottam, and Surve)
- Northwell Health-Lenox Hill Hospital, New York City, New York, USA. (Dr Roslin)
| | - Daniel Cottam
- Bariatric Medicine Institute, Salt Lake City, Utah, USA. (Drs A. Cottam, D. Cottam, and Surve)
- Northwell Health-Lenox Hill Hospital, New York City, New York, USA. (Dr Roslin)
| | - Mitchell Roslin
- Bariatric Medicine Institute, Salt Lake City, Utah, USA. (Drs A. Cottam, D. Cottam, and Surve)
- Northwell Health-Lenox Hill Hospital, New York City, New York, USA. (Dr Roslin)
| | - Amit Surve
- Bariatric Medicine Institute, Salt Lake City, Utah, USA. (Drs A. Cottam, D. Cottam, and Surve)
- Northwell Health-Lenox Hill Hospital, New York City, New York, USA. (Dr Roslin)
| |
Collapse
|
45
|
Barreto-Peixoto JA, Silva C, Costa ASG, Álvarez-Rivera G, Cifuentes A, Ibáñez E, Oliveira MBPP, Alves RC, Martel F, Andrade N. A Prunus avium L. Infusion Inhibits Sugar Uptake and Counteracts Oxidative Stress-Induced Stimulation of Glucose Uptake by Intestinal Epithelial (Caco-2) Cells. Antioxidants (Basel) 2023; 13:59. [PMID: 38247483 PMCID: PMC10812648 DOI: 10.3390/antiox13010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Sweet cherry (Prunus avium L.) is among the most valued fruits due to its organoleptic properties and nutritional worth. Cherry stems are rich in bioactive compounds, known for their anti-inflammatory and antioxidant properties. Innumerable studies have indicated that some bioactive compounds can modulate sugar absorption in the small intestine. In this study, the phenolic profile of a cherry stem infusion was investigated, as well as its capacity to modulate intestinal glucose and fructose transport in Caco-2 cells. Long-term (24 h) exposure to cherry stem infusion (25%, v/v) significantly reduced glucose (3H-DG) and fructose (14C-FRU) apical uptake, reduced the apical-to-basolateral Papp to 3H-DG, and decreased mRNA expression levels of the sugar transporters SGLT1, GLUT2 and GLUT5. Oxidative stress (induced by tert-butyl hydroperoxide) caused an increase in 3H-DG uptake, which was abolished by the cherry stem infusion. These findings suggest that cherry stem infusion can reduce the intestinal absorption of both glucose and fructose by decreasing the gene expression of their membrane transporters. Moreover, this infusion also appears to be able to counteract the stimulatory effect of oxidative stress upon glucose intestinal uptake. Therefore, it can be a potentially useful compound for controlling hyperglycemia, especially in the presence of increased intestinal oxidative stress levels.
Collapse
Affiliation(s)
- Juliana A. Barreto-Peixoto
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.B.-P.); (C.S.); (A.S.G.C.); (M.B.P.P.O.); (R.C.A.)
| | - Cláudia Silva
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.B.-P.); (C.S.); (A.S.G.C.); (M.B.P.P.O.); (R.C.A.)
| | - Anabela S. G. Costa
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.B.-P.); (C.S.); (A.S.G.C.); (M.B.P.P.O.); (R.C.A.)
| | - Gerardo Álvarez-Rivera
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolas Cabrera 9, 28049 Madrid, Spain; (G.Á.-R.); (A.C.); (E.I.)
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolas Cabrera 9, 28049 Madrid, Spain; (G.Á.-R.); (A.C.); (E.I.)
| | - Elena Ibáñez
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolas Cabrera 9, 28049 Madrid, Spain; (G.Á.-R.); (A.C.); (E.I.)
| | - M. Beatriz P. P. Oliveira
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.B.-P.); (C.S.); (A.S.G.C.); (M.B.P.P.O.); (R.C.A.)
| | - Rita C. Alves
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.B.-P.); (C.S.); (A.S.G.C.); (M.B.P.P.O.); (R.C.A.)
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, 4200-135 Porto, Portugal
| | - Nelson Andrade
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.B.-P.); (C.S.); (A.S.G.C.); (M.B.P.P.O.); (R.C.A.)
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
46
|
Pérez-Castillo ÍM, Williams JA, López-Chicharro J, Mihic N, Rueda R, Bouzamondo H, Horswill CA. Compositional Aspects of Beverages Designed to Promote Hydration Before, During, and After Exercise: Concepts Revisited. Nutrients 2023; 16:17. [PMID: 38201848 PMCID: PMC10781183 DOI: 10.3390/nu16010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Hypohydration can impair aerobic performance and deteriorate cognitive function during exercise. To minimize hypohydration, athletes are recommended to commence exercise at least euhydrated, ingest fluids containing sodium during long-duration and/or high-intensity exercise to prevent body mass loss over 2% and maintain elevated plasma osmolality, and rapidly restore and retain fluid and electrolyte homeostasis before a second exercise session. To achieve these goals, the compositions of the fluids consumed are key; however, it remains unclear what can be considered an optimal formulation for a hydration beverage in different settings. While carbohydrate-electrolyte solutions such as sports drinks have been extensively explored as a source of carbohydrates to meet fuel demands during intense and long-duration exercise, these formulas might not be ideal in situations where fluid and electrolyte balance is impaired, such as practicing exercise in the heat. Alternately, hypotonic compositions consisting of moderate to high levels of electrolytes (i.e., ≥45 mmol/L), mainly sodium, combined with low amounts of carbohydrates (i.e., <6%) might be useful to accelerate intestinal water absorption, maintain plasma volume and osmolality during exercise, and improve fluid retention during recovery. Future studies should compare hypotonic formulas and sports drinks in different exercise settings, evaluating different levels of sodium and/or other electrolytes, blends of carbohydrates, and novel ingredients for addressing hydration and rehydration before, during, and after exercise.
Collapse
Affiliation(s)
| | | | | | - Niko Mihic
- Real Madrid, Medical Services, 28055 Madrid, Spain; (J.L.-C.); (N.M.)
| | | | | | - Craig A. Horswill
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60608, USA;
| |
Collapse
|
47
|
Sharp LS, Sharp WT, Ng P. Remission of Type II Diabetes Mellitus after Duodenal Switch: the Contribution of Common Channel Length. Obes Surg 2023; 33:3841-3849. [PMID: 37816973 PMCID: PMC10687107 DOI: 10.1007/s11695-023-06870-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION The role of the common channel length in duodenal switch (DS) on remission of type II diabetes mellitus (DM), when stratifying patients based on diabetes severity, is not well understood. METHODS We retrospectively reviewed 341 consecutive patients with DM undergoing DS with one of three different common channel (CC) lengths (100 cm, 150 cm, and 200 cm), each with a fixed 300 cm alimentary limb (AL). Patients were stratified by insulin dependence (IDDM) versus non-insulin dependent diabetes (NIDDM). Data was collected at one year and at the last available follow-up. RESULTS The NIDDM group had a similar average HbA1c at last follow-up for each of the CC lengths. However, the IDDM group had lower average HbA1c with shorter CC lengths (100 cm = 5.4%, 150 cm = 6%, 200 cm = 6.4%, p < 0.05). Shorter CC lengths resulted in a greater proportion of patients achieving remission in the IDDM group (66%, 50%, 32% in the 100 cm, 150 cm, and 200 cm CC, respectively, p < 0.01). Improvements in HbA1c were independent of weight loss and average DiaRem scores were similar between CC lengths. Rates of nutritional deficiencies were higher in shorter common channel lengths. Revision for malnutrition was similar between common channel lengths (100 cm group: 3.7%; 150 cm group: 1.8%; 200 cm group: 0%, p = NS). CONCLUSIONS When the AL is fixed, shortening CC lengths results in improved glycemic control and remission of DM in patients with the need for insulin preoperatively. Milder forms of DM are treated well with any of the CC lengths.
Collapse
Affiliation(s)
- Lindsey S Sharp
- UNC Rex Healthcare, Rex Bariatric Specialists, 4207 Lake Boone Trail, Suite 210, Raleigh, NC, 27607, USA.
| | - William T Sharp
- UNC Rex Healthcare, Rex Bariatric Specialists, 4207 Lake Boone Trail, Suite 210, Raleigh, NC, 27607, USA
| | - Peter Ng
- UNC Rex Healthcare, Rex Bariatric Specialists, 4207 Lake Boone Trail, Suite 210, Raleigh, NC, 27607, USA
| |
Collapse
|
48
|
Subramaniam M, Loewen ME. Review: A species comparison of the kinetic homogeneous and heterogeneous organization of sodium-dependent glucose transport systems along the intestine. Comp Biochem Physiol A Mol Integr Physiol 2023; 285:111492. [PMID: 37536429 DOI: 10.1016/j.cbpa.2023.111492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
The targeted use of carbohydrates by feed and food industries to create balanced and cost-effective diets has generated a tremendous amount of research in carbohydrate digestion and absorption in different species. Specifically, this research has led us to a larger observation that identified different organizations of intestinal sodium-dependent glucose absorption across species, which has not been previously collated and reviewed. Thus, this review will compare the kinetic segregation of sodium-dependent glucose transport across the intestine of different species, which we have termed either homogeneous or heterogeneous systems. For instance, the pig follows a heterogeneous system of sodium-dependent glucose transport with a high-affinity, super-low-capacity (Ha/sLc) in the jejunum, and a high-affinity, super-high-capacity (Ha/sHc) in the ileum. This is achieved by multiple sodium-dependent glucose transporters contributing to each segment. In contrast, tilapia have a homogenous system characterized by high-affinity, high-capacity (Ha/Hc) throughout the intestine. Additionally, we are the first to report glucose transporter patterns across species presented from vertebrates to invertebrates. Finally, other kinetic transport systems are briefly covered to illustrate possible contributions/modulations to sodium-dependent glucose transporter organization. Overall, we present a new perspective on the organization of glucose absorption along the intestinal tract.
Collapse
Affiliation(s)
- Marina Subramaniam
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada
| | - Matthew E Loewen
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada.
| |
Collapse
|
49
|
Liu M, Shen J, Zhu X, Ju T, Willing BP, Wu X, Lu Q, Liu R. Peanut skin procyanidins reduce intestinal glucose transport protein expression, regulate serum metabolites and ameliorate hyperglycemia in diabetic mice. Food Res Int 2023; 173:113471. [PMID: 37803795 DOI: 10.1016/j.foodres.2023.113471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/05/2023] [Accepted: 09/10/2023] [Indexed: 10/08/2023]
Abstract
One of diabetic characteristics is the postprandial hyperglycemia. Inhibiting glucose uptake may be beneficial for controlling postprandial blood glucose levels and regulating the glucose metabolism Peanut skin procyanidins (PSP) have shown a potential for lowering blood glucose; however, the underlying mechanism through which PSP regulate glucose metabolism remains unknown. In the current study, we investigated the effect of PSP on intestinal glucose transporters and serum metabolites using a mouse model of diabetic mice. Results showed that PSP improved glucose tolerance and systemic insulin sensitivity, which coincided with decreased expression of sodium-glucose cotransporter 1 and glucose transporter 2 in the intestinal epithelium induced by an activation of the phospholipase C β2/protein kinase C signaling pathway. Moreover, untargeted metabolomic analysis of serum samples revealed that PSP altered arachidonic acid, sphingolipid, glycerophospholipid, bile acids, and arginine metabolic pathways. The study provides new insight into the anti-diabetic mechanism of PSP and a basis for further research.
Collapse
Affiliation(s)
- Min Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China
| | - Jinxin Shen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China
| | - Xiaoling Zhu
- Hubei Provincial Institute for Food Supervision and Test, Wuhan 430070, China
| | - Tingting Ju
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Benjamin P Willing
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Xin Wu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China
| | - Qun Lu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan 430000, China
| | - Rui Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430000, China; Wuhan Engineering Research Center of Bee Products on Quality and Safety Control, Wuhan 430000, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan 430000, China; Key Laboratory of Urban Agriculture in Central China, Ministry of Agriculture and Rural Affairs, China.
| |
Collapse
|
50
|
Nomier Y, Asaad GF, Salama A, Shabana ME, Alshahrani S, Firoz Alam M, Anwer T, Sultana S, ur Rehman Z, Khalid A. Explicit mechanistic insights of Prosopis juliflora extract in streptozotocin-induced diabetic rats at the molecular level. Saudi Pharm J 2023; 31:101755. [PMID: 37727228 PMCID: PMC10505680 DOI: 10.1016/j.jsps.2023.101755] [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: 04/11/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
Background The Ancient system of medicine showed the limelight on the use of herbal remedies and was found to possess minimal side effects and acceptable therapeutic outcomes. In this context, Prosopis juliflora gained importance in managing chronic diseases such as cancer, dermatological diseases, and chronic inflammatory disorders. Hence, P. juliflora was selected for further investigation associated with diabetes and inflammation. Aim The present study aimed to evaluate the anti-diabetic activity in chemically induced experimental rats and explore the nature of phytocomponents that may produce this activity. Methods Experimentally, diabetes was induced by a single administration of streptozotocin at 50 mg/kg intraperitoneally in Wistar rats. The animals were treated orally with P. juliflora at low and high doses (200 and 400 mg/kg) for 10 days. Blood collected from the retro-orbital plexus was analyzed for parameters like blood glucose levels, insulin, adiponectin, Keap1 and Nrf2. PPAR-γ, AMPK and GLUT 2 levels were analyzed in the pancreatic tissue. Besides, at the end of the experiment, animals were sacrificed, and the pancreatic tissue sections were subjected for histopathological, morphometrical and immune histochemical exploration. The phytochemical composition of the plant was investigated by GC-MS. Results The administration of P. juliflora higher dose showed a significant decrease (**p< 0.001) in blood glucose levels with a rise in adiponectin, PPARγ, Keap1, Nrf2, Glut 2, and AMPK significantly (**p< 0.001). The inflammatory cytokine TNFα was also estimated and was found to be lowered significantly (**p< 0.001) in test drug-treated animals. Furthermore, in the pancreatic tissue, the number of Islets, the area, and the number of β-cells were improved significantly with the sub-chronic treatment of P. juliflora extract. The structure and function of β-cells were also revamped. Conclusion The study results demonstrated a significant effect of P. juliflora on glycemic status, inflammatory condition, and the architecture of pancreatic tissue. In the identification and isolation process by GC MS, it was noticed that P. juliflora contained few phytochemical constituents from which it might be considered a promising drug for type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yousra Nomier
- Department of Pharmacology and Clinical Pharmacy, College of Medicine, and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Department of Pharmacology and Toxicology, Pharmacy College, Jazan University, P.O. Box 114 45142, Jazan, Saudi Arabia
| | - Gihan F. Asaad
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Abeer Salama
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Marwa E. Shabana
- Department of Pathology, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, Pharmacy College, Jazan University, P.O. Box 114 45142, Jazan, Saudi Arabia
| | - Mohammad Firoz Alam
- Department of Pharmacology and Toxicology, Pharmacy College, Jazan University, P.O. Box 114 45142, Jazan, Saudi Arabia
| | - Tarique Anwer
- Department of Pharmacology and Toxicology, Pharmacy College, Jazan University, P.O. Box 114 45142, Jazan, Saudi Arabia
| | - Shahnaz Sultana
- Department of Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114 45142, Jazan, Saudi Arabia
| | - Zia ur Rehman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114 45142, Jazan, Saudi Arabia
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, P.O. Box 114 45142, Jazan, Saudi Arabia
- Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Centre for Research, P.O. Box: 2424, Khartoum 11111, Sudan
| |
Collapse
|