1
|
Talebi G, Saffarian P, Hakemi-Vala M, Sadeghi A, Yadegar A. The effect of Helicobacter pylori-derived extracellular vesicles on glucose metabolism and induction of insulin resistance in HepG2 cells. Arch Physiol Biochem 2025; 131:316-327. [PMID: 39431628 DOI: 10.1080/13813455.2024.2418494] [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: 06/10/2024] [Revised: 09/23/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
Helicobacter pylori infection has been associated with the development of insulin resistance (IR). This study aimed to examine the effect of H. pylori-derived extracellular vesicles (EVs) on IR induction. EVs were derived from two H. pylori strains, and characterised by transmission electron microscopy and dynamic light scattering. Different concentrations of insulin were added to HepG2 cells to induce IR model. HepG2 cells were exposed to various concentrations of H. pylori-derived EVs to assess IR development. The gene expression of IRS1, AKT2, GLUT2, IL-6, SOCS3, c-Jun and miR-140 was examined using RT-qPCR. Glucose uptake analysis revealed insulin at 5 × 10 -7 mol/l and EVs at 50 µg/ml induced IR model in HepG2 cells. H. pylori-derived EVs downregulated the expression level of IRS1, AKT2, and GLUT2, and upregulated IL-6, SOCS3, c-Jun, and miR-140 expression in HepG2 cells. In conclusion, our findings propose a novel mechanism by which H. pylori-derived EVs could potentially induce IR.
Collapse
Affiliation(s)
- Ghazaleh Talebi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parvaneh Saffarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mojdeh Hakemi-Vala
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Ruan Z, Wang Y, Shi L, Yang XJ. Progress of research on glucose transporter proteins in hepatocellular carcinoma. World J Hepatol 2025; 17:104715. [DOI: 10.4254/wjh.v17.i3.104715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumour with high prevalence and mortality rate worldwide. Metabolic reprogramming of cancer cells may be a major factor in the process of this disease. Glucose transporter proteins (GLUTs) are members of the major facilitator superfamily of membrane transporters, playing a pivotal role in the metabolic reprogramming and tumour progression in HCC. This review discusses the advances in the study of GLUTs in HCC, including the expression patterns, functions and possibilities of GLUTs. In HCC, the expression levels of GLUTs are closely associated with tumour aggressiveness, metabolic reprogramming and prognosis. A series of inhibitors have been demonstrated efficacy in inhibiting HCC cell growth and glucose uptake in in vitro and in vivo models. These inhibitors offer a novel approach to HCC treatment by reducing the glucose metabolism of tumour cells, thereby impeding tumour growth, and concurrently enhancing the sensitivity to chemotherapeutic agents. This reminds us of the urgent need to elucidate GLUTs’ roles in HCC and to determine the most effective ways to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Zheng Ruan
- The First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yan Wang
- Division of Personnel, Gansu Provincial People’s Hospital, Lanzhou 730000, Gansu Province, China
| | - Lei Shi
- Department of General Surgery, The Second people’s Hospital of Lanzhou, Lanzhou 730000, Gansu Province, China
| | - Xiao-Jun Yang
- Department of General Surgery, Gansu Provincial People’s Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
3
|
Lee JI, Lee HM, Park JH, Lee YG. Improvement of Glucose Metabolism by Pennogenin 3-O-β-Chacotrioside via Activation of IRS/PI3K/Akt Signaling and Mitochondrial Respiration in Insulin-Resistant Hepatocytes. Mol Nutr Food Res 2025:e70010. [PMID: 40103416 DOI: 10.1002/mnfr.70010] [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: 09/01/2024] [Revised: 01/06/2025] [Accepted: 02/12/2025] [Indexed: 03/20/2025]
Abstract
SCOPE Insulin resistance (IR), which causes chronic hyperglycemia, has been one of the most prevalent components of metabolic syndrome over the centuries. Pennogenin 3-O-β-chacotrioside (P3C), the main steroid glycoside derived from Paris polyphylla, has been found to exert various biological activities. However, the exact role of P3C on glucose metabolism in the IR state remains unexplored. METHODS AND RESULTS To induce IR, AML12 cells were exposed to glucose (27 mM) and insulin (10 µg/mL) and then incubated with P3C (0.25 or 0.5 µM) for 24 h. The effects of P3C on glucose metabolism in insulin-resistant AML12 cells were evaluated through glucose consumption assays, real-time quantitative polymerase chain reaction (qPCR), Western blotting, and metabolic analysis for extracellular acidification rate (ECAR) and oxygen consumption rate (OCR). Our data showed that P3C significantly improved insulin sensitivity in AML12 hepatocytes with high glucose-induced IR. P3C stimulated insulin sensitivity and glucose uptake by activating the IRS/PI3K/Akt signaling pathway, which enhances glycogen synthesis and suppresses gluconeogenesis in insulin-resistant AML12 cells. In addition, P3C treatment increased the protein expression of p-AMPK and PGC1α, as well as the expression of oxidative phosphorylation complex proteins, potentially enhancing mitochondrial oxidative respiration. CONCLUSIONS Our findings imply that P3C could be a therapeutic option for improving metabolic abnormalities associated with IR.
Collapse
Affiliation(s)
- Jae-In Lee
- Precision Nutrition Research Group, Korea Food Research Institute (KFRI), Wanju, Republic of Korea
| | - Hee Min Lee
- Kimchi Industry Promotion Division, World Institute of Kimchi, Gwangju, Republic of Korea
| | - Jae-Ho Park
- Precision Nutrition Research Group, Korea Food Research Institute (KFRI), Wanju, Republic of Korea
| | - Yu Geon Lee
- Precision Nutrition Research Group, Korea Food Research Institute (KFRI), Wanju, Republic of Korea
| |
Collapse
|
4
|
Guan Y, Liu G, Tang F, Wu X, Shi J, Huang Q. Stress hyperglycemia in acute pancreatitis: From mechanisms to prognostic implications. Life Sci 2025; 365:123469. [PMID: 39956188 DOI: 10.1016/j.lfs.2025.123469] [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: 11/16/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Acute pancreatitis (AP) is an inflammatory reaction of the pancreas. When the disease is severe, it is often accompanied by destruction of the pancreatic islets, resulting in dysfunction of the endocrine system of the pancreas. Stress hyperglycemia is a transient increase in glucose during a critical illness, and its possible mechanism is related to abnormal glucose metabolism and insulin resistance due to the increased release of counterregulatory hormones and cytokines, such as glucagon, cortisol, and catecholamines. Numerous studies have shown that stress hyperglycemia is strongly associated with morbidity, mortality, and increased risk of post-acute pancreatitis diabetes in AP patients. Therefore, stress hyperglycemia may be a significant independent risk factor for poor clinical outcomes and prognosis in patients with AP. This article reviews the clinical features, risk factors, and mechanisms of action of stress hyperglycemia in AP and its influence on adverse clinical outcomes and the prognosis of inpatients with AP. For AP patients with stress hyperglycemia, it is necessary to comprehensively consider their blood glucose levels, daily habits, and complications to develop an appropriate treatment plan for hyperglycemia. Limited evidence indicates that in the case of acute hyperglycemia in critically ill patients, especially during the first 3 days of hospitalization, insulin therapy should not be undertaken if the blood glucose level does not exceed 10 mmol/L. However, some important questions related to clinical practice remain to be answered. More clinical trials and studies are needed in the future to provide a sufficient basis for clinical practice.
Collapse
Affiliation(s)
- Yuting Guan
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guoqing Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Feimin Tang
- Guangxi Medical University, Nanning, Guangxi, China
| | - Xiangmin Wu
- Guangxi Medical University, Nanning, Guangxi, China
| | - Jian Shi
- Department of Cardiology, The People's Hospital of Laibin, Laibin, Guangxi, China.
| | - Qiongguang Huang
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China; Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
5
|
Nguyen LTG, Tawfik SM, Jin J, Durwin A, Zhong XB. Impact on efficacy of target reduction of two FDA-approved ASO drugs by intracellular glucose levels in in vitro cell models. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102487. [PMID: 40104114 PMCID: PMC11919288 DOI: 10.1016/j.omtn.2025.102487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/12/2025] [Indexed: 03/20/2025]
Abstract
Antisense oligonucleotides (ASOs) have emerged as a new therapeutic modality for the treatment of both rare and common human diseases. A significant proportion of the patient population that may benefit from ASO therapy may also have common diseases, such as diabetes mellitus. The potential influence of prevalent diseases on the effectiveness of ASO drugs in silencing their target mRNAs remains largely unexplored. The present study utilized in vitro cell models to determine the impact on the efficacy of target reduction of two US Food and Drug Administration (FDA)-approved ASO drugs by intracellular glucose levels. Using inotersen and mipomersen as the FDA-approved ASO model drugs, this study demonstrated that a higher intracellular level of glucose resulted in decreased silencing efficacy of target reduction of inotersen and mipomersen in HepG2 cells. Reducing intracellular glucose levels in HepG2 cells, either by knocking down the glucose transporter GLUT2 or by treating with the antidiabetic drug metformin, reversed the decreased silencing efficacy of inotersen and mipomersen. This study brings to light the first indication about the significant impact of intracellular glucose levels on the silencing efficacy of the FDA-approved ASO drugs in an in vitro model.
Collapse
Affiliation(s)
- Le Tra Giang Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Sherouk M Tawfik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Jing Jin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Andrea Durwin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
6
|
Zafar MH, Li C, Lu Z, Lu Y, Zhang Z, Qi R, Nazir U, Yang K, Wang M. Duodenum and Caecum Microbial Shift Modulates Immune and Antioxidant Response Through Energy Homeostasis in Hu Sheep Fed Vegetable Waste and Rice Straw Silage. Antioxidants (Basel) 2024; 13:1546. [PMID: 39765874 PMCID: PMC11727283 DOI: 10.3390/antiox13121546] [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: 11/12/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
The gradual decline in feed resources for livestock needs alternate ways to ensure non-stop feed supply throughout the year. The objective of this study was to evaluate the impact of vegetable waste and rice straw silage (VTRS) on immune response, antioxidant status, and microbial changes in duodenum and caecum in Hu sheep. Eight healthy male Hu sheep were randomly distributed into control (fed farm roughage) and VTRS (fed vegetable waste silage) groups for 35 days. Results had shown that silage had less mycotoxin content (p < 0.05). The VTRS increased butyrate content in duodenal digesta, while acetate, butyrate, total volatile fatty acids (TVFA), and valerate were enhanced in caecal digesta (p < 0.05). The VTRS also increased amylase activity in duodenum and ileum tissues, along with GLUT2 and SGLT1 expressions. In serum, Interleukin-10 (IL-10) concentration and total antioxidant capacity (T-AOC) were increased while malondialdehyde (MDA) was decreased. An increase in T-AOC and GSH-Px activity was also observed, along with increased IL-6, immunoglobulin A (IgA), and catalase in duodenum tissue (p < 0.05). Prevotella was increased in the duodenum and caecum, with Prevotellacae UCG-001 and Christensenellacae R-7 group representing the VTRS group in the duodenum (p < 0.05). KEGG pathway prediction also indicated the enrichment of energy metabolism-related pathways. Significant microbes had shown a significant correlation with immune parameters. It can be concluded that vegetable waste silage has the ability to improve antioxidant status, enhance energy metabolism, and balance intestinal microbiota in Hu sheep.
Collapse
Affiliation(s)
- Muhammad Hammad Zafar
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
| | - Chuang Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China
| | - Zhiqi Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
| | - Yue Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
| | - Zhenbin Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
| | - Ruxin Qi
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
| | - Usman Nazir
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
| | - Kailun Yang
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China
| | - Mengzhi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (M.H.Z.)
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi 832000, China
| |
Collapse
|
7
|
Ji H, Zhu X, Qiu J, Zhang S, Li J, Liu L, Li X, Muneeb M. Milk fat globule membranes ameliorate diet-induced obesity in mice by modulating glucolipid metabolism, body inflammation, and oxidative stress. Food Funct 2024; 15:11903-11917. [PMID: 39584542 DOI: 10.1039/d4fo04072d] [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: 11/26/2024]
Abstract
This study aimed to explore the lipid-lowering effect and the mechanism of action of the milk fat globule membrane (MFGM) in obese mice. All findings indicated that MFGM supplementation impeded weight gain in mice with obesity. qPCR and western blot analysis further revealed that MFGM could reduce lipid deposition and improve lipid metabolism by downregulating the expression levels of Fas, Scd1, PPARγ, and Srebp-1c and increasing the expression levels of Mcad, Cpt-1c, and PPAR-α. MFGM also reduced glucose metabolism disorders by downregulating the expression levels of Pepck and G6pase and upregulating the expression levels of PK and GK. MFGM can reduce the expression levels of TNF-α, IL-6, and IL-1β, thus reducing inflammation in the body. In addition, MFGM also increased the expression of the Nrf2 gene, strengthening the antioxidant enzymes' (GSH, CAT, and SOD) vitality, which strengthened the body's defenses against oxidative stress. In summary, our experiment demonstrated that the MFGM has the potential to treat obesity by controlling the metabolism of fat and glucose, thereby reducing oxidative stress and inflammation, which provides a theoretical foundation for the development of products related to the treatment of obesity.
Collapse
Affiliation(s)
- Haowen Ji
- Food College, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China
| | - Xiaojun Zhu
- Food College, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China
| | - Jiaxin Qiu
- Food College, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China
| | - Shouwen Zhang
- Postdoctoral Research Station of Heilongjiang Yaolan Dairy Technology Stock Company Ltd, 150010, Harbin, China
| | - Jiajun Li
- Heilongjiang Yaolan Dairy Technology Stock Company Ltd, 150010, Harbin, China
| | - Lu Liu
- Food College, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China
| | - Xiaodong Li
- Food College, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China
| | - Muhammad Muneeb
- Food College, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, No. 600 Changjiang St., Xiangfang Dist., 150030, Harbin, China
| |
Collapse
|
8
|
Cherubini A, Della Torre S, Pelusi S, Valenti L. Sexual dimorphism of metabolic dysfunction-associated steatotic liver disease. Trends Mol Med 2024; 30:1126-1136. [PMID: 38890029 DOI: 10.1016/j.molmed.2024.05.013] [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: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver condition. MASLD is a sexually dimorphic condition, with its development and progression influenced by sex chromosomes and hormones. Estrogens typically protect against, whereas androgens promote, MASLD. Therapeutic approaches for a sex-specific personalized medicine include estrogen replacement, androgen blockers, and novel drugs targeting hormonal pathways. However, the interactions between hormonal factors and inherited genetic variation impacts MASLD risk, necessitating more tailored therapies. Understanding sex disparities and the role of estrogens could improve MASLD interventions and management, whereas clinical trials addressing sex differences are crucial for advancing personalized treatment. This review explores the underappreciated impact of sexual dimorphism in MASLD and discusses the potential therapeutic application of sex-related hormones.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Serena Pelusi
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Department of Transfusion Medicine, Precision Medicine Lab, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
9
|
Kwan SH, Esteves F, Davis E, Gonzalez de Mejia E. Chemical characterization and DPP IV inhibitory capacity of purified adzuki bean β-vignin digest in comparison to soybean β-conglycinin and in vitro effect of β-vignin on diabetic-related outcomes. Food Chem 2024; 467:142285. [PMID: 39644661 DOI: 10.1016/j.foodchem.2024.142285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/10/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Adzuki bean (AB) is a legume with a low glycemic index and is traditionally used in Asian cultures to modulate type 2 diabetes (T2D). Our objectives were to characterize the functional peptides from purified AB β-vignin after simulated gastrointestinal digestion in comparison to soybean β-conglycinin, to evaluate their DPP IV inhibitory capacity, and to determine in vitro the effect of digested AB β-vignin on diabetic-related outcomes using HepG2 cells in healthy and insulin-resistant states. Five peptides (215-742 Da) from AB β-vignin and five peptides (215-447 Da) from β-conglycinin were identified to exhibit bioactivity as DPP IV inhibitors. Molecular docking demonstrated peptides could bind to DPP IV at the same binding site as a diabetic medication, linagliptin. Digested AB β-vignin significantly increased (p < 0.05) hepatic glucose uptake (> 290 %) via DPP IV inhibition (> 40 %) in healthy and insulin-resistant states. IRS-1, Akt-1, and Glut 2 increased after treating cells with digested AB β-vignin in healthy and insulin-resistant states.
Collapse
Affiliation(s)
- Shu Hang Kwan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
| | - Frida Esteves
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA; Tecnologico de Monterrey, Monterrey, Mexico
| | - Emily Davis
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
| | - Elvira Gonzalez de Mejia
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA.
| |
Collapse
|
10
|
Liu Y, Jia Y, Wu Y, Zhang H, Ren F, Zhou S. Review on mechanisms of hypoglycemic effects of compounds from highland barley and potential applications. Food Funct 2024; 15:11365-11382. [PMID: 39495067 DOI: 10.1039/d4fo00940a] [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: 11/05/2024]
Abstract
The rising prevalence of metabolic diseases, such as diabetes and obesity, presents a significant global health challenge. Dietary interventions, with their minimal side effects, hold great promise as effective strategies for blood sugar management. Highland barley (HB) boasts a comprehensive and unique nutritional composition, characterized by high protein, high fiber, high vitamins, low fat, low sugar, and diverse bioactive components. These attributes make it a promising candidate for alleviating high blood sugar. This review explores the mechanisms underlying the glucose-lowering properties of HB, emphasizing its nutritional profile and bioactive constituents. Additionally, it examines the impact of common HB processing techniques on its nutrient composition and highlights its applications in food products. By advancing the understanding of HB's value and mechanisms in diabetes prevention, this review aims to facilitate the development of HB-based foods suitable for diabetic patients.
Collapse
Affiliation(s)
- Yanan Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Yuanqiang Jia
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Yingying Wu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Huijuan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Feiyue Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Sumei Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| |
Collapse
|
11
|
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
|
12
|
Ye Y, Cao Z. Glucose Metabolism and Glucose Transporters in Head and Neck Squamous Cell Carcinoma. Cancer Invest 2024; 42:827-844. [PMID: 39324504 DOI: 10.1080/07357907.2024.2407424] [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: 05/11/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Head and neck squamous cell carcinoma ranks seventh globally in malignancy prevalence, with persistent high mortality rates despite treatment advancements. Glucose, pivotal in cancer metabolism via the Warburg effect, enters cells via glucose transporters, notably GLUT proteins. Glycolysis, aerobic oxidation, and the pentose phosphate pathway in glucose metabolism significantly impact HNSCC progression. HNSCC exhibits elevated expression of glucose metabolism enzymes and GLUT proteins, correlating with prognosis. Heterogeneity in HNSCC yields varied metabolic profiles, influenced by factors like HPV status and disease stage. This review highlights glucose metabolism's role and potential as therapeutic targets and cancer imaging tracers in HNSCC.
Collapse
Affiliation(s)
- Yanyan Ye
- Department of Otolaryngology, Shulan (Hangzhou) Hospital, affiliated to Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Zaizai Cao
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Ballesteros-Pla C, Sevillano J, Sánchez-Alonso MG, Limones M, Pita J, Zapatería B, Sanz-Cuadrado MI, Pizarro-Delgado J, Izquierdo-Lahuerta A, Medina-Gómez G, Herradón G, Ramos-Álvarez MDP. Constitutive Pleiotrophin Deletion Results in a Phenotype with an Altered Pancreatic Morphology and Function in Old Mice. Int J Mol Sci 2024; 25:10960. [PMID: 39456743 PMCID: PMC11507919 DOI: 10.3390/ijms252010960] [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: 09/16/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Pleiotrophin (PTN) is crucial for embryonic development and pancreas organogenesis as it regulates metainflammation, metabolic homeostasis, thermogenesis, and glucose tolerance. Pleiotrophin deletion is associated with a lipodystrophic phenotype in which adipose tissue plasticity is altered in late life. This study explored the impact of pleiotrophin deletion on pancreatic morphology and function in later life. We analyzed glucose tolerance and circulating parameters on female wild-type (Ptn+/+) and knock-out (Ptn-/-) mice. At 9 and 15 months, we conducted morphometric analyses of pancreatic islets and evaluated the levels of insulin, glucagon, somatostatin, glucose transporter 2 (GLUT2), vesicle-associated membrane protein 2 (VAMP2), and synaptosome-associated protein 25 (SNAP25) via immunofluorescence. The effect of PTN on glucose-stimulated insulin secretion (GSIS) was evaluated in INS1E cells and isolated islets. Ptn-/- mice showed hyperinsulinemia, impaired glucose tolerance, and increased homeostatic model assessment for insulin resistance (HOMA-IR) with age. While Ptn+/+ islets enlarge with age, in Ptn-/- mice, the median size decreased, and insulin content increased. Vesicle transport and exocytosis proteins were significantly increased in 9-month-old Ptn-/- islets. Islets from Ptn-/- mice showed impaired GSIS and decreased cell membrane localization of GLUT2 whereas, PTN increased GSIS in INS1E cells. Ptn deletion accelerated age-related changes in the endocrine pancreas, affecting islet number and size, and altering VAMP2 and SNAP25 levels and GLUT2 localization leading to impaired GSIS and insulin accumulation in islets.
Collapse
Affiliation(s)
- Cristina Ballesteros-Pla
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Julio Sevillano
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - María Gracia Sánchez-Alonso
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - María Limones
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Jimena Pita
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Begoña Zapatería
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marta Inmaculada Sanz-Cuadrado
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Javier Pizarro-Delgado
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| | - Adriana Izquierdo-Lahuerta
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, 28922 Madrid, Spain; (A.I.-L.); (G.M.-G.)
| | - Gema Medina-Gómez
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, 28922 Madrid, Spain; (A.I.-L.); (G.M.-G.)
| | - Gonzalo Herradón
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte Urbanización Montepríncipe, 28660 Madrid, Spain;
| | - María del Pilar Ramos-Álvarez
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660 Madrid, Spain; (C.B.-P.); (J.S.); (M.G.S.-A.); (M.L.); (J.P.); (B.Z.); (M.I.S.-C.); (J.P.-D.)
| |
Collapse
|
14
|
Milfort MC, Ghareeb AFA, Ariyo OW, Kwakye J, Hartono E, Sovi S, Aryal B, Fuller AL, El Sabry MI, Stino F, Rekaya R, Aggrey SE. Renal Sugar Metabolites and mRNA Expression of Glucose Transporters in Meat-Type Chickens with Differing Residual Water Intake. Animals (Basel) 2024; 14:2912. [PMID: 39409861 PMCID: PMC11482541 DOI: 10.3390/ani14192912] [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: 09/04/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Molecular differences exist between birds with high residual water intake (HRWI) compared to those with low residual water intake (LRWI). Residual water intake (RWI) is defined as the difference between the water intake of a bird and the expected water intake corrected for metabolic body weight, feed intake, and body weight gain. Tissue metabolomic analysis revealed significantly increased kidney glucose, fructose, and arabitol in the LRWI group compared to the HRWI group. mRNA expression analysis of apical sodium glucose cotransporters SGLT1, SGLT4, SGLT5, and SGLT6 showed decreased expression of SGLTs 1, 5, and 6 in LRWI birds (p < 0.05), whereas SGLT4 expression was increased compared with HRWI birds (p < 0.01). An analysis of basal glucose transporters GLUT1, GLUT2, GLUT5, and GLUT9 showed significantly increased GLUT2 expression in LRWI birds compared with HRWI birds (p < 0.01). We postulate that SGLT4 is the main apical transporter in chicken kidneys and that its increased expression reduces these birds' need for water, resulting in less drinking. This is balanced by the increased expression of the basal transporter GLUT2, indicating better glucose retention, which may partly explain the physiological mechanism behind why these birds drink less water. Innately driven broiler water intake could therefore be influenced by the expression of kidney solute transporters.
Collapse
Affiliation(s)
- Marie C. Milfort
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Ahmed F. A. Ghareeb
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Oluwatomide W. Ariyo
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Josephine Kwakye
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Evan Hartono
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Selorm Sovi
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Bikash Aryal
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Alberta L. Fuller
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| | - Mohamed I. El Sabry
- Department of Animal Production, Cairo University, Giza 12613, Egypt; (M.I.E.S.); (F.S.)
| | - Farid Stino
- Department of Animal Production, Cairo University, Giza 12613, Egypt; (M.I.E.S.); (F.S.)
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA;
| | - Samuel E. Aggrey
- Water Intake Genomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (M.C.M.); (A.F.A.G.); (O.W.A.); (J.K.); (E.H.); (S.S.); (B.A.); (A.L.F.)
| |
Collapse
|
15
|
U N, R C T, R KR, Mahalingam G. Glucose transporters and their energy homeostasis function in various organs. VITAMINS AND HORMONES 2024; 128:1-47. [PMID: 40097247 DOI: 10.1016/bs.vh.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Glucose transporters (GLUTs) belong to a membrane-protein family that essentially participates in easing the transportation and absorption of glucose molecules throughout the cellular membranes. From the brain to the eyes, each section delves into the intricate mechanisms of glucose uptake and utilization, shedding light on the unique adaptations and regulatory pathways in different anatomical structures. Beginning with the brain, known for its high energy demands, the chapter explicates the specialized GLUT expression patterns crucial for neuronal function and synaptic transmission. Moving to metabolic powerhouses like the liver, muscles, and adipose tissue, it elucidates the dynamic interplay of GLUT isoforms in energy storage, mobilization, and insulin responsiveness. Furthermore, the chapter navigates through the kidneys, lungs, skin, and reproductive organs, unveiling the diverse roles of GLUTs in renal glucose reabsorption, pulmonary-epithelial transportation, skin barrier associated functions, and gonadal development. It also explores the significance of placental GLUTs in fatal nutrient supply and the implications of cardiac GLUTs in myocardial energy metabolism. Additionally, it examines the intricate regulation of GLUTs at key barriers like the BBB (Blood-Brain Barrier) and placenta, as well as in endocrine glands such as the pancreas, adrenal medulla and thyroid. Moreover, it further elucidates the less-explored territories of GLUT expression in the bones, gastrointestinal tract, and ocular tissues like the retina, unraveling their implications in immune function, bone metabolism, intestinal glucose-sensing, and retinal physiology.
Collapse
Affiliation(s)
- Nithya U
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Theijeswini R C
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Karthick Raja R
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Gayathri Mahalingam
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
16
|
Wei Y, Li X, Cui R, Liu J, Wang G. Associations between sensitivity to thyroid hormones and insulin resistance in euthyroid adults with obesity. Front Endocrinol (Lausanne) 2024; 15:1366830. [PMID: 39175570 PMCID: PMC11338882 DOI: 10.3389/fendo.2024.1366830] [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: 02/06/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
Background Impaired sensitivity to thyroid hormones (TH) was associated with metabolic syndrome. The study aimed to explore the association between central TH sensitivity indices and insulin resistance (IR) in euthyroid adults with obesity. Methods This cross-sectional study enrolled 293 euthyroid outpatients with obesity in Beijing Chao-Yang Hospital. We used the thyroid feedback quantile-based index (TFQI), thyroid stimulating hormone index (TSHI), and thyrotrophic T4 resistance index (TT4RI) to indicate central TH sensitivity. IR was assessed by homeostasis model assessment of insulin resistance (HOMA-IR), hepatic insulin resistance index (hepatic-IR), the Matsuda index, and the adipose tissue insulin resistance index (Adipo-IR). Participants were categorized according to tertiles of TH sensitivity indices. We used multiple linear regressions to explore the associations. Results There was a significant stepwise increase in HOMA-IR and Adipo-IR from the lowest to the highest tertiles of TH sensitivity indices (all P<0.05). After adjustment for age, sex, body mass index, hypertension, hyperlipidemia, and diabetes, only Adipo-IR was significantly associated with TH sensitivity indices. On average, each unit increased in TFQI, TSHI, and TT4RI was associated with 1.19 (P=0.053), 1.16 (P=0.04), and 1.01 (P=0.03) units increased in Adipo-IR, respectively. There was no significant association between TH sensitivity indices and HOMA-IR, hepatic-IR, and the Matsuda index after adjustment for other risk factors. Conclusions Reduced central TH sensitivity was associated with increased adipose tissue insulin resistance in euthyroid adults with obesity. The results further confirmed the importance of TH sensitivity on metabolic diseases.
Collapse
Affiliation(s)
| | | | | | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Nawaz AH, Setthaya P, Feng C. Exploring Evolutionary Adaptations and Genomic Advancements to Improve Heat Tolerance in Chickens. Animals (Basel) 2024; 14:2215. [PMID: 39123741 PMCID: PMC11311085 DOI: 10.3390/ani14152215] [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: 05/31/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Climate change poses a significant threat to the poultry industry, especially in hot climates that adversely affect chicken growth, development, and productivity through heat stress. This literature review evaluates the evolutionary background of chickens with the specific genetic characteristics that can help chickens to cope with hot conditions. Both natural selection and human interventions have influenced the genetic characteristics of the breeds used in the current poultry production system. The domestication of chickens from the Red junglefowl (Gallus gallus) has resulted in the development of various breeds with distinct genetic differences. Over the past few years, deliberate breeding for desirable traits (such as meat production and egg quality) in chickens has resulted in the emergence of various economically valuable breeds. However, this selective breeding has also caused a decrease in the genetic diversity of chickens, making them more susceptible to environmental stressors like heat stress. Consequently, the chicken breeds currently in use may possess a limited ability to adapt to challenging conditions, such as extreme heat. This review focuses on evaluating potential genes and pathways responsible for heat tolerance, including heat shock response, antioxidant defense systems, immune function, and cellular homeostasis. This article will also discuss the physiological and behavioral responses of chicken varieties that exhibit genetic resistance to heat, such as the naked neck and dwarf traits in different indigenous chickens. This article intends to review the current genomic findings related to heat tolerance in chickens that used methods such as the genome-wide association study (GWAS) and quantitative trait loci (QTL) mapping, offering valuable insights for the sustainability of poultry in the face of global warming.
Collapse
Affiliation(s)
- Ali Hassan Nawaz
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
| | - Phatthawin Setthaya
- Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Chungang Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
| |
Collapse
|
18
|
Zhao Y, Veysman B, Antolijao K, Zhao Y, Papagni Y, Wang H, Ross R, Tibbot T, Povrzenic D, Fox R. Increase in the Expression of Glucose Transporter 2 (GLUT2) on the Peripheral Blood Insulin-Producing Cells (PB-IPC) in Type 1 Diabetic Patients after Receiving Stem Cell Educator Therapy. Int J Mol Sci 2024; 25:8337. [PMID: 39125908 PMCID: PMC11313087 DOI: 10.3390/ijms25158337] [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: 06/14/2024] [Revised: 07/28/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Multicenter international clinical trials demonstrated the clinical safety and efficacy by using stem cell educator therapy to treat type 1 diabetes (T1D) and other autoimmune diseases. Previous studies characterized the peripheral blood insulin-producing cells (PB-IPC) from healthy donors with high potential to give rise to insulin-producing cells. PB-IPC displayed the molecular marker glucose transporter 2 (GLUT2), contributing to the glucose transport and sensing. To improve the clinical efficacy of stem cell educator therapy in the restoration of islet β-cell function, we explored the GLUT2 expression on PB-IPC in recent onset and longstanding T1D patients. In the Food and Drug Administration (FDA)-approved phase 2 clinical studies, patients received one treatment with the stem cell educator therapy. Peripheral blood mononuclear cells (PBMC) were isolated for flow cytometry analysis of PB-IPC and other immune markers before and after the treatment with stem cell educator therapy. Flow cytometry revealed that both recent onset and longstanding T1D patients displayed very low levels of GLUT2 on PB-IPC. After the treatment with stem cell educator therapy, the percentages of GLUT2+CD45RO+ PB-IPC were markedly increased in these T1D subjects. Notably, we found that T1D patients shared common clinical features with patients with other autoimmune and inflammation-associated diseases, such as displaying low or no expression of GLUT2 on PB-IPC at baseline and exhibiting a high profile of the inflammatory cytokine interleukin (IL)-1β. Flow cytometry demonstrated that their GLUT2 expressions on PB-IPC were also markedly upregulated, and the levels of IL-1β-positive cells were significantly downregulated after the treatment with stem cell educator therapy. Stem cell educator therapy could upregulate the GLUT2 expression on PB-IPC and restore their function in T1D patients, leading to the improvement of clinical outcomes. The clinical data advances current understanding about the molecular mechanisms underlying the stem cell educator therapy, which can be expanded to treat patients with other autoimmune and inflammation-associated diseases.
Collapse
Affiliation(s)
- Yong Zhao
- Throne Biotechnologies, Paramus, NJ 07652, USA
| | | | | | - Yelu Zhao
- Throne Biotechnologies, Paramus, NJ 07652, USA
| | | | | | - Robin Ross
- Throne Biotechnologies, Paramus, NJ 07652, USA
| | - Terri Tibbot
- Life Line Stem Cell Tissue, Cord Blood Bank, New Haven, IN 46774, USA
| | | | - Richard Fox
- Throne Biotechnologies, Paramus, NJ 07652, USA
| |
Collapse
|
19
|
Wang Z, Liu H. Roles of Lysine Methylation in Glucose and Lipid Metabolism: Functions, Regulatory Mechanisms, and Therapeutic Implications. Biomolecules 2024; 14:862. [PMID: 39062577 PMCID: PMC11274642 DOI: 10.3390/biom14070862] [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: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Glucose and lipid metabolism are essential energy sources for the body. Dysregulation in these metabolic pathways is a significant risk factor for numerous acute and chronic diseases, including type 2 diabetes (T2DM), Alzheimer's disease (AD), obesity, and cancer. Post-translational modifications (PTMs), which regulate protein structure, localization, function, and activity, play a crucial role in managing cellular glucose and lipid metabolism. Among these PTMs, lysine methylation stands out as a key dynamic modification vital for the epigenetic regulation of gene transcription. Emerging evidence indicates that lysine methylation significantly impacts glucose and lipid metabolism by modifying key enzymes and proteins. This review summarizes the current understanding of lysine methylation's role and regulatory mechanisms in glucose and lipid metabolism. We highlight the involvement of methyltransferases (KMTs) and demethylases (KDMs) in generating abnormal methylation signals affecting these metabolic pathways. Additionally, we discuss the chemical biology and pharmacology of KMT and KDM inhibitors and targeted protein degraders, emphasizing their clinical implications for diseases such as diabetes, obesity, neurodegenerative disorders, and cancers. This review suggests that targeting lysine methylation in glucose and lipid metabolism could be an ideal therapeutic strategy for treating these diseases.
Collapse
Affiliation(s)
| | - Huadong Liu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China;
| |
Collapse
|
20
|
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
|
21
|
Tran M, Gilling S, Wu J, Wang L, Shin DJ. miR-141/200c contributes to ethanol-mediated hepatic glycogen metabolism. Mol Metab 2024; 84:101942. [PMID: 38642890 PMCID: PMC11060962 DOI: 10.1016/j.molmet.2024.101942] [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: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024] Open
Abstract
OBJECTIVE Hepatic glucose metabolism is profoundly perturbed by excessive alcohol intake. miR-141/200c expression is significantly induced by chronic ethanol feeding. This study aimed at identifying the role of miR-141/200c in glucose homeostasis during chronic ethanol exposure. METHODS WT and miR-141/200c KO mice were fed a control or an ethanol diet for 30 days, followed by a single binge of maltose dextrin or ethanol, respectively. Untargeted metabolomics analysis of hepatic primary metabolites was performed along with analyses for liver histology, gene expression, intracellular signaling pathways, and physiological relevance. Primary hepatocytes were used for mechanistic studies. RESULTS miR-141/200c deficiency rewires hepatic glucose metabolism during chronic ethanol feeding, increasing the abundance of glucose intermediates including G6P, an allosteric activator for GS. miR-141/200c deficiency replenished glycogen depletion during chronic ethanol feeding accompanied by reduced GS phosphorylation in parallel with increased expression of PP1 glycogen targeting subunits. Moreover, miR-141/200c deficiency prevented ethanol-mediated increases in AMPK and CaMKK2 activity. Ethanol treatment reduced glycogen content in WT-hepatocytes, which was reversed by dorsomorphin, a selective AMPK inhibitor, while KO-hepatocytes displayed higher glycogen content than WT-hepatocytes in response to ethanol treatment. Furthermore, treatment of hepatocytes with A23187, a calcium ionophore activating CaMKK2, lowered glycogen content in WT-hepatocytes. Notably, the suppressive effect of A23187 on glycogen deposition was reversed by dorsomorphin, demonstrating that the glycogen depletion by A23187 is mediated by AMPK. KO-hepatocytes exhibited higher glycogen content than WT-hepatocytes in response to A23187. Finally, miR-141/200c deficiency led to improved glucose tolerance and insulin sensitivity during chronic ethanol feeding. CONCLUSIONS miR-141/200c deficiency replenishes ethanol-mediated hepatic glycogen depletion through the regulation of GS activity and calcium signaling coupled with the AMPK pathway, improving glucose homeostasis and insulin sensitivity. These results underscore miR-141/200c as a potential therapeutic target for the management of alcohol intoxication.
Collapse
Affiliation(s)
- Melanie Tran
- Department of Physiology and Neurobiology, University of Connecticut, 75 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Shaynian Gilling
- Department of Physiology and Neurobiology, University of Connecticut, 75 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Jianguo Wu
- Department of Physiology and Neurobiology, University of Connecticut, 75 N. Eagleville Rd, Storrs, CT 06269, USA
| | - Li Wang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, 333 Cedar St, New Haven, CT 06510, USA
| | - Dong-Ju Shin
- Department of Physiology and Neurobiology, University of Connecticut, 75 N. Eagleville Rd, Storrs, CT 06269, USA.
| |
Collapse
|
22
|
Soleimanifar F, Aghapur N, Rezaei-Kiasari Z, Mahboudi H, Kaabi M, Mansour RN, Kehtari M, Abazari M, Enderami SE, Hassannia H. The generation of islet-like insulin-producing cells from Wharton's jelly-derived mesenchymal stem cells on the PES/fish gelatin scaffold. Regen Ther 2024; 26:251-259. [PMID: 38974324 PMCID: PMC11225687 DOI: 10.1016/j.reth.2024.05.019] [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: 03/21/2024] [Revised: 05/12/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024] Open
Abstract
Diabetes Mellitus (DM) disrupts the body's capability to control blood glucose statuses. Type 1 diabetes mellitus (T1DM) arises from inadequate insulin production and is treated with insulin replacement therapy. Stem cell therapy is a hopeful treatment for T1DM that involves using adult stem cells to generate insulin-producing cells (IPCs). Mesenchymal stem cells (MSCs) are particularly advantageous for generating IPCs. The islet cells require interactions with the extracellular matrix for survival, which is lacking in conventional 2D culture systems. Natural or synthetic polymers create a supportive 3D microenvironment in tissue engineering. We aim to construct superior differentiation conditions employing polyethersulfone (PES)/Fish gelatin scaffolds to differentiate Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) to IPCs. In this study, the PES/fish gelatin scaffold (3D) was manufactured by electrospinning, and then its biocompatibility and non-toxicity were investigated by MTT assay. After that, scaffold-supportive effects on WJ-MSCs differentiation to IPCs were studied at the gene and protein levels. After exposure to the differentiation media, 2D and 3D (PES/Fish gelatin) cultured cells were slowly aggregated and developed spherical-shaped clusters. The viability of cells was found to be comparable in both 2D and 3D cultures. The gene expression analysis showed that efficiency of differentiation was more elevated in 3D culture. Additionally, ELISA results indicated that C-peptide and insulin release were more significant in 3D than in 2D culture. In conclusion, the PES/fish gelatin scaffold is highly promising for pancreatic tissue engineering because it supports the viability, growth, and differentiation of WJ-MSCs into IPCs.
Collapse
Affiliation(s)
- Fatemeh Soleimanifar
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Zeinab Rezaei-Kiasari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hosein Mahboudi
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Reyhaneh Nassiri Mansour
- Department of Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mousa Kehtari
- Department of Animal Biology, School of Biology, Faculty of Science, University of Tehran, Tehran, Iran
| | - Mohammadfoad Abazari
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hadi Hassannia
- Amol Faculty of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
23
|
Yoshino S, Ishida E, Horiguchi K, Matsumoto S, Nakajima Y, Ozawa A, Yamada M, Yamada E. Mixed-Lineage Leukaemia Gene Regulates Glucose-Sensitive Gene Expression and Insulin Secretion in Pancreatic Beta Cells. Int J Mol Sci 2024; 25:4704. [PMID: 38731926 PMCID: PMC11082990 DOI: 10.3390/ijms25094704] [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: 03/29/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
The escalating prevalence of diabetes mellitus underscores the need for a comprehensive understanding of pancreatic beta cell function. Interest in glucose effectiveness has prompted the exploration of novel regulatory factors. The myeloid/lymphoid or mixed-lineage leukaemia gene (MLL) is widely recognised for its role in leukemogenesis and nuclear regulatory mechanisms through its histone methyltransferase activity in active chromatin. However, its function within pancreatic endocrine tissues remains elusive. Herein, we unveil a novel role of MLL in glucose metabolism and insulin secretion. MLL knockdown in βHC-9 pancreatic beta cells diminished insulin secretion in response to glucose loading, paralleled by the downregulation of the glucose-sensitive genes SLC2a1 and SLC2a2. Similar observations were made in MLL heterozygous knockout mice (MLL+/-), which exhibited impaired glucose tolerance and reduced insulin secretion without morphological anomalies in pancreatic endocrine cells. The reduction in insulin secretion was independent of changes in beta cell mass or insulin granule morphology, suggesting the regulatory role of MLL in glucose-sensitive gene expression. The current results suggest that MLL interacts with circadian-related complexes to modulate the expression of glucose transporter genes, thereby regulating glucose sensing and insulin secretion. Our findings shed light on insulin secretion control, providing potential avenues for therapeutics against diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Eijiro Yamada
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (S.Y.); (K.H.); (S.M.); (Y.N.); (A.O.); (M.Y.)
| |
Collapse
|
24
|
Zhang Q, Hu S, Jin Z, Wang S, Zhang B, Zhao L. Mechanism of traditional Chinese medicine in elderly diabetes mellitus and a systematic review of its clinical application. Front Pharmacol 2024; 15:1339148. [PMID: 38510656 PMCID: PMC10953506 DOI: 10.3389/fphar.2024.1339148] [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: 11/15/2023] [Accepted: 01/31/2024] [Indexed: 03/22/2024] Open
Abstract
Objective: Affected by aging, the elderly diabetes patients have many pathological characteristics different from the young people, including more complications, vascular aging, cognitive impairment, osteoporosis, and sarcopenia. This article will explore their pathogenesis and the mechanism of Traditional Chinese medicine (TCM) intervention, and use the method of systematic review to evaluate the clinical application of TCM in elderly diabetes. Method: Searching for randomized controlled trials (RCTs) published from January 2000 to November 2023 in the following databases: Web of Science, Pubmed, Embase, Cochrane Library, Sinomed, China National Knowledge Internet, Wanfang and VIP. They were evaluated by three subgroups of Traditional Chinese Prescription, Traditional Chinese patent medicines and Traditional Chinese medicine extracts for their common prescriptions, drugs, adverse reactions and the quality of them. Results and Conclusion: TCM has the advantages of multi-target and synergistic treatment in the treatment of elderly diabetes. However, current clinical researches have shortcomings including the inclusion of age criteria and diagnosis of subjects are unclear, imprecise research design, non-standard intervention measures, and its safety needs further exploration. In the future, the diagnosis of elderly people with diabetes needs to be further clarified. Traditional Chinese patent medicines included in the pharmacopoeia can be used to conduct more rigorous RCTs, and then gradually standardize the traditional Chinese medicine prescriptions and traditional Chinese medicine extracts, providing higher level evidence for the treatment of elderly diabetes with traditional Chinese medicine.
Collapse
Affiliation(s)
- Qiqi Zhang
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Shiwan Hu
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zishan Jin
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Sicheng Wang
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Boxun Zhang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Sahu P, Camarillo IG, Sundararajan R. Efficacy of metformin and electrical pulses in breast cancer MDA-MB-231 cells. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:54-73. [PMID: 38464382 PMCID: PMC10918234 DOI: 10.37349/etat.2024.00204] [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: 08/19/2023] [Accepted: 10/30/2023] [Indexed: 03/12/2024] Open
Abstract
Aim Triple-negative breast cancer (TNBC) is a very aggressive subset of breast cancer, with limited treatment options, due to the lack of three commonly targeted receptors, which merits the need for novel treatments for TNBC. Towards this need, the use of metformin (Met), the most widely used type-2 diabetes drug worldwide, was explored as a repurposed anticancer agent. Cancer being a metabolic disease, the modulation of two crucial metabolites, glucose, and reactive oxygen species (ROS), is studied in MDA-MB-231 TNBC cells, using Met in the presence of electrical pulses (EP) to enhance the drug efficacy. Methods MDA-MB-231, human TNBC cells were treated with Met in the presence of EP, with various concentrations Met of 1 mmol/L, 2.5 mmol/L, 5 mmol/L, and 10 mmol/L. EP of 500 V/cm, 800 V/cm, and 1,000 V/cm (with a pulse width of 100 µs at 1 s intervals) were applied to TNBC and the impact of these two treatments was studied. Various assays, including cell viability, microscopic inspection, glucose, ROS, and wound healing assay, were performed to characterize the response of the cells to the combination treatment. Results Combining 1,000 V/cm with 5 mmol/L Met yielded cell viability as low as 42.6% at 24 h. The glucose level was reduced by 5.60-fold and the ROS levels were increased by 9.56-fold compared to the control, leading to apoptotic cell death. Conclusions The results indicate the enhanced anticancer effect of Met in the presence of electric pulses. The cell growth is inhibited by suppressing glucose levels and elevated ROS. This shows a synergistic interplay between electroporation, Met, glucose, and ROS metabolic alterations. The results show promises for combinational therapy in TNBC patients.
Collapse
Affiliation(s)
- Praveen Sahu
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Ignacio G. Camarillo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
26
|
Shen Z, Hou Y, Zhao G, Tan L, Chen J, Dong Z, Ni C, Pei L. Physiological functions of glucose transporter-2: From cell physiology to links with diabetes mellitus. Heliyon 2024; 10:e25459. [PMID: 38333863 PMCID: PMC10850595 DOI: 10.1016/j.heliyon.2024.e25459] [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: 07/24/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Glucose is a sugar crucial for human health since it participates in many biochemical reactions. It produces adenosine 5'-triphosphate (ATP) and nucleosides through glucose metabolic and pentose phosphate pathways. These processes require many transporter proteins to assist in transferring glucose across cells, and the most notable ones are glucose transporter-2 (GLUT-2) and sodium/glucose cotransporter 1 (SGLT1). Glucose enters small intestinal epithelial cells from the intestinal lumen by crossing the brush boundary membrane via the SGLT1 cotransporter. It exits the cells by traversing the basolateral membrane through the activity of the GLUT-2 transporter, supplying energy throughout the body. Dysregulation of these glucose transporters is involved in the pathogenesis of several metabolic diseases, such as diabetes. Natural loss of GLUT-2 or its downregulation causes abnormal blood glucose concentrations in the body, such as fasting hypoglycemia and glucose tolerance. Therefore, understanding GLUT-2 physiology is necessary for exploring the mechanisms of diabetes and targeted treatment development. This article reviews how the apical GLUT-2 transporter maintains normal physiological functions of the human body and the adaptive changes this transporter produces under pathological conditions such as diabetes.
Collapse
Affiliation(s)
- Zhean Shen
- Xinjiang Institute of Technology, Aksu, China
| | - Yingze Hou
- Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Guo Zhao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Libi Tan
- School of Laboratory Medicine and Biotechnology, Southern Medical University, China
| | - Jili Chen
- Department of Nutrition and Food Hygiene School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziqi Dong
- School of Public Health, Peking University Health Science Center, Beijing 100021, China
| | - Chunxiao Ni
- Hangzhou Lin ‘an District Center for Disease Control and Prevention, Hangzhou, China
| | | |
Collapse
|
27
|
Mohsin S, Elabadlah H, Alotaiba MK, AlAmry S, Almehairbi SJ, Harara MMK, Almuhsin AMH, Tariq S, Howarth FC, Adeghate EA. High-Density Lipoprotein Is Located Alongside Insulin in the Islets of Langerhans of Normal and Rodent Models of Diabetes. Nutrients 2024; 16:313. [PMID: 38276551 PMCID: PMC10818677 DOI: 10.3390/nu16020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Recent studies have implicated pre-beta and beta lipoproteins (VLDL and LDL) in the etiopathogenesis of complications of diabetes mellitus (DM). In contrast, alpha lipoprotein (HDL) is protective of the beta cells of the pancreas. This study examined the distribution of HDL in the islets of Langerhans of murine models of type 1 diabetic rats (streptozotocin (STZ)-induced DM in Wistar rats) and type 2 models of DM rats (Goto-Kakizaki (GK), non-diabetic Zucker lean (ZL), and Zucker diabetic and fatty (ZDF)). The extent by which HDL co-localizes with insulin or glucagon in the islets of the pancreas was also investigated. Pancreatic tissues of Wistar non-diabetic, diabetic Wistar, GK, ZL, and ZDF rats were processed for immunohistochemistry. Pancreatic samples of GK rats fed with either a low-fat or a high-fat diet were prepared for transmission immune-electron microscopy (TIEM) to establish the cytoplasmic localization of HDL in islet cells. HDL was detected in the core and periphery of pancreatic islets of Wistar non-diabetic and diabetic, GK, ZL, and ZDF rats. The average total of islet cells immune positive for HDL was markedly (<0.05) reduced in GK and ZDF rats in comparison to Wistar controls. The number of islet cells containing HDL was also remarkably (p < 0.05) reduced in Wistar diabetic rats and GK models fed on high-fat food. The co-localization study using immunofluorescence and TIEM techniques showed that HDL is detected alongside insulin within the secretory granules of β-cells. HDL did not co-localize with glucagon. This observation implies that HDL may contribute to the metabolism of insulin.
Collapse
Affiliation(s)
- Sahar Mohsin
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
| | - Haba Elabadlah
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
- Cambridge Medical and Rehabilitation Center, Al Ain P.O. Box 222297, United Arab Emirates
| | - Mariam K. Alotaiba
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
| | - Suhail AlAmry
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
| | - Shamma J. Almehairbi
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
| | - Maha M. K. Harara
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
| | - Aisha M. H. Almuhsin
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
| | - Saeed Tariq
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
| | - Frank Christopher Howarth
- Department of Physiology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Ernest A. Adeghate
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.M.)
- Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|