1
|
Suryaningtyas IT, Jung WK, Lee SJ, Je JY. Bioactive peptides PIISVYWK and FSVVPSPK improve glucose homeostasis by targeting DPP-IV and glucose transport in type 2 diabetic mice. Int Immunopharmacol 2025; 158:114844. [PMID: 40359889 DOI: 10.1016/j.intimp.2025.114844] [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: 03/07/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
Type 2 Diabetes Mellitus (T2DM) is a metabolic disorder characterized by chronic hyperglycemia, insulin resistance, and progressive β-cell dysfunction, often exacerbated by inflammation and oxidative stress. Effective management requires multi-targeted approaches, including modulation of glucose metabolism, suppression of inflammatory pathways, and pancreatic protection. This study investigates the antidiabetic and immunomodulatory potential of PIISVYWK (P1) and FSVVPSPK (P2), bioactive peptides from blue mussel, Mytilus edulis, in regulating these pathways. In vitro, P1 and P2 were assessed for their ability to inhibit α-glucosidase and DPP-IV activity in Caco-2 cells, alongside glucose uptake and transporter protein expression (SGLT-1 and GLUT2). In vivo, HFD/STZ-induced diabetic mice were administered P1 or P2 (1 mg/kg or 10 mg/kg) or metformin (200 mg/kg) for four weeks. Peptide treatment significantly improved glycemic control by inhibiting α-glucosidase and DPP-IV, increasing GLP-1 levels, and reducing intestinal glucose uptake. Additionally, P1 and P2 exhibited strong anti-inflammatory effects by suppressing NF-κB activation and reducing circulating IL-6, TNF-α, and IL-1β levels. Enhanced antioxidant enzyme activity (SOD, GPx, CAT) further mitigated oxidative stress, preventing pancreatic damage. Peptides also preserved β-cell function by enhancing insulin secretion and regulating glucagon levels. These findings suggest that P1 and P2 peptides exert antidiabetic effects through multi-targeted mechanisms, including immunomodulation, making them promising therapeutic candidates for T2DM management.
Collapse
Affiliation(s)
- Indyaswan T Suryaningtyas
- Research Center for Marine-Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Food Technology and Processing, National Research and Innovation Agency, Yogyakarta 55861, Indonesia
| | - Won-Kyo Jung
- Research Center for Marine-Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Sei-Jung Lee
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
2
|
Wang N, Zhu X, Xu Z, Ning X, Guo L, Liang D, Li G, Zhu N. Photoaged polystyrene nanoplastics induce perturbation of glucose metabolism in HepG2 cells via oxidative stress. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 379:126534. [PMID: 40425061 DOI: 10.1016/j.envpol.2025.126534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/29/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
MICRO: & nano-plastics (MNPs) have been considered an emerging persistent pollutant in the environment. Most of the works focus on the potential toxicity of pristine, rather than photoaged, MNPs, let alone the underlying mechanisms of toxicity. To address this gap, we exposed human liver cancer cells (HepG2) to polystyrene nanoplastics (PS-NPs) with varying degrees of photodegradation, including pristine PS-NPs and photoaged PS-NPs irradiated with UV for 8 days (short-term) and 32 days (long-term).The surface characteristics of PS-NPs exhibited a significant alteration as characterized by SEM, FTIR, XPS, and Zetasizer. Exposure to PS-NPs affected cell viability, ion transport capacity and glucose metabolism, and also induced oxidative stress. Photoaged PS-NPs posed relatively higher impacts than pristine ones on HepG2 cells. Long-term photoaged PS-NPs induced the glucose metabolic disorders in a dose-dependent manner, while pristine and short-term photoaged PS-NPs induced the metabolic disorders only at high concentrations. The severe cellular metabolic toxicity of PS-NPs was attributed to the changes in physicochemical properties induced by UV irradiation, such as the production of oxygen-containing functional groups (hydroxyl, carboxyl, and carbonyl groups). Taken together, the long-term photoaged PS-NPs suppressed more than 10% of cell vitality compared to the pristine ones, and disrupted the glucose metabolism in HepG2 cells, particularly gene expression associated with glucose homeostasis.
Collapse
Affiliation(s)
- Ning Wang
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, 030006, P. R. CHINA
| | - Xin Zhu
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, 030006, P. R. CHINA
| | - Zhiqiang Xu
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, 030006, P. R. CHINA
| | - Xia Ning
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, 030006, P. R. CHINA
| | - Lin Guo
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, 030006, P. R. CHINA
| | - Dong Liang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan, 030051, P. R. CHINA
| | - Guangke Li
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, 030006, P. R. CHINA
| | - Na Zhu
- Shanxi Key Laboratory of Coal-based Emerging Pollutant Identification and Risk Control, Research Center of Environment and Health, College of Environment and Resource, Shanxi University, Taiyuan, 030006, P. R. CHINA.
| |
Collapse
|
3
|
Abruscato G, Tarantino R, Mauro M, Chiarelli R, Vizzini A, Arizza V, Vazzana M, Luparello C. Glucose consumption and uptake in HepG2 cells is improved by aqueous extracts from leaves, but not rhizomes, of Posidonia oceanica (L.) Delile via GLUT-4 upregulation. PROTOPLASMA 2025:10.1007/s00709-025-02076-8. [PMID: 40413340 DOI: 10.1007/s00709-025-02076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
The endemic Mediterranean seagrass Posidonia oceanica is a valuable source of natural bioactive compounds that possess significant therapeutic potential. Here, we examined whether aqueous extracts of rhizomes (RE) and green leaves (GLE) of P. oceanica could exert a glucose-lowering effect on the HepG2 cell line, chosen as an in vitro model of liver cells. We assessed glucose uptake and storage, expression levels of GLUT-2 and -4 transporters and the exposure of the latter one at cell surface, as well as modulation of the expression, synthesis and/or activation of the GLUT2-transcription factor hepatocyte nuclear factor-1 alpha (HNF1α), and insulin receptor substrate-1 (IRS-1), AKT and protein kinase Cζ (PKCζ), which regulate GLUT-4 translocation. Glucose consumption/uptake and glycogen storage were increased with exposure to GLE alone. Furthermore, at the molecular level GLE-induced upregulation of (i) IRS-1, AKT, and PKCζ activation levels, (ii) GLUT-4 translation levels, and (iii) GLUT-4 exposure on the cell surface. Conversely, GLUT-2 protein was downregulated. Therefore, the application of the aqueous extract of green leaves of P. oceanica may be suitable for the development of new treatment agents or dietary supplements for diabetes mellitus acting through GLUT-4 mediated glucose import.
Collapse
Affiliation(s)
- Giulia Abruscato
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Roberta Tarantino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Manuela Mauro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Roberto Chiarelli
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
- NBFC, National Biodiversity Future Center, 90133, Palermo, Italy
| | - Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
- NBFC, National Biodiversity Future Center, 90133, Palermo, Italy
| | - Mirella Vazzana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
- NBFC, National Biodiversity Future Center, 90133, Palermo, Italy
| | - Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy.
- NBFC, National Biodiversity Future Center, 90133, Palermo, Italy.
| |
Collapse
|
4
|
Maharwal N, Shrivastava R, Majumder SK. Insight into Optogenetics for Diabetes Management. ACS Synth Biol 2025; 14:1324-1335. [PMID: 40279455 DOI: 10.1021/acssynbio.4c00549] [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: 04/27/2025]
Abstract
Optogenetics is an interdisciplinary field wherein optical and genetic engineering methods are employed together to impart photounresponsive cells (usually of higher animals) the ability to respond to light through expression of light-sensitive proteins sourced generally from algae or bacteria. It enables precise spatiotemporal control of various cellular activities through light stimulation. Recently, emerging as a synthetic biology-based approach for diabetes management, optogenetics can provide user-control of hormonal secretion by photoactivation of a suitably modified cell. For around a decade, studies have been performed on the applicability of various light-sensitive proteins and their incorporation into pancreatic and nonpancreatic cells for photoinduced insulin secretion. Further, in vivo studies demonstrated amelioration of diabetes in mouse models through photoactivation of the implanted engineered cells. Here, we attempt to highlight the various optogenetic approaches explored in terms of influencing the insulin secretion pathway at different points in light of the natural insulin secretion pathway in pancreatic β cells. We also discuss how transgenic cells of both pancreatic as well as nonpancreatic origin are exploited for photoinduced secretion of insulin. Recent advances on integration of "smart" technologies for remote control of light irradiation and thereby insulin secretion from implanted engineered cells in preclinical models are also described. Additionally, the need for further comprehensive studies on irradiation parameters, red-shifted opsins, and host-cell interaction is stressed to realize the full potential of optogenetics as a clinically applicable modality providing user-controlled "on demand" hormonal secretion for better management of diabetes.
Collapse
Affiliation(s)
- Nidhi Maharwal
- Laser Biomedical Applications Division, Laser R&D Block-A1, Raja Ramanna Centre for Advanced Technology, Indore 452013, India
- Homi Bhabha National Institute, 2nd floor, BARC Training School Complex, Anushakti Nagar, Mumbai 400 094, India
| | - Rashmi Shrivastava
- Laser Biomedical Applications Division, Laser R&D Block-A1, Raja Ramanna Centre for Advanced Technology, Indore 452013, India
- Homi Bhabha National Institute, 2nd floor, BARC Training School Complex, Anushakti Nagar, Mumbai 400 094, India
| | - Shovan Kumar Majumder
- Laser Biomedical Applications Division, Laser R&D Block-A1, Raja Ramanna Centre for Advanced Technology, Indore 452013, India
- Homi Bhabha National Institute, 2nd floor, BARC Training School Complex, Anushakti Nagar, Mumbai 400 094, India
| |
Collapse
|
5
|
Musso N, Bonacci PG, Letizia Consoli GM, Maugeri L, Terrana M, Lanzanò L, Longo E, Buscarino G, Consoli A, Petralia S. Biofriendly glucose-derived carbon nanodots: GLUT2-mediated cell internalization for an efficient targeted drug delivery and light-triggered cancer cell damage. J Colloid Interface Sci 2025; 696:137873. [PMID: 40403481 DOI: 10.1016/j.jcis.2025.137873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/09/2025] [Accepted: 05/11/2025] [Indexed: 05/24/2025]
Abstract
Personalized medicine holds great promise for treating the underlying causes of many human diseases with high precision. Low-dimensional carbon-based materials are being designed to more closely match specific delivery efficiency for targeted cancer treatment, while enabling the benefits of increased biocompatibility, high cargo-loading capacity and excellent light-responsive properties, including photoluminescence and photothermal effects. Here, we report an unprecedented example of glucose-based carbon-nanodots (CDs-gluc) obtained via a one-pot thermal process from glucose, without using organic solvent and additional reagents. The CDs-gluc nanostructures, composed of a C-sp2 inner core and a glucose outer shell, showed a high photothermal conversion efficiency (η = 42.7 % at 532 nm), good photoluminescence quantum yield (ϕPL = 6 %), and low cytotoxicity. Measurements of cellular Zeta-potential demonstrated the effective interaction of CDs-gluc with the surface of cancer cells overexpressing the Glucose Transporter Type 2 (GLUT2). The effective and specific GLUT2-mediated internalization mechanism was demonstrated by inducing up- and down-regulation of the transporter expression under conditions of glucose excess and deprivation, through fluorescence correlation spectroscopy. The potential of the CDs-gluc as drug nanocarriers was demonstrated by entrapping the anticancer drug 5-fluorouracil, achieving a drug loading capacity of 4.5 ± 0.8 %. In vitro experiments confirmed the excellent light-triggered cell damage activity and remarkable cell-targeting ability of CDs-gluc driven by GLUT2 expression. The easy and green preparation, biocompatibility, effective and specific cellular uptake, photoluminescence and hyperthermia make CDs-gluc appealing candidates in the research of novel nanostructures for cancer cell targeting.
Collapse
Affiliation(s)
- Nicolo Musso
- Faculty of Medicine and Surgery, "Kore" University of Enna, Contrada Santa Panasia, 94100 Enna, Italy; Department of Biomedicals and Biotechnologies Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy.
| | - Paolo Giuseppe Bonacci
- Department of Biomedicals and Biotechnologies Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | | | - Ludovica Maugeri
- Department of Drug and Health Sciences, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Morena Terrana
- Department of Biomedicals and Biotechnologies Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | - Luca Lanzanò
- Department of Physic and Astronomy, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Elisa Longo
- Department of Physic and Astronomy, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Gianpiero Buscarino
- Department of Physics and Chemistry, University of Palermo, Via Archirafi 36, 90123 Palermo, Italy
| | - Antonino Consoli
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Salvatore Petralia
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; Department of Drug and Health Sciences, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy.
| |
Collapse
|
6
|
Rusanuar ND, Aminuddin A, Hamid AA, Kumar J, Hui CK, Mahadi MK, Ugusman A. The Potential of Edible Bird's Nests in Reducing Cardiovascular Disease Risk Factors: A Narrative Review. Int J Mol Sci 2025; 26:4619. [PMID: 40429763 PMCID: PMC12111720 DOI: 10.3390/ijms26104619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiovascular disease (CVD) remains a leading cause of mortality worldwide, with dyslipidemia, obesity, diabetes mellitus, and hypertension being major modifiable risk factors. Functional foods with antioxidant and anti-inflammatory properties have gained attention for their potential for reducing CVD risk. Edible bird's nest (EBN), a functional food rich in bioactive compounds such as sialic acid, lactoferrin, and glycoproteins, has been shown to exhibit antioxidant and anti-inflammatory effects. This review explores the potential of EBN in mitigating CVD risk factors, focusing on its role in improving lipid profiles, managing obesity, and enhancing glucose metabolism. EBN has been shown to improve the lipid profile by regulating the hepatic cholesterol metabolism and gut-liver axis interactions. Additionally, EBN reduces body weight gain and visceral fat accumulation, improves adipokine regulation, and enhances insulin sensitivity, which may collectively support cardiovascular health. Despite promising findings, clinical evidence remains limited. Future research should focus on clinical trials to validate its efficacy, determine optimal dosages, and assess its long-term safety. Additionally, further studies on EBN's effects on hypertension and its interaction with conventional therapies could enhance its potential role in CVD prevention and management.
Collapse
Affiliation(s)
- Nina Diyana Rusanuar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia; (N.D.R.); (A.A.); (A.A.H.); (J.K.)
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia; (N.D.R.); (A.A.); (A.A.H.); (J.K.)
- Cardiovascular and Pulmonary (CardioResp) Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Adila A. Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia; (N.D.R.); (A.A.); (A.A.H.); (J.K.)
- Cardiovascular and Pulmonary (CardioResp) Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia; (N.D.R.); (A.A.); (A.A.H.); (J.K.)
| | | | - Mohd Kaisan Mahadi
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia; (N.D.R.); (A.A.); (A.A.H.); (J.K.)
- Cardiovascular and Pulmonary (CardioResp) Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
7
|
Singh P, Singh DP, Patel MK, Binwal M, Kaushik A, Mall M, Sahu M, Khare P, Shanker K, Bawankule DU, Sundaresan V, Mani DN, Shukla AK. Vindoline is a key component of Catharanthus roseus leaf juice extract prepared through an Ayurveda-based method for ameliorating insulin-resistant type 2 diabetes. PROTOPLASMA 2025; 262:667-681. [PMID: 39794517 DOI: 10.1007/s00709-024-02026-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025]
Abstract
Catharanthus roseus leaves have been traditionally described to possess potent antidiabetic activity and some leaf-specific alkaloids, including vindoline, have been studied for their antidiabetic potential. The aim of the present study was to validate the antidiabetic property of the plant with special reference to vindoline. An Ayurveda-based method was used to prepare the Swaras [leaf juice extract (LJE)] of three familial C. roseus genotypes differing in their vindoline content [CIM-Sushil (CS) > Dhawal (D) > Nirmal (N)]. In vivo experiments using LJE were performed in Charles Foster rats, whereby metformin (M100, 100 mg/kg BW) and vindoline (V20, 20 mg/kg BW) were used for comparison. OGTT-based screening for LJE doses (N100, N300, N500, D100, D200, D300, CS100, CS200, CS300 mg/kg BW) was carried out. Further analysis of the effective doses (D100, D200, D300, CS100, CS200, CS300) in streptozotocin-induced diabetic rats indicated highest blood glucose depletion in D300 (52.51%) and CS200 (64.55%) together with V20 (56.96%) on the 14th day. CS-LJE was found to be safe up to 2000 mg/kg BW. The role of LJE/vindoline in maintaining glucose homeostasis in liver was found to be mediated through the expression of insulin pathway genes (IRS-1, PI3K, AKT, GLUT2). TNF-α-induced insulin resistance in L6 skeletal muscle cells was used to analyze the effect of LJE/vindoline through glucose uptake assay and expression analysis of insulin pathway genes (IRS-1, PI3K, AKT, GLUT4). The results indicated that the antidiabetic effect of LJE/vindoline is mediated through activation of IRS/PI3K/AKT/GLUT signaling pathway.
Collapse
Affiliation(s)
- Pooja Singh
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
| | - Dewasya P Singh
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Manish K Patel
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
| | - Monika Binwal
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Amit Kaushik
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Maneesha Mall
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
| | - Mridula Sahu
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
| | - Puja Khare
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Karuna Shanker
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Dnyaneshwar U Bawankule
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Velusamy Sundaresan
- CSIR-Central Institute of Medicinal and Aromatic Plants, Research Centre, Allalasandra, GKVK Post, Bengaluru, 560065, Karnataka, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Daya N Mani
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| | - Ashutosh K Shukla
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
8
|
Toyoda Y, Shigesawa R, Merriman TR, Matsuo H, Takada T. GLUT2/SLC2A2 is a bi-directional urate transporter. J Biol Chem 2025; 301:108485. [PMID: 40209957 DOI: 10.1016/j.jbc.2025.108485] [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: 01/30/2025] [Accepted: 03/31/2025] [Indexed: 04/12/2025] Open
Abstract
Recent genetic studies showed an association between solute carrier 2A2 (SLC2A2), which encodes glucose transporter 2 (GLUT2), and serum urate concentrations; however, urate transport activity of GLUT2 has not been studied contrary to its function as a sugar transporter. Here, we hypothesized that GLUT2 acts also as a urate transporter, which led us to conduct cell-based functional analyses using HEK-derived 293A cells. We found that radiolabeled [8-14C]-urate was incorporated into GLUT2-expressing cells more compared to control cells and this elevated cellular activity was almost completely inhibited by GLUT2 inhibitors, demonstrating that GLUT2 is a urate transporter. Regarding the concentration dependence of GLUT2-mediated urate transport, no saturable properties were observed within an experimentally achievable range (0-500 μM), suggesting that GLUT2 mediates the robust transport of urate. Moreover, the GLUT2-mediated urate transport was not inhibited by 10 mM glucose; GLUT2-mediated sugar transport was hardly affected by 500 μM urate. As these concentrations of urate and glucose were relevant to their maximum levels in healthy humans, our results suggest that GLUT2 maintains its urate transport ability under physiological conditions. Furthermore, using a cell-based urate efflux assay system, we successfully demonstrated that urate secretion was accelerated in GLUT2-expressing cells than in control cells. Therefore, GLUT2 may also function as a urate exporter. The present study revealed that GLUT2 is a bi-directional urate transporter. Our findings contribute to a deeper understanding of urate-handling systems in the body. To elucidate the physiological role of GLUT2 as a urate transporter, further studies are required.
Collapse
Affiliation(s)
- Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan; Department of Pharmacy, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Ryuichiro Shigesawa
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan.
| |
Collapse
|
9
|
Kim SA, Kim HG, Wijesinghe WCB, Min D, Yoon TY. Emerging Patterns in Membrane Protein Folding Pathways. Annu Rev Biophys 2025; 54:141-162. [PMID: 40327440 DOI: 10.1146/annurev-biophys-070524-100658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Studies of membrane protein folding have progressed from simple systems such as bacteriorhodopsin to complex structures such as ATP-binding cassette transporters and voltage-gated ion channels. Advances in techniques such as single-molecule force spectroscopy and in vivo force profiling now allow for the detailed examination of membrane protein folding pathways at amino acid resolutions. These proteins navigate rugged energy landscapes partly shaped by the absence of hydrophobic collapse and the viscous nature of the lipid bilayer, imposing biophysical limitations on folding speeds. Furthermore, many transmembrane (TM) helices display reduced hydrophobicity to support functional requirements, simultaneously increasing the energy barriers for membrane insertion, a manifestation of the evolutionary trade-off between functionality and foldability. These less hydrophobic TM helices typically insert and fold as helical hairpins, following the protein synthesis direction from the N terminus to the C terminus, with assistance from endoplasmic reticulum (ER) chaperones like the Sec61 translocon and the ER membrane protein complex. The folding pathways of multidomain membrane proteins are defined by allosteric networks that extend across various domains, where mutations and folding correctors affect seemingly distant domains. A common evolutionary strategy is likely to be domain specialization, where N-terminal domains enhance foldability and C-terminal domains enhance functionality. Thus, despite inherent biophysical constraints, evolution has finely tuned membrane protein sequences to optimize foldability, stability, and functionality.
Collapse
Affiliation(s)
- Sang Ah Kim
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea;
| | - Hyun Gyu Kim
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea;
| | - W C Bhashini Wijesinghe
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, South Korea;
| | - Duyoung Min
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, South Korea;
| | - Tae-Young Yoon
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea;
| |
Collapse
|
10
|
Ida-Yonemochi H. Role of glucose metabolism in amelogenesis. J Oral Biosci 2025; 67:100667. [PMID: 40306383 DOI: 10.1016/j.job.2025.100667] [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: 11/28/2024] [Revised: 04/27/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Cell energy metabolism plays a pivotal role in organ development and function by regulating cell behavior in pathophysiological conditions. Glucose metabolism is the central cascade for obtaining energy in mammalian cells, and cells alter the glucose metabolic pathway depending on intra- and extracellular environments. Therefore, glucose metabolism is closely associated with cell differentiation stages, and cell energy metabolism plays a vital role not only in energy production but also in cell fate regulation in organogenesis. HIGHLIGHT During enamel formation, the timing of the expression of passive and active glucose transporters, glycogen synthesis, and glycogen degradation is strictly regulated according to the energy demand of ameloblast-lineage cells. These glucose metabolic reactions are particularly active in the maturation stage of ameloblasts. Furthermore, autophagy, a key regulator of cellular energy homeostasis that modulates glucose metabolism, occurs during both the secretory and maturation stages of ameloblasts. Disruption of glucose metabolism cascade and autophagy induces enamel hypoplasia, as demonstrated in both in vitro and in vivo models. CONCLUSION Adequate energy supply via glucose metabolism is essential for enamel matrix secretion and maturation. A thorough understanding of the precise regulation of energy metabolism in amelogenesis facilitates comprehension of the normal enamel formation process and pathological conditions affecting it. This review summarizes glucose metabolic processes during amelogenesis, focusing on glucose uptake, glycogenesis, and glycogenolysis.
Collapse
Affiliation(s)
- Hiroko Ida-Yonemochi
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan.
| |
Collapse
|
11
|
Ma GY, Hou LK, Guo JH, Sun JC, Nong Q, Wang YF, Hu SW, Zhao WJ, Tan J, Liu XF, Guo Y, Zhang BJ, Wang HL, Wang SY, Li XQ, Cao W. The structures of two polysaccharides from Fructus Corni and their protective effect on insulin resistance. Carbohydr Polym 2025; 353:123290. [PMID: 39914965 DOI: 10.1016/j.carbpol.2025.123290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 05/07/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by impaired cellular or organ responsiveness to insulin, which results in ineffective glucose disposal and lacks a definitive treatment. Previous studies suggest that polysaccharides hold promise as natural agents for the prevention and treatment of T2DM. In this study, we identified two polysaccharides from Fructus Corni, PFC-CI and PFC-CII, with molecular weights of 59.0 kDa and 26.0 kDa, respectively. PFC-CI consists of T-α-Galp-(1→6)-α-Galp-(1→6)-α-Galp-(1→[4)-GalpA-(1→4)-GalpA-OMe-(1→4)-α-GalpA-3-OAc-(1→]m→[2)-Rhap-(1→4)-α-GalpA-(1→]n, with side chains composed of α-Araf-(1→, →3)-α-Araf-(1→, →3,5)-α-Araf-(1→, →5)-α-Araf-(1→. PFC-CII is a linear α-1,6-glucan. Among them, PFC-CI showed superior effects on promoting glucose consumption and uptake in insulin-resistant (IR) HepG2 cells. In vivo, PFC-CI significantly alleviate IR in high-fat diet and streptozotocin-induced diabetic rats, indicating its potential as a therapeutic agent for T2DM. Mechanistically, PFC-CI modulates the glucose transporter 2 (Glut2)/glucokinase signaling pathway, thereby exerting anti-T2DM effects. This research indicates that Fructus Corni contains a potential therapeutic polysaccharide as a specific regulator of Glut2 for treating T2DM.
Collapse
Affiliation(s)
- Guang-Yuan Ma
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Lu-Kuan Hou
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Jia-Hui Guo
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Jing-Chun Sun
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Qiuna Nong
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Yu-Fan Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Sheng-Wei Hu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Wen-Jing Zhao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Jin Tan
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Xiao-Feng Liu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Yuan Guo
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Bo-Jing Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Hong-Ling Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Shu-Yao Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Xiao-Qiang Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Wei Cao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
12
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [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] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
13
|
Attrill EH, Scharapow O, Perera S, Mayne S, Sumargo N, Ross RM, Richards SM, Sutherland BA, Premilovac D. Controlled induction of type 2 diabetes in mice using high fat diet and osmotic-mini pump infused streptozotocin. Sci Rep 2025; 15:8812. [PMID: 40087321 PMCID: PMC11909212 DOI: 10.1038/s41598-025-89162-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/03/2025] [Indexed: 03/17/2025] Open
Abstract
Type 2 diabetes (T2D) is a progressive metabolic disorder characterised by obesity, insulin resistance, impaired glucose tolerance, and hyperglycaemia. The long time-course of T2D in humans makes accurate modelling of sustained T2D in animal models difficult. The goal of this study was to develop and characterise an accurate and reproducible, non-transgenic model of sustained T2D in mice. Adult, male C57BL/6 mice were placed on a high-fat diet (HFD) for 17 weeks. From weeks 3-5, osmotic mini-pumps were implanted subcutaneously to slowly infuse streptozotocin (STZ; 200-350 mg/kg) for 14-days after which mini-pumps were removed. Body weight, blood glucose concentration, and glucose tolerance were monitored for 12 weeks post STZ treatment. Our data demonstrate that the combination of HFD and 200 mg/kg STZ delivered by mini-pump leads to increased blood glucose concentrations and impaired glucose tolerance, while maintaining obesity and hepatic dyslipidaemia. In week 17, plasma insulin concentration was assessed and showed that with STZ treatment, mice still produce insulin, but that this is reduced compared with mice on HFD only. Lastly, we examined pancreas sections using immunohistochemistry and show that there is no overt loss of beta cell mass. In conclusion, we demonstrate development of a reproducible in vivo model of T2D in mice that replicates a number of key pathophysiological changes seen in humans with T2D.
Collapse
Affiliation(s)
- Emily H Attrill
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Oscar Scharapow
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Sathya Perera
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Sophie Mayne
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Nicole Sumargo
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Renee M Ross
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Stephen M Richards
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Dino Premilovac
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000, Australia.
| |
Collapse
|
14
|
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
|
15
|
Yang J, Li L, An Z, Lv Y, Li R, Li J, Guo M, Sun H, Yang H, Wang L, Liu Y, Guo H. Role of hepatocyte-specific FOXO1 in hepatic glucolipid metabolic disorders induced by perfluorooctane sulfonate. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125632. [PMID: 39755352 DOI: 10.1016/j.envpol.2025.125632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/25/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Perfluorooctane sulfonate (PFOS), a prevalent perfluoroalkyl substance (PFAS), is widely present in various environmental media, animals, and even human bodies. It primarily accumulates in the liver, contributing to the disruption of hepatic metabolic homeostasis. However, the precise mechanism underlying PFOS-induced hepatic glucolipid metabolic disorders remains elusive. The transcription factor forkhead box protein O 1 (FOXO1) plays a crucial role in regulating hepatic glucolipid metabolism; however, its involvement in PFOS-induced hepatic glucolipid metabolic disorders has not been thoroughly explored. Molecular docking revealed high binding affinity between PFOS and FOXO1. Male C57BL/6 mice were exposed to PFOS at doses of 0.3, 1.0, and 3.0 mg/kg body weight for 12 weeks to assess its subchronic effects on hepatic glucolipid metabolism in this work. The results indicate that PFOS exposure increases hepatic acetylated FOXO1 expression, promotes liver lipid accumulation, suppresses gluconeogenesis, whereas fasting blood glucose levels remain unaffected but this dysregulation results in insulin resistance. Furthermore, hepatic deletion of FOXO1 in PFOS-exposed mice ameliorates liver injury and reduces lipid accumulation by suppressing hepatic autophagy without significantly affecting gluconeogenesis. In conclusion, FOXO1 may play a pivotal role in the development of PFOS-induced hepatic glucolipid metabolic disorder.
Collapse
Affiliation(s)
- Jing Yang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China; Ningxia Hui Autonomous Region Center for Disease Control and Prevention, Yinchuan, 750001, PR China
| | - Longfei Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Ziwen An
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yi Lv
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Ran Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Jing Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mingmei Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Heming Sun
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Huiling Yang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Lei Wang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yi Liu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Huicai Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Province, Shijiazhuang, 050017, PR China.
| |
Collapse
|
16
|
Farhadi R, Daniali M, Baeeri M, Khorasani R, Haghi-Aminjan H, Gholami M, Rahimifard M, Navaei-Nigjeh M, Abdollahi M. Molecular evidence of the inhibitory potential of melatonin against sodium arsenite toxicity. Heliyon 2025; 11:e42113. [PMID: 39916822 PMCID: PMC11799970 DOI: 10.1016/j.heliyon.2025.e42113] [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: 07/10/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Introduction Sodium arsenite (SA), NaAsO2, is among the most hazardous toxicants, and wide use and presence of this toxicant leads to a severe environmental threat. Exposure to SA is associated with many health concerns, such as the prevalence of cancer and diabetes mellitus type 2 (DMT2). Many studies suggest that SA induces inflammation and biochemical impairments through different mechanisms, including increasing oxidative stress and altering vital genes such as biochemical and anti-inflammatory. Recent studies on melatonin (MLT), a harmless hormone secreted in the body generally for induction of sleepiness, find many beneficial and positive effects. Mitigating different harms and toxicities through different mechanisms, such as antioxidant properties, anti-inflammatory effects, and critical gene regulation, is essential. Due to these findings, this study aimed to evaluate the hypothesis that MLT may ameliorate pancreatic damage caused by exposure to SA. Methods Forty-eight adult healthy male wistar rats aged 7-8 weeks were divided into eight for this research. Group 1 did not receive any intervention. Group 2 received 10 mg/kg/day MLT through intraperitoneal (IP) injection. Groups 3, 4, and 5 received 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg SA, respectively. Groups 6, 7, and 8 were given 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg of SA along with 10 mg/kg/day MLT, respectively, during the last ten days of the experiment. After 28 days of the experiment, the blood and tissue samples of the pancreas were removed for biochemical and pathological examination. Results MLT attenuates SA toxicity by reducing oxidative stress biomarkers and inflammation markers. Moreover, MLT improves SA exposure's biochemical and functional damages by regulating related genes and pathways. Conclusion MLT poses protective and preventive effects on the pancreas against exposure to SA. However, MLT's therapeutic and beneficial impacts have great potential for further investigation.
Collapse
Affiliation(s)
- Ramtin Farhadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Marzieh Daniali
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Reza Khorasani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Haghi-Aminjan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahban Rahimifard
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mona Navaei-Nigjeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
17
|
Li M, Ding L, Cao L, Zhang Z, Li X, Li Z, Xia Q, Yin K, Song S, Wang Z, Du H, Zhao D, Li X, Wang Z. Natural products targeting AMPK signaling pathway therapy, diabetes mellitus and its complications. Front Pharmacol 2025; 16:1534634. [PMID: 39963239 PMCID: PMC11830733 DOI: 10.3389/fphar.2025.1534634] [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: 11/26/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Diabetes mellitus (DM) ranks among the most prevalent chronic metabolic diseases, characterized primarily by a persistent elevation in blood glucose levels. This condition typically stems from either insufficient insulin secretion or a functional defect in the insulin itself. Clinically, diabetes is primarily classified into type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM), with T2DM comprising nearly 90% of all diagnosed cases. Notably, the global incidence of T2DM has surged dramatically over recent decades. The adenylate-activated protein kinase (AMPK) signaling pathway is crucial in regulating cellular energy metabolism, marking it as a significant therapeutic target for diabetes and related complications. Natural products, characterized by their diverse origins, multifaceted bioactivities, and relative safety, hold considerable promise in modulating the AMPK pathway. This review article explores the advances in research on natural products that target the AMPK signaling pathway, aiming to inform the development of innovative antidiabetic therapies.
Collapse
Affiliation(s)
- Min Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Lu Ding
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Liyuan Cao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Xueyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zirui Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Qinjing Xia
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Kai Yin
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Siyu Song
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zihan Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Haijian Du
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zeyu Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
18
|
Zhou J, Shi Y, Zhao L, Wang R, Luo L, Yin Z. γ-Glutamylcysteine restores glucolipotoxicity-induced islet β-cell apoptosis and dysfunction via inhibiting endoplasmic reticulum stress. Toxicol Appl Pharmacol 2025; 495:117206. [PMID: 39701215 DOI: 10.1016/j.taap.2024.117206] [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: 09/25/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE The impaired function of islet β-cell is associated with the pathogenesis of type 2 diabetes mellitus (T2DM). γ-glutamylcysteine (γ-GC), an immediate precursor of glutathione (GSH), has antioxidant and neuroprotective functions. Its level has been reported to be down-regulated in hyperglycemia. However, whether γ-GC has a protective effect on islet β-cell dysfunction remains elusive. Recently, we explore the molecular mechanism by which γ-GC protects islet β-cell from glucolipotoxicity-induced dysfunction. METHODS In vivo mice models and in vitro cell models were established to examine the therapeutic effects and molecular mechanisms of γ-GC. RESULTS db mice develop impaired glucose-stimulated insulin secretion (GSIS) due to reduced islet number and damaged islet microstructure. Serious oxidative damage, apoptosis and lipid accumulation are also observed in β-cell stimulated by glucolipotoxicity. Mechanistic studies suggest that glucolipotoxicity inhibits PDX-1 nuclear translocation by inducing endoplasmic reticulum (ER) stress, which leads to impaired insulin (INS) secretion in β-cell. Nevertheless, γ-GC as an inhibitor of ER stress can alleviate the damage of islet microstructure in db mice. Importantly, γ-GC promotes INS gene expression and GSIS through driving nuclear translocation of PDX-1, thereby enhancing intracellular INS content. Moreover, treatment with γ-GC can also mitigate oxidative damage, apoptosis and lipid accumulation of β-cell, resulting in ameliorating islet β-cell dysfunction induced by glucolipotoxicity. CONCLUSION Our results support the use of γ-GC as an inhibitor of ER stress for prevention and treatment of T2DM in the future.
Collapse
Affiliation(s)
- Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Yingying Shi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lishuang Zhao
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Rong Wang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
19
|
Okoro FO, Markus V. Artificial sweeteners and Type 2 Diabetes Mellitus: A review of current developments and future research directions. J Diabetes Complications 2025; 39:108954. [PMID: 39854925 DOI: 10.1016/j.jdiacomp.2025.108954] [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: 09/05/2024] [Revised: 11/12/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
While artificial sweeteners are Generally Regarded as Safe (GRAS), the scientific community remains divided on their safety status. The previous assumption that artificial sweeteners are inert within the body is no longer valid. Artificial sweeteners, known for their high intense sweetness and low or zero calories, are extensively used today in food and beverage products as sugar substitutes and are sometimes recommended for weight management and Type 2 Diabetes Mellitus (T2DM) patients. The general omission of information about the concentration of artificial sweeteners on market product labels makes it challenging to determine the amounts of artificial sweeteners consumed by people. Despite regulatory authorization for their usage, such as from the United States Food and Drug Administration (FDA), concerns remain about their potential association with metabolic diseases, such as T2DM, which the artificial sweeteners were supposed to reduce. This review discusses the relationship between artificial sweetener consumption and the risk of developing T2DM. With the increasing number of recent scientific studies adding to the debate on this subject matter, we assessed recent literature and up-to-date evidence. Importantly, we highlight future research directions toward furthering knowledge in this field of study.
Collapse
Affiliation(s)
- Francisca Obianuju Okoro
- Department of Medical Biochemistry, Faculty of Medicine, Near East University, Nicosia, 99138 TRNC, Mersin 10, Turkey
| | - Victor Markus
- Department of Medical Biochemistry, Faculty of Medicine, Near East University, Nicosia, 99138 TRNC, Mersin 10, Turkey.
| |
Collapse
|
20
|
Zhu Y, Verkhratsky A, Chen H, Yi C. Understanding glucose metabolism and insulin action at the blood-brain barrier: Implications for brain health and neurodegenerative diseases. Acta Physiol (Oxf) 2025; 241:e14283. [PMID: 39822067 PMCID: PMC11737474 DOI: 10.1111/apha.14283] [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: 11/11/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
The blood-brain barrier (BBB) is a highly selective, semipermeable barrier critical for maintaining brain homeostasis. The BBB regulates the transport of essential nutrients, hormones, and signaling molecules between the bloodstream and the central nervous system (CNS), while simultaneously protecting the brain from potentially harmful substances and pathogens. This selective permeability ensures that the brain is nourished and shielded from toxins. An exception to this are brain regions, such as the hypothalamus and circumventricular organs, which are irrigated by fenestrated capillaries, allowing rapid and direct response to various blood components. We overview the metabolic functions of the BBB, with an emphasis on the impact of altered glucose metabolism and insulin signaling on BBB in the pathogenesis of neurodegenerative diseases. Notably, endothelial cells constituting the BBB exhibit distinct metabolic characteristics, primarily generating ATP through aerobic glycolysis. This occurs despite their direct exposure to the abundant oxygen in the bloodstream, which typically supports oxidative phosphorylation. The effects of insulin on astrocytes, which form the glial limitans component of the BBB, show a marked sexual dimorphism. BBB nutrient sensing in the hypothalamus, along with insulin signaling, regulates systemic metabolism. Insulin modifies BBB permeability by regulating the expression of tight junction proteins, angiogenesis, and vascular remodeling, as well as modulating blood flow in the brain. The disruptions in glucose and insulin signaling are particularly evident in neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, where BBB breakdown accelerates cognitive decline. This review highlights the critical role of normal glucose metabolism and insulin signaling in maintaining BBB functionality and investigates how disruptions in these pathways contribute to the onset and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiyi Zhu
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Department of NeurosciencesUniversity of the Basque Country, CIBERNEDLeioaBizkaiaSpain
- IKERBASQUE Basque Foundation for ScienceBilbaoSpain
- Department of Forensic Analytical Toxicology, School of Forensic MedicineChina Medical UniversityShenyangChina
| | - Hui Chen
- School of Life Sciences, Faculty of ScienceUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Chenju Yi
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseGuangzhouChina
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational ResearchShenzhenChina
| |
Collapse
|
21
|
Msane S, Khathi A, Sosibo AM. The Effect of the 14:10-Hour Time-Restricted Feeding (TRF) Regimen on Selected Markers of Glucose Homeostasis in Diet-Induced Prediabetic Male Sprague Dawley Rats. Nutrients 2025; 17:292. [PMID: 39861423 PMCID: PMC11768421 DOI: 10.3390/nu17020292] [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: 11/23/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Prediabetes is a condition that often precedes the onset of type 2 diabetes mellitus (T2DM). Literature evidence indicates that prediabetes is reversible, making it an important therapeutic target for preventing the progression to T2DM. Several studies have investigated intermittent fasting as a possible method to manage or treat prediabetes. OBJECTIVES This study evaluated the impact of a 14:10-hour time-restricted feeding (TRF) regimen on leptin concentration, insulin sensitivity and selected markers associated with the insulin signalling pathway and glucose homeostasis in diet-induced prediabetic rats. METHODS Twenty-four male Sprague Dawley rats were obtained and randomly divided into two dietary groups: group 1 (n = 6) received a standard diet and water, while group 2 (n = 18) was provided a high-fat, high-carbohydrate (HFHC) diet supplemented with 15% fructose for a period of 20 weeks to induce prediabetes. After confirming prediabetes, an intermittent fasting (IF) regimen was assigned to the rats while also having untreated and metformin-treated prediabetic rats serving as controls. RESULTS Both IF and HFHC-Met groups yield significantly lower blood glucose, leptin and BMI results compared to the prediabetic group. The IF group yielded significantly lower insulin, HOMA-IR and HbA1C than both controls. CONCLUSIONS The study showed the potential of IF in alleviating prediabetes-induced dysregulation of glucose homeostasis and therefore warrants further investigations into its use in the management of prediabetes.
Collapse
Affiliation(s)
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (S.M.); (A.M.S.)
| | | |
Collapse
|
22
|
Du J, Shen M, Chen J, Yan H, Xu Z, Yang X, Yang B, Luo P, Ding K, Hu Y, He Q. The impact of solute carrier proteins on disrupting substance regulation in metabolic disorders: insights and clinical applications. Front Pharmacol 2025; 15:1510080. [PMID: 39850557 PMCID: PMC11754210 DOI: 10.3389/fphar.2024.1510080] [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: 10/12/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Carbohydrates, lipids, bile acids, various inorganic salt ions and organic acids are the main nutrients or indispensable components of the human body. Dysregulation in the processes of absorption, transport, metabolism, and excretion of these metabolites can lead to the onset of severe metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, gout and hyperbilirubinemia. As the second largest membrane receptor supergroup, several major families in the solute carrier (SLC) supergroup have been found to play key roles in the transport of substances such as carbohydrates, lipids, urate, bile acids, monocarboxylates and zinc ions. Based on common metabolic dysregulation and related metabolic substances, we explored the relationship between several major families of SLC supergroup and metabolic diseases, providing examples of drugs targeting SLC proteins that have been approved or are currently in clinical/preclinical research as well as SLC-related diagnostic techniques that are in clinical use or under investigation. By highlighting these connections, we aim to provide insights that may contribute to the development of improved treatment strategies and targeted therapies for metabolic disorders.
Collapse
Affiliation(s)
- Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minhui Shen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhuai Hu
- Yuhong Pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Qiaojun He
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Liu Y, Wang W, Liang B, Zou Z, Zhang A. NLRP3 inflammasome activation and disruption of IRS-1/PI3K/AKT signaling: Potential mechanisms of arsenic-induced pancreatic beta cells dysfunction in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117504. [PMID: 39657381 DOI: 10.1016/j.ecoenv.2024.117504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Environmental exposure to arsenic is associated with significant health risks, including diabetogenic effects linked to pancreatic dysfunction. The NOD-like receptor protein 3 (NLRP3) inflammasome has been implicated in various metabolic abnormalities; however, its specific role in arsenic-induced pancreatic dysfunction remains insufficiently understood. This study aimed to elucidate the involvement and underlying mechanisms of the NLRP3 inflammasome in arsenic-induced pancreatic beta cells dysfunction through in vivo and in vitro models. In rat models, arsenic exposure was found to activate the NLRP3 inflammasome, as evidenced by pathomorphological changes and the expression of inflammasome activation markers. These pathological changes were accompanied by disruptions in the insulin signaling pathway, characterized by increased phosphorylation of insulin receptor substrate 1 (IRS-1) at Ser616, reduced expression of phosphatidylinositol 3-kinase (PI3K) and phosphorylated protein kinase B (AKT) at Ser473, and significant decreases in downstream targets, including the nuclear translocation of PDX-1, membrane translocation of glucose transporter 2 (GLUT2), and glucokinase (GCK) expression. In vitro, NaAsO2-treated INS-1 cells exhibited a dose-dependent reduction in glucose-stimulated insulin secretion. Furthermore, arsenic exposure in these cells activated the NLRP3 inflammasome, suppressed the IRS-1/PI3K/AKT signaling pathway, and downregulated insulin secretion regulatory molecules (PDX-1, GLUT2, and GCK). Notably, these arsenic-induced effects were reversed by MCC950, an NLRP3 inflammasome inhibitor, and Extendin-4, an agonist of the IRS-1/PI3K/AKT signaling pathway. Collectively, these findings demonstrate that NLRP3 inflammasome activation disrupts the IRS-1/PI3K/AKT signaling pathway, contributing to arsenic-induced pancreatic beta cells dysfunction in rats.
Collapse
Affiliation(s)
- Yonglian Liu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Bing Liang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Zhonglan Zou
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou 550025, PR China.
| |
Collapse
|
24
|
Shafi S, Khan MA, Ahmad J, Rabbani SA, Singh S, Najmi AK. Envisioning Glucose Transporters (GLUTs and SGLTs) as Novel Intervention against Cancer: Drug Discovery Perspective and Targeting Approach. Curr Drug Targets 2025; 26:109-131. [PMID: 39377414 DOI: 10.2174/0113894501335877240926101134] [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: 06/27/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 10/09/2024]
Abstract
Metabolic reprogramming and altered cellular energetics have been recently established as an important cancer hallmark. The modulation of glucose metabolism is one of the important characteristic features of metabolic reprogramming in cancer. It contributes to oncogenic progression by supporting the increased biosynthetic and bio-energetic demands of tumor cells. This oncogenic transformation consequently results in elevated expression of glucose transporters in these cells. Moreover, various cancers exhibit abnormal transporter expression patterns compared to normal tissues. Recent investigations have underlined the significance of glucose transporters in regulating cancer cell survival, proliferation, and metastasis. Abnormal regulation of these transporters, which exhibit varying affinities for hexoses, could enable cancer cells to efficiently manage their energy supply, offering a crucial edge for proliferation. Exploiting the upregulated expression of glucose transporters, GLUTs, and Sodium Linked Glucose Transporters (SGLTs), could serve as a novel therapeutic intervention for anti-cancer drug discovery as well as provide a unique targeting approach for drug delivery to specific tumor tissues. This review aims to discussthe previous and emerging research on the expression of various types of glucose transporters in tumor tissues, the role of glucose transport inhibitors as a cancer therapy intervention as well as emerging GLUT/SGLT-mediated drug delivery strategies that can be therapeutically employed to target various cancers.
Collapse
Affiliation(s)
- Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Kingdom of Saudi Arabia (KSA)
| | - Syed Arman Rabbani
- Department of Clinical Pharmacy and Pharmacology, Ras Al Khaimah College of Pharmacy, Ras Al Khaimah Medical and Health Science University, Ras Al Khaimah, United Arab Emirates
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
25
|
Aljani B, Lindner A, Weigelt M, Zhao M, Sharma V, Bonifacio E, Jones P, Eugster A. Small RNA-Seq and real time rt-qPCR reveal islet miRNA released under stress conditions. Islets 2024; 16:2392343. [PMID: 39154325 PMCID: PMC11332650 DOI: 10.1080/19382014.2024.2392343] [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: 04/29/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
Replacement of beta cells through transplantation is a potential therapeutic approach for individuals with pancreas removal or poorly controllable type 1 diabetes. However, stress and death of beta cells pose significant challenges. Circulating miRNA has emerged as potential biomarkers reflecting early beta cell stress and death, allowing for timely intervention. The aim of this study was to identify miRNAs as potential biomarkers for beta cell health. Literature review combined with small RNA sequencing was employed to select islet-enriched miRNA. The release of those miRNA was assessed by RT-qPCR in vivo, using a streptozotocin induced diabetes mouse model and in vitro, through mouse and human islets exposed to varying degrees of hypoxic and cytokine stressors. Utilizing the streptozotocin induced model, we identified 18 miRNAs out of 39 candidate islet-enriched miRNA to be released upon islet stress in vivo. In vitro analysis of culture supernatants from cytokine and/or hypoxia stressed islets identified the release of 45 miRNAs from mouse and 8 miRNAs from human islets. Investigation into the biological pathways targeted by the cytokine- and/or hypoxia-induced miRNA suggested the involvement of MAPK and PI3K-Akt signaling pathways in both mouse and human islets. We have identified miRNAs associated with beta cell health and stress. The findings allowed us to propose a panel of 47 islet-related human miRNA that is potentially valuable for application in clinical contexts of beta cell transplantation and presymptomatic early-stage type 1 diabetes.
Collapse
Affiliation(s)
- Bssam Aljani
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Annett Lindner
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Marc Weigelt
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Min Zhao
- German Center for Environmental Health, Institute of Diabetes Research, Helmholtz Munich, Munich, Germany
| | - Virag Sharma
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ezio Bonifacio
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
- Faculty of Medicine, German Center for Diabetes Research (DZD), Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Peter Jones
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London, UK
| | - Anne Eugster
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
26
|
Zou C, Liu X, He M, Sun Y, Sang Y, Peng G, Ma Y, Geng H, Liang J. Insulin Resistance Mediates the Association Between Vitamin D and Non-Alcoholic Fatty Liver Disease. Int J Prev Med 2024; 15:77. [PMID: 39867257 PMCID: PMC11759225 DOI: 10.4103/ijpvm.ijpvm_221_23] [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: 08/18/2023] [Accepted: 11/14/2023] [Indexed: 01/28/2025] Open
Abstract
Background Vitamin D (VD) deficiency and insulin resistance (IR) increase the risk of non-alcoholic fatty liver disease (NAFLD), but few studies have explored the potential mechanisms by which IR mediates the association between VD and the pathogenesis of NAFLD at the genetic level using publicly available databases. Methods This is a cross-sectional study, and we utilized the National Health and Nutrition Examination Survey (NHANES) dataset, as well as data from GSE200765 obtained from the Gene Expression Omnibus (GEO) website. A total of 723 individuals who had completed liver ultrasound examination and the detection of VD levels were included in the final analysis. A gene expression dataset, GSE200765, was also downloaded from the GEO website, to explore the potential mechanism of VD and NAFLD. Results In the NHANES data, covariates significantly differed in four VD categories, and the controlled attenuation parameter (CAP), vibration-controlled transient elastography-liver stiffness measurement (VCTE-LSM), and IR were reduced with an increase in VD levels. Mediation analysis revealed that IR mediated the association between VD and both CAP and LSM, and the estimated mediation effects were 29.0% and 39.8%, respectively. Bioinformatics analysis showed that seven differentially expressed genes (DEGs) (solute carrier family 2 member 2 [SLC2A2], protein phosphatase 1 regulatory subunit 3E [PPP1R3E], CAMP responsive element binding protein 3-like 3 [CREB3L3], Interleukin-6 [IL-6], peroxisome proliferator-activated receptor gamma coactivator 1-alpha [PPARGC1A], nuclear factor kappa B inhibitor alpha [NFKBIA], and phosphoenolpyruvate carboxykinase 2 [PCK2]) were enriched in the IR pathway in comparison groups (VD group vs. lipid group), suggesting that VD improved NAFLD via changed IR. Conclusions VD deficiency and IR were the risk factors for NAFLD, and increased VD levels improved the status of NAFLD. The underlying mechanism may be that elevated VD levels reduced IR, which improved the expression of DEGs involved in the IR pathway.
Collapse
Affiliation(s)
- Caiyan Zou
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Affiliated Hospital of Southeast University, Xuzhou Clinical School of Nanjing Medical University, Xuzhou, Jiangsu, China
| | - Xuekui Liu
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Affiliated Hospital of Southeast University, Xuzhou Clinical School of Nanjing Medical University, Xuzhou, Jiangsu, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Institute of Medical Science, Jiangsu Province, China
| | - Maosheng He
- Department of Ultrasound, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Yan Sun
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Affiliated Hospital of Southeast University, Xuzhou Clinical School of Nanjing Medical University, Xuzhou, Jiangsu, China
| | - Yiquan Sang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Affiliated Hospital of Southeast University, Xuzhou Clinical School of Nanjing Medical University, Xuzhou, Jiangsu, China
| | - Gangshan Peng
- Department of Graduate School, Xuzhou Medical University, Xuzhou, China
| | - Yamei Ma
- Department of Bengbu Medical College, Bengbu, China
| | - Houfa Geng
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Affiliated Hospital of Southeast University, Xuzhou Clinical School of Nanjing Medical University, Xuzhou, Jiangsu, China
| | - Jun Liang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Affiliated Hospital of Southeast University, Xuzhou Clinical School of Nanjing Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
27
|
Xia B, Zhao D, Hao Q, Yu J, Han Y, Ling L, Zhao R, Zhao J. Effects of fishing stress on fatty acid and amino acid composition and glycolipid metabolism in triploid rainbow trout. Food Chem 2024; 461:140904. [PMID: 39181054 DOI: 10.1016/j.foodchem.2024.140904] [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: 05/24/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Triploid Oncorhynchus mykiss is an important economic fish worldwide. Fishing stress can affect its growth and meat quality. This study first explored the effects of fishing stress on fatty acid and amino acid in triploid O. mykiss. Results showed fishing stress significantly reduced the content of docosadienoic acid, Gly, Arg, and DAA (P < 0.05). Targeted lipidomics analysis furthered suggested that some lipid molecules belonging to TG, DG, PC, Cer, ChE, and So were significantly up-regulated; while some lipid molecules belonging to Cer, LPE, LPC, PS, PC, and SM were significantly down-regulated, suggesting an accelerated glycolipid metabolism. Eventually, the glycolipid metabolism-related enzyme activity and gene expressions were examined, and the results indicated that O. mykiss was anti-oxidative stress by affecting relevant glycolipid metabolism signaling pathways and participating in cellular redox homeostasis. Findings of this study provide a theoretical foundation for further investigation into the mechanisms through which fishing stress affects O. mykiss.
Collapse
Affiliation(s)
- Banghua Xia
- Northeast Agricultural University, Harbin 150030, China
| | - Dandan Zhao
- Northeast Agricultural University, Harbin 150030, China
| | - Qirui Hao
- Northeast Agricultural University, Harbin 150030, China; Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Junfei Yu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Yue Han
- Liaoyang Development and Reform Service Center, Liaoyang 111001, China
| | - Ling Ling
- Northeast Agricultural University, Harbin 150030, China
| | - Rongwei Zhao
- Harbin Agricultural Technology Extension Master station, Harbin 150023, China
| | - Junwei Zhao
- Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
28
|
Amer J, Amleh A, Salhab A, Kolodny Y, Yochelis S, Saffouri B, Paltiel Y, Safadi R. High-fat diet mouse model receiving L-glucose supplementations propagates liver injury. Front Nutr 2024; 11:1469952. [PMID: 39742098 PMCID: PMC11687001 DOI: 10.3389/fnut.2024.1469952] [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: 07/24/2024] [Accepted: 11/11/2024] [Indexed: 01/03/2025] Open
Abstract
Background and aims Limited data link manufactured sweeteners impact on metabolic dysfunction-associated steatotic liver disease (MASLD). We aimed to evaluate the effects of manufactured sugars (L-glucose) compared to natural sugars (D-glucose) on phenotype, molecular and metabolic changes in mice models fed with either regular diet (RD) or high fat diet (HFD). Methods C57BL/6 mice fed 16-weeks with either RD; 70% carbohydrate or HFD; 60% fat, with or without additional glucose (Glu, at 18% w/v) to drinking tap water at weeks 8-16; of either natural (D-Glu) or manufactured (L-Glu) sugars. Liver inflammation (ALT and AST serum levels, liver H&E histologic stains and cell viability profile by p-AKT), liver fibrosis [quantitated α smooth-muscle-actin (αSMA) by western blot and RT-PCR, Masson Trichrome staining (MTC) of liver tissue], liver lipid [steatosis stain by H&E, Adipose Differentiation-Related Protein (ADRP) lipid accumulation, serum and lipid peroxidation Malondialdehyde (MDA) markers by ELISA], glucose hemostasis (serum Glucose and C-peptide with HOMA-IR score calculation) and liver aspects [hepatic glucose transporter 2 (GLUT2), insulin receptor (IR) expressions and GYS2/PYGL ratio] evaluated. Results D- and L-Glu supplementations propagate hepatocytes ballooning and steatosis in HFD-fed mice and were associated with αSMA down-expressions by 1.5-fold compared to the untreated group while showed an acceleration in liver fibrosis in the RD-fed mice. Lipid profile (Steatosis, ADRP and MDA) significantly increased in HFD-fed mice, both Glu supplementations (mainly the L-Glu) increased serum MDA while decreased ADRP. HOMA-IR score and IR significantly increased in HFD-fed mice, with further elevation in HOMA-IR score following Glu supplementations (mainly L-Glu). The increase in HOMA-IR negatively correlated with IR and Glut2 expressions. D- and L-Glu supplementations showed significant decrease of Glycogenesis (low GYS2/PYGL ratio) and unchanged p-AKT pattern compared to their RD counterparts. Conclusion Our data indicate an increase in rate of de-novo lipogenesis (DNL) in RD-fed mice (High carbohydrate diet) and liver fibrosis following additional sugar supplementations. In contrast, HFD-fed mice (with pre-existing high lipid profile) supplemented with sugar showed less liver fibrosis, because of reduced de-novo fatty acids synthesis and subsequently, the lipid oxidation pathways become dominated and induce the net results of lipid clearance.
Collapse
Affiliation(s)
- Johnny Amer
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Athar Amleh
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Ahmad Salhab
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Yuval Kolodny
- Applied Physics Department, Center for Nanoscience and Nanotechnology, Hebrew University Givaat Ram, Jerusalem, Israel
| | - Shira Yochelis
- Applied Physics Department, Center for Nanoscience and Nanotechnology, Hebrew University Givaat Ram, Jerusalem, Israel
| | - Baker Saffouri
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Yossi Paltiel
- Applied Physics Department, Center for Nanoscience and Nanotechnology, Hebrew University Givaat Ram, Jerusalem, Israel
| | - Rifaat Safadi
- Liver Institute, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| |
Collapse
|
29
|
Sun B, Chen H. Nickel Sulfate-Induced GSIS Injury in MIN6 Cells by Activating the JNK Pathway Through Oxidative Stress. Biol Trace Elem Res 2024:10.1007/s12011-024-04477-x. [PMID: 39661292 DOI: 10.1007/s12011-024-04477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Nickel has an impact on human health, especially in the context of the new energy industries. Nickel's influence on glycemia remains controversial, and the effects and mechanisms of nickel on islet function still need further exploration. MIN6 cells were treated with different concentrations of nickel sulfate (NiSO4) (0, 75, 150, and 300 µg/mL) for different durations (0, 12, 24, and 48 h). The study measured cell cycle progression, apoptosis, reactive oxygen species (ROS) production, oxidative stress-related indexes (T-SOD, TBARS, 8-OHdG, and GSH), glucose-induced insulin secretion (GSIS), and the expression of JNK pathway-related proteins, pancreaticoduodenal homeobox-1 (PDX-1), glucose transporter 2 (GLUT2), and forkhead box protein O1 (FOXO1). NiSO4 damaged MIN6 cells in a time- and dose-dependent manner. NiSO4 blocked the cell cycle, induced apoptosis, and reduced insulin secretion in the GSIS experiment. NiSO4 also induced ROS production, increased oxidative stress-related indexes (TRABS and 8-OHdG), and decreased antioxidant stress-related indexes (GSH and T-SOD). In addition, NiSO4 activated the JNK pathway, upregulated FOXO1 protein expression, and inhibited PDX-1 and GLUT2 protein expression, affecting insulin release during GSIS. NiSO4 inhibited the proliferation of MIN6 cells through oxidative stress, aggravated apoptosis, caused functional impairment, upregulated the expression of FOXO1 by activating the JNK pathway, inhibited the expression of PDX-1 and GLUT2 proteins, and impaired the GSIS function of islets.
Collapse
Affiliation(s)
- Bo Sun
- Endocrine and Metabolism Department, Lanzhou University Second Hospital, Chengguan District, No. 82, CuyingmenLanzhou, 730000, Gansu, China
- Department of Infantile Endocrine Genetic Metabolism, Gansu Maternal and Child Health Care Hospital, Lanzhou, 730000, Gansu, China
| | - Hui Chen
- Endocrine and Metabolism Department, Lanzhou University Second Hospital, Chengguan District, No. 82, CuyingmenLanzhou, 730000, Gansu, China.
| |
Collapse
|
30
|
Bin Pan, Xie Y, Shao W, Fang X, Han D, Li J, Hong X, Tu W, Shi J, Yang M, Tian F, Xia M, Hu J, Ren J, Kan H, Xu Y, Li W. Prenatal exposure to PM 2.5 disturbs the glucose metabolism of offspring fed with high-fat diet in a gender-dependent manner. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117404. [PMID: 39615301 DOI: 10.1016/j.ecoenv.2024.117404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Studies have shown that maternal exposure to PM2.5 could potentially disrupt glucose and lipid metabolism in offspring supplied with high-fat diet, yet whether this effect is gender-dependent or not and the underlying biological mechanisms are not well understood. In our current research, female ICR mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CAP) before and during pregnancy. The offspring mice were fed with control diet (CD) or high-fat diet (HFD) for 9 weeks, and their metabolic conditions were analyzed. Our findings reveal that maternal exposure to PM2.5 induced glucose intolerance and insulin resistance in female offspring fed with HFD but not in males. Specifically, hepatic insulin resistance as indicated by significantly decreased AKT phosphorylation (p-AKT) level, changed liver structure as indicated by increased ballooning and steatosis based on H&E staining images, and impaired liver function as indicated by up-regulated ALT activity were observed in HFD-fed female offspring from CAP-exposed mothers in comparison to those from FA-exposed ones. Further analysis indicated that these impacts of prenatal PM2.5 exposure on glucose metabolism in offspring may result from disturbed gluconeogenesis and induced inflammatory response in liver. Our research underscores that prenatal PM2.5 exposure induces glucose metabolism abnormalities in offspring fed with HFD in a gender-dependent manner, and the liver potentially serves as a key player in mediating these effects of maternal PM2.5 exposure.
Collapse
Affiliation(s)
- Bin Pan
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China; Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yuanting Xie
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Wenpu Shao
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xinyi Fang
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Dongyang Han
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jingyu Li
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xiaoqing Hong
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Wenyue Tu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jiayi Shi
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Mingjun Yang
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Fang Tian
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Minjie Xia
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Jingying Hu
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Jianke Ren
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China.
| | - Weihua Li
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drugs and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of pharmacy, Fudan University, Shanghai 200237, China.
| |
Collapse
|
31
|
Farhadi Z, Khaksari M, Alivirdiloo V, Mehrjerdi FZ, Alborzi N, Baktash KB, Rezvani ME. Review on the role of hypothalamic astrocytes in the neuroendocrine control of metabolism. J Diabetes Metab Disord 2024; 23:1635-1643. [PMID: 39610541 PMCID: PMC11599663 DOI: 10.1007/s40200-024-01465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 11/30/2024]
Abstract
Astrocytes are the most numerous type of glial cells found in the nervous system. They regulate energy homeostasis in collaboration with the neuronal circuits involved in energy balance. These glial cells are equipped with sensors and receptors for nutrients and metabolic hormones in order to control energy homeostasis. Astrocytes, like hypothalamic appetite-regulating neurons, are vulnerable to the negative consequences of a high-fat diet (HFD) feeding, which is associated with an inflammatory response and transforms them into a reactive astrocyte state, consequently leading to the disruption of energy balance. Additionally, these cells have sexually dimorphic characteristics, which will lead to different metabolic outcomes in males and females. In this review, we will discuss the various physiological and pathophysiological roles of astrocytes in regulating energy balance. Finally, we will discuss the sexual dimorphism in astrocytes and the impact of estrogen on eliciting distinct responses.
Collapse
Affiliation(s)
- Zeinab Farhadi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Khaksari
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Alivirdiloo
- Medical Doctor Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Fatemeh Zare Mehrjerdi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Nasrin Alborzi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | | | - Mohammad Ebrahim Rezvani
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| |
Collapse
|
32
|
Solak H, Gormus ZIS, Koca RO, Gunes CE, Iyisoy MS, Kurar E, Kutlu S. 'The effect of neuropeptide Y1 receptor agonist on hypothalamic neurogenesis in rat experimental depression model'. Metab Brain Dis 2024; 40:39. [PMID: 39576364 DOI: 10.1007/s11011-024-01445-1] [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: 04/24/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024]
Abstract
Depression is responsible for neuropathies such as decreased neurogenesis and increased dendritic atrophy. There is information that antidepressant treatments have an effect by increasing hippocampal neurogenesis and neurotrophic factor expression. The neuropeptide Y1 (NPY1R) receptor agonist has been suggested to have anxiolytic effects. Based on this information, it was aimed to investigate the effect of NPY1R agonist on depression in rats with depression using the CMS model and to determine how depression affects cell proliferation in the hypothalamus and hypothalamic peptide levels. Forty-eight adult, male Wistar albino rats were divided into groups as Control, Depression (D), Depression + NPY1R and NPY1R. Various stressors were applied to D for 30 days. An open field test (OFT) and forced swim test (FST) were performed to check whether the animals were depressed. On the 16th day, an osmotic mini pump was placed under the skin and NPY1R (130 ul/kg/day) was applied for 15 days. Behavioral tests were performed, hypothalamic peptide gene expression levels were analyzed by quantitative RT-PCR and statistical evaluations were made using ANOVA. A decrease in the percentage of movement in the D and control groups were noted in the OFT, an increase in the immobility time in the D group in the FST, and an increase in swimming behavior in the DNPY1R group. The animals did not display any anxiety behavior based on the elevated plus maze test results. It caused a decrease in IGF1R, FGF2, POMC, NPY and GLUT2 gene expression in the hypothalamus of depression group animals, and an increase in NPY gene expression in NPY1R treatment. This study compellingly demonstrated that exposure to chronic mild stress simultaneously downregulates gene expression in the hypothalamus; we observed that NPY receptor NPY1R treatment increased the effect of NPY. Therefore, adjunctive treatments with appropriate molecules such as NPY, Y1 receptor agonists or pharmacological derivatives may have significant potential in the treatment of depression.
Collapse
Affiliation(s)
- Hatice Solak
- Department of Physiology, Faculty of Medicine, Kutahya Health Science University, Kutahya, Turkey.
| | - Z Isik Solak Gormus
- Department of Physiology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Raviye Ozen Koca
- Department of Physiology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Canan Eroglu Gunes
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Mehmet Sinan Iyisoy
- Department of Medical Education and Informatics, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ercan Kurar
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Selim Kutlu
- Department of Physiology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
33
|
Zhang M, Yin YS, May KS, Wang S, Purcell H, Zhang XS, Blaser MJ, den Hartigh LJ. The role of intestinal microbiota in physiologic and body compositional changes that accompany CLA-mediated weight loss in obese mice. Mol Metab 2024; 89:102029. [PMID: 39293564 PMCID: PMC11447304 DOI: 10.1016/j.molmet.2024.102029] [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: 04/05/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
OBJECTIVE Obesity continues to be a major problem, despite known treatment strategies such as lifestyle modifications, pharmaceuticals, and surgical options, necessitating the development of novel weight loss approaches. The naturally occurring fatty acid, 10,12 conjugated linoleic acid (10,12 CLA), promotes weight loss by increasing fat oxidation and browning of white adipose tissue, leading to increased energy expenditure in obese mice. Coincident with weight loss, 10,12 CLA also alters the murine gut microbiota by enriching for microbes that produce short chain fatty acids (SCFAs), with concurrent elevations in fecal butyrate and plasma acetate. METHODS To determine if the observed microbiota changes are required for 10,12 CLA-mediated weight loss, adult male mice with diet-induced obesity were given broad-spectrum antibiotics (ABX) to perturb the microbiota prior to and during 10,12 CLA-mediated weight loss. Conversely, to determine whether gut microbes were sufficient to induce weight loss, conventionally-raised and germ-free mice were transplanted with cecal contents from mice that had undergone weight loss by 10,12 CLA supplementation. RESULTS While body weight was minimally modulated by ABX-mediated perturbation of gut bacterial populations, adult male mice given ABX were more resistant to the increased energy expenditure and fat loss that are induced by 10,12 CLA supplementation. Transplanting cecal contents from donor mice losing weight due to oral 10,12 CLA consumption into conventional or germ-free mice led to improved glucose metabolism with increased butyrate production. CONCLUSIONS These data suggest a critical role for the microbiota in diet-modulated changes in energy balance and glucose metabolism, and distinguish the metabolic effects of orally delivered 10,12 CLA from cecal transplantation of the resulting microbiota.
Collapse
Affiliation(s)
- Meifan Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Yue S Yin
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Karolline S May
- Department of Medicine: Metabolism, Endocrinology, and Nutrition, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Shari Wang
- Department of Medicine: Metabolism, Endocrinology, and Nutrition, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Hayley Purcell
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Laura J den Hartigh
- Department of Medicine: Metabolism, Endocrinology, and Nutrition, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
34
|
Jiménez-Sánchez C, Oberhauser L, Maechler P. Role of fatty acids in the pathogenesis of ß-cell failure and Type-2 diabetes. Atherosclerosis 2024; 398:118623. [PMID: 39389828 DOI: 10.1016/j.atherosclerosis.2024.118623] [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: 07/31/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Pancreatic ß-cells are glucose sensors in charge of regulated insulin delivery to the organism, achieving glucose homeostasis and overall energy storage. The latter function promotes obesity when nutrient intake chronically exceeds daily expenditure. In case of ß-cell failure, such weight gain may pave the way for the development of Type-2 diabetes. However, the causal link between excessive body fat mass and potential degradation of ß-cells remains largely unknown and debated. Over the last decades, intensive research has been conducted on the role of lipids in the pathogenesis of ß-cells, also referred to as lipotoxicity. Among various lipid species, the usual suspects are essentially the non-esterified fatty acids (NEFA), in particular the saturated ones such as palmitate. This review describes the fundamentals and the latest advances of research on the role of fatty acids in ß-cells. This includes intracellular pathways and receptor-mediated signaling, both participating in regulated glucose-stimulated insulin secretion as well as being implicated in ß-cell dysfunction. The discussion extends to the contribution of high glucose exposure, or glucotoxicity, to ß-cell defects. Combining glucotoxicity and lipotoxicity results in the synergistic and more deleterious glucolipotoxicity effect. In recent years, alternative roles for intracellular lipids have been uncovered, pointing to a protective function in case of nutrient overload. This requires dynamic storage of NEFA as neutral lipid droplets within the ß-cell, along with active glycerolipid/NEFA cycle allowing subsequent recruitment of lipid species supporting glucose-stimulated insulin secretion. Overall, the latest studies have revealed the two faces of the same coin.
Collapse
Affiliation(s)
- Cecilia Jiménez-Sánchez
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Lucie Oberhauser
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland.
| |
Collapse
|
35
|
Liu X, Du P, Xu J, Wang W, Zhang C. Therapeutic Effects of Intermittent Fasting Combined with SLBZS and Prebiotics on STZ-HFD-Induced Type 2 Diabetic Mice. Diabetes Metab Syndr Obes 2024; 17:4013-4030. [PMID: 39492963 PMCID: PMC11531242 DOI: 10.2147/dmso.s474196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose This study aims to assess the therapeutic potential of combining Shen-Ling-Bai-Zhu-San (SLBZS) or prebiotics with intermittent fasting (IF) in type 2 diabetes mellitus (T2DM) mice and to investigate the synergistic effects and underlying mechanisms. Methods Type 2 diabetic mouse models were induced using high-fat diet (HFD) and streptozotocin (STZ), followed by IF treatment. Mice were then grouped for combined therapy with different doses of SLBZS and prebiotics. Fasting blood glucose (FBG) levels, body weight variations, and oral glucose tolerance tests were assessed to elucidate metabolic alterations. The hepatic and renal parameters were evaluated to determine systemic changes in T2DM mice, while the insulin levels were quantified by ELISA to assess glucose homeostasis. Gut microbiota alterations were examined via 16S rRNA sequencing. Alterations of the genes in relevant signaling pathways were analyzed using RT-qPCR. Results IF improved FBG, body weight, insulin levels, and other diabetes indicators. Combined IF with SLBZS or prebiotics yielded similar effects. Furthermore, it ameliorated dyslipidemia and mitigated hepatic and renal parameters in T2DM mice. Pancreatic tissue histopathology showed islet cell restoration post-intervention. IF therapy reduced the abnormally elevated GSK-3β gene expression and increased the abnormally reduced GLUT2 genes. Further analysis indicated that the combination of IF with prebiotics and high doses of SLBZS upregulated the expression of the INSR and IRS1 genes. Gut microbiota analysis revealed restored diversity and structure, with notable changes in specific bacterial families. At the family level, the contents of Akkermansiaceae and Bifidobacteriaceae were restored. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt2) analysis suggested metabolic pathway alterations. Conclusion IF improved type 2 diabetic symptoms, with combined SLBZS and prebiotics showing similar effects. IF with high concentration of SLBZS and prebiotics doses upregulated the INSR and IRS1 genes and had superior effects on gut microbiota compared to IF alone.
Collapse
Affiliation(s)
- Xiaoyu Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Pengyun Du
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jianing Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wei Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Chenggang Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
36
|
Li Y, Li W, Zhu X, Xu N, Meng Q, Jiang W, Zhang L, Yang M, Xu F, Li Y. VEGFB ameliorates insulin resistance in NAFLD via the PI3K/AKT signal pathway. J Transl Med 2024; 22:976. [PMID: 39468621 PMCID: PMC11520811 DOI: 10.1186/s12967-024-05621-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: 04/29/2024] [Accepted: 08/19/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is one of the most universal liver diseases with complicated pathogenesis throughout the world. Insulin resistance is a leading risk factor that contributes to the development of NAFLD. Vascular endothelial growth factor B (VEGFB) was described by researchers as contributing to regulating lipid metabolic disorders. Here, we investigated VEGFB as a main target to regulate insulin resistance and metabolic syndrome. METHODS In this study, bioinformatics, transcriptomics, morphological experiments, and molecular biology were used to explore the role of VEGFB in regulating insulin resistance in NAFLD and its molecular mechanism based on human samples, animal models, and cell models. RNA-seq was performed to analyze the signal pathways associated with VEGFB and NAFLD; Palmitic acid and High-fat diet were used to induce insulin-resistant HepG2 cells model and NAFLD animal model. Intracellular glucolipid contents, glucose uptake, hepatic and serum glucose and lipid levels were examined by Microassay and Elisa. Hematoxylin-eosin staining, Oil Red O staining, and Periodic acid-schiff staining were used to analyze the hepatic steatosis, lipid droplet, and glycogen content in the liver. Western blot and quantitative real-time fluorescent PCR were used to verify the expression levels of the VEGFB and insulin resistance-related signals PI3K/AKT pathway. RESULTS We observed that VEGFB is genetically associated with NAFLD and the PI3K/AKT signal pathway. After VEGFB knockout, glucolipids levels were increased, and glucose uptake ability was decreased in insulin-resistant HepG2 cells. Meanwhile, body weight, blood glucose, blood lipids, and hepatic glucose of NAFLD mice were increased, and hepatic glycogen, glucose tolerance, and insulin sensitivity were decreased. Moreover, VEGFB overexpression reduced glucolipids and insulin resistance levels in HepG2 cells. Specifically, VEGFB/VEGFR1 activates the PI3K/AKT signals by activating p-IRS1Ser307 expression, inhibiting p-FOXO1pS256 and p-GSK3Ser9 expressions to reduce gluconeogenesis and glycogen synthesis in the liver. Moreover, VEGFB could also enhance the expression level of GLUT2 to accelerate glucose transport and reduce blood glucose levels, maintaining glucose homeostasis. CONCLUSIONS Our studies suggest that VEGFB could present a novel strategy for treating NAFLD as a positive factor.
Collapse
Affiliation(s)
- Yuqi Li
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Wenhao Li
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Xiaonan Zhu
- Department of Intensive Care Medicine, The Second School of Clinical Medical, Binzhou Medical University, Yantai, Shandong, China
| | - Nuo Xu
- Department of Intensive Care Medicine, The Second School of Clinical Medical, Binzhou Medical University, Yantai, Shandong, China
| | - Qinyu Meng
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Wenguo Jiang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Lei Zhang
- Department of Infectious Diseases, The Second School clinical Medicine, YanTai Affiliated Hospital of Bin Zhou Medical University, Yantai, China
| | - Meizi Yang
- Department of Pharmacology, School of Basic Medicine of Binzhou Medical University, Yantai, Chian, China
| | - Fang Xu
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China.
| | - Yana Li
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China.
| |
Collapse
|
37
|
Hu J, Zheng L, Fan X, Lang H, Xie H, Lin N. Ameliorative effects of Penthorum chinense Pursh on insulin resistance and oxidative stress in diabetic obesity db/db mice. PLoS One 2024; 19:e0311502. [PMID: 39374222 PMCID: PMC11458015 DOI: 10.1371/journal.pone.0311502] [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: 05/10/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Penthorum chinense Pursh (PCP), a medicinal and edible plant, has been reported to protect against liver damage by suppressing oxidative stress. Type 2 diabetes mellitus (T2DM) is associated with liver dysfunction and oxidative stress. In the present study, we aim to investigate the hypoglycemic effect of PCP on db/db mice and further explore the underlying mechanisms. METHODS Thirty-two db/db mice were randomized into four groups, including a diabetic model control group (MC) and three diabetic groups treated with low (LPCP, 300 mg/kg/d), medium (MPLP, 600 mg/kg/d), and high doses of PCP (HPCP, 1200 mg/kg/d), and the normal control group (NC) of eight db/m mice were included. Mice in the NC and MC groups received the ultrapure water. After four weeks of intervention, parameters of fasting blood glucose (FBG), insulin resistance (IR), blood lipid levels, hepatic oxidative stress, and enzymes related to hepatic glucose metabolism were compared in the groups. RESULTS PCP administration significantly reduced FBG and IR in diabetic db/db mice, and improved hepatic glucose metabolism by increasing glucose transporter 2 (GLUT2) and glucokinase (GCK) protein expression. Meanwhile, PCP supplementation ameliorated hepatic oxidative stress by decreasing malonaldehyde content and increasing the activities of superoxide dismutase and glutathione peroxidase in db/db mice. Furthermore, PCP treatment reduced obesity and food intake in db/db mice, and improved dyslipidemia demonstrated by increasing high-density lipoprotein cholesterol (HDL-C) while decreasing total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (HDL-C). All doses of PCP treatment decreased the values of LDL-C/HDL-C in a dose-response relationship. CONCLUSION PCP significantly alleviated hyperglycemia, hyperinsulinemia, hyperlipidemia, and obesity, inhibited hepatic oxidative stress, and enhanced hepatic glucose transport in T2DM mice. Based on the above findings, the hypoglycemic effect of PCP may be attributed to the activation of the GLUT2/GCK expression in the liver and the reduction of hepatic oxidative stress.
Collapse
Affiliation(s)
- Jilei Hu
- Department of Clinical Nutrition, The General Hospital of Western Theater Command, Chengdu, P. R. China
- School of Public Health, Southwest Medical University, Luzhou, P. R. China
| | - Leyu Zheng
- School of Public Health, Southwest Medical University, Luzhou, P. R. China
- Wanzhou District Market Supervision Administration, Chongqing, P. R. China
| | - Xi Fan
- School of Public Health, Southwest Medical University, Luzhou, P. R. China
| | - Hongmei Lang
- General Medicine, Chengdu Second People’s Hospital, Chengdu, P. R. China
| | - Huibo Xie
- School of Public Health, Southwest Medical University, Luzhou, P. R. China
| | - Ning Lin
- Department of Clinical Nutrition, The General Hospital of Western Theater Command, Chengdu, P. R. China
| |
Collapse
|
38
|
Zhang Y, Jiang X, Wu D, Huang H, Jia G, Zhao G. Sema4D deficiency enhances glucose tolerance through GLUT2 retention in hepatocytes. J Transl Med 2024; 22:864. [PMID: 39334386 PMCID: PMC11429007 DOI: 10.1186/s12967-024-05694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The glucose transporter 2 (GLUT2) is constitutively expressed in pancreatic beta cells and hepatocytes of mice. It is the most important receptor in glucose-stimulated insulin release and hepatic glucose transport. The Sema4D is a signalin receptor on cell membranes. The correlation between Sema4D and GLUT2 has not been reported previously. We investigated whether knockdown of Sema4D could exert a hypoglycemic effect based on the increased GLUT2 expression in Sema4D -/- mice hepatocytes. METHODS The glucose tolerance test and insulin tolerance test in sema4D -/- and sema4D +/+ mice were compared before and after streptozotocin (STZ) injection; the expression of GLUT2 content on the membrane surface of both groups was verified by Western blot. Then, the levels of insulin and C-peptide in the serum of the two groups of mice after STZ injection were measured by ELISA; the differentially expressed mRNAs in the liver of the two groups of mice were analyzed by transcriptomic analysis; then the differences in the expression of GLUT2, glycogen, insulin and glucagon in the two groups of mice were compared by tissue section staining. Finally, metabolomics analysis was performed to analyze the metabolites differentially expressed in the two groups of mice. KEY FINDINGS First, Sema4D -/- male mice exhibited significantly greater glucose tolerance than wild-type mice in a hyperglycemic environment. Secondly, Sema4D -/- mice had more retained GLUT2 in liver membranes after STZ injection according to an immunofluorescence assay. After STZ injection, Sema4D -/- male mice did not exhibit fasting hyperinsulinemia like wild-type mice. Finally, analysis of metabolomic and immunohistochemical data also revealed that Sema4D -/- mice produce hypoglycemic effects by enhancing the pentose phosphate pathway, but not glycogen synthesis. CONCLUSIONS Thus, Sema4D may play an important role in the regulation of glucose homeostasis by affecting GLUT2 synthesis.
Collapse
Affiliation(s)
- Yanling Zhang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Xiaomei Jiang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Dongsong Wu
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Hao Huang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China.
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
39
|
Wang W, Chen S, Jiang Y, Ji J, Cong R. Expression of the C-allele of intronic rs8192675 in SLC2A2 is associated with improved glucose response to metformin. Genet Mol Biol 2024; 47:e20230281. [PMID: 39535164 PMCID: PMC11559485 DOI: 10.1590/1678-4685-gmb-2023-0281] [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: 10/07/2023] [Accepted: 05/30/2024] [Indexed: 11/16/2024] Open
Abstract
Glucose is a critical nutrient for energy metabolism. The SLC2A2 gene is essential for glucose sensing and homeostasis, as it encodes the facilitated glucose transporter GLUT2. During diabetes treatment, the C-allele of rs8192675 in SLC2A2 has been found to regulate the action of metformin and reduce the absolute level of HbA1c more effectively than the T-allele. In this study, stable HEK293T cell lines carrying the CC, CT, and TT genotypes of rs8192675 in SLC2A2 were generated using CRISPR/Cas9-mediated genome editing. GLUT2 mRNA and protein levels were elevated in cell clones with the TC genotype compared to those with the CC genotype but were reduced relative to the TT genotype. Additionally, high concentrations of glucose or fructose induced more GLUT2 protein production in CT-genotype cells than that induced in CC-genotype cells, yet less than that induced in TT-genotype cells. Metformin induced a greater increase in GLUT2 expression and a smaller increase in activated AMPK protein expression in CC-genotype cells than those induced in TT-genotype cells, resulting in a remarkable reduction in activated mTOR and S6 levels. This study directly supports the biological mechanism linking the C-allele of rs8192675 with improved treatment outcomes in metformin therapy for diabetes.
Collapse
Affiliation(s)
- Wanjun Wang
- Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Suying Chen
- Affiliated Hospital 2 of Nantong University, Department of Radiology, No.666 Shengli Road, Nantong, Jiangsu Province, China
| | - Yilei Jiang
- Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Jianhong Ji
- Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Intensive Care Unit, Nantong, People's Republic of China
| | - Ruochen Cong
- Affiliated Hospital 2 of Nantong University, Department of Radiology, No.666 Shengli Road, Nantong, Jiangsu Province, China
| |
Collapse
|
40
|
Zhang L, Wu M, Zhang J, Liu T, Fu S, Wang Y, Xu Z. The pivotal role of glucose transporter 1 in diabetic kidney disease. Life Sci 2024; 353:122932. [PMID: 39067659 DOI: 10.1016/j.lfs.2024.122932] [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: 04/05/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Diabetes mellitus (DM) is a significant public health problem. Diabetic kidney disease (DKD) is the most common complication of DM, and its incidence has been increasing with the increasing prevalence of DM. Given the association between DKD and mortality in patients with DM, DKD is a significant burden on public health resources. Despite its significance in DM progression, the pathogenesis of DKD remains unclear. Aberrant glucose uptake by cells is an important pathophysiological mechanism underlying DKD renal injury. Glucose is transported across the bilayer cell membrane by a glucose transporter (GLUT) located on the cell membrane. Multiple GLUT proteins have been identified in the kidney, and GLUT1 is one of the most abundantly expressed isoforms. GLUT1 is a crucial regulator of intracellular glucose metabolism and plays a key pathological role in the phenotypic changes in DKD mesangial cells. In an attempt to understand the pathogenesis of DKD better, we here present a review of studies on the role of GLUT1 in the development and progression of DKD.
Collapse
Affiliation(s)
- Li Zhang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Meiyan Wu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jizhou Zhang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Tingting Liu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shaojie Fu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhonggao Xu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
41
|
Simha N A, Patil SM, M K J, N C, Wong LS, Kijsomporn J, Raj R, Ramu R. From sugar binders to diabetes fighters: the lectin saga of antihyperglycemic activity through systematic review and meta-analysis. Front Pharmacol 2024; 15:1382876. [PMID: 39323638 PMCID: PMC11422237 DOI: 10.3389/fphar.2024.1382876] [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: 02/06/2024] [Accepted: 08/13/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Lectins are carbohydrate-binding proteins that are extremely selective for sugar groups in the other molecules. As a result, they perform a variety of roles in biological processes involving cell, carbohydrate, and protein recognition at the cellular and molecular levels. Because lectins can bind to carbohydrates, they may play a role in determining the rate of carbohydrate digestion. They also bind to some proteins involved in diabetes mellitus (DM) pathophysiology. The present review aims to summarize the efficiency of lectins from different sources as potential antihyperglycemic agents. Methods The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were employed for the drafting. In this regard, published scientific articles on the effects of different lectins on blood glucose (BG), glucose tolerance, hormonal effects, carbohydrate-digesting enzymes, oxidative stress, and insulin production process were collected from reputed journals using electronic databases. Furthermore, the toxicity effects of lectins from different sources were collected. A specific keyword search was completed to collect numerous articles with unique experimental designs and significant results. This was followed by the selection of the requisite articles based on the criteria designed by the authors. Data extraction was based on the common research elements included in the articles. Results and Discussion Of 13 identified studies, 11 studies were considered after double screening based on the inclusion criteria. All 11 pharmacological investigations were considered for review. Subsequent studies reflected on the pharmacological properties of lectins on the levels of BG, oxidative stress, β-cell proliferation, insulin resistance, inhibition of carbohydrate digesting enzymes, body weight, food and water intake, lipid profile, and other parameters. This review highlights lectins as potential anti-diabetic agents. Conclusion However, due to limited research, systematic evaluation is recommended for their development and promotion as effective potential antihyperglycemic agents. The clinical efficacy and safety of lectins against diabetes mellitus must also be evaluated.
Collapse
Affiliation(s)
- Akshaya Simha N
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Shashank M Patil
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Jayanthi M K
- Department of Pharmacology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Chaitra N
- Division of Medical Statistics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, Malaysia
| | | | - Ranjith Raj
- Department of Pharmacology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Ramith Ramu
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| |
Collapse
|
42
|
Barros LF, Schirmeier S, Weber B. The Astrocyte: Metabolic Hub of the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041355. [PMID: 38438188 PMCID: PMC11368191 DOI: 10.1101/cshperspect.a041355] [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: 03/06/2024]
Abstract
Astrocytic metabolism has taken center stage. Interposed between the neuron and the vasculature, astrocytes exert control over the fluxes of energy and building blocks required for neuronal activity and plasticity. They are also key to local detoxification and waste recycling. Whereas neurons are metabolically rigid, astrocytes can switch between different metabolic profiles according to local demand and the nutritional state of the organism. Their metabolic state even seems to be instructive for peripheral nutrient mobilization and has been implicated in information processing and behavior. Here, we summarize recent progress in our understanding of astrocytic metabolism and its effects on metabolic homeostasis and cognition.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos, Valdivia 5110465, Chile
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Valdivia 5110693, Chile
| | - Stefanie Schirmeier
- Technische Universität Dresden, Department of Biology, 01217 Dresden, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, 8057 Zurich, Switzerland
| |
Collapse
|
43
|
Yin R, Ke J, Zhao M, Ding Y, Li W, Li M, Hu L, Dai X, Hong G. Synaptotagmin-1 antagonizes paraquat intracellular accumulation and nephrocyte toxicity by up-regulating SERBP1/GLUT2 expression. Chem Biol Interact 2024; 400:111165. [PMID: 39059605 DOI: 10.1016/j.cbi.2024.111165] [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: 06/18/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Acute kidney injury (AKI) is common and an independent risk factor for mortality in patients with paraquat (PQ) poisoning. Currently, no specific antidote is available. Synaptotagmin-1 (SYT1) has been identified as a key protein that facilitates PQ efflux in PQ-resistant A549 cells, thereby preventing PQ-induced lung injury. However, the protective effect of STY1 on PQ-induced AKI remains to be elucidated. This study exposed human kidney 2 (HK-2) cells overexpressing SYT1 to PQ. These cells exhibited significantly lower levels of growth inhibition, reactive oxygen species production, early apoptosis, and PQ accumulation compared to the parent HK-2 cells. Transcriptomic screening and Western blot analysis revealed that SYT1 overexpression significantly promoted the expression of glucose transporter 2 (GLUT2). Inhibition of GLUT2 completely abolished the protective effects of SYT1 overexpression in HK-2 cells and restored intracellular PQ concentrations. Further immunoprecipitation-shotgun and RNA interference experiments revealed that SYT1 binds to and stabilizes the protein SERPINE1 mRNA-binding protein 1 (SERBP1), enhancing the stability of GLUT2 mRNA and its protein levels. In summary, SYT1 antagonizes PQ intracellular accumulation and prevents nephrocyte toxicity by up-regulating SERBP1/GLUT2 expression. This study identifies a potential target for the treatment of PQ-induced AKI.
Collapse
Affiliation(s)
- Ran Yin
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingjing Ke
- Wenzhou Medical University, Wenzhou, 325000, China; Emergency Department, Taizhou First People's Hospital, Taizhou, 318020, China
| | - Mingming Zhao
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China
| | - Yitian Ding
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenwen Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Mengxuan Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China
| | - Lufeng Hu
- Pharmacy Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoqin Dai
- Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Guangliang Hong
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
44
|
Camacho J, Bernal-Rivera A, Peña V, Morales-Sosa P, Robb SMC, Russell J, Yi K, Wang Y, Tsuchiya D, Murillo-García OE, Rohner N. Sugar assimilation underlying dietary evolution of Neotropical bats. Nat Ecol Evol 2024; 8:1735-1750. [PMID: 39198571 PMCID: PMC11383804 DOI: 10.1038/s41559-024-02485-7] [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: 07/06/2023] [Accepted: 06/27/2024] [Indexed: 09/01/2024]
Abstract
Dietary specializations in animals lead to adaptations in morphology, anatomy and physiology. Neotropical bats, with their high taxonomic and trophic diversity, offer a unique perspective on diet-driven evolutionary adaptations. Here we assess the metabolic response to different dietary sugars among wild-caught bats. We found that insectivorous bats had a pronounced metabolic response to trehalose, whereas bats with nectar and fruit-based diets showed significantly higher blood glucose levels in response to glucose and sucrose, reaching levels over 750 mg dl-1. The genomic analysis of 22 focal species and two outgroup species identified positive selection for the digestive enzyme trehalase in insect eaters, while sucrase-isomaltase showed selection in lineages with omnivorous and nectar diets. By examining anatomical and cellular features of the small intestine, we discovered that dietary sugar proportion strongly impacted numerous digestive traits, providing valuable insight into the physiological implications of molecular adaptations. Using hybridization chain reaction (HCR) RNA fluorescence in situ hybridization, we observed unusually high expression in the glucose transporter gene Slc2a2 in nectar bats, while fruit bats increased levels of Slc5a1 and Slc2a5. Overall, this study highlights the intricate interplay between molecular, morphological and physiological aspects of diet evolution, offering new insights into the mechanisms of dietary diversification and sugar assimilation in mammals.
Collapse
Affiliation(s)
- Jasmin Camacho
- Stowers Institute for Medical Research, Kansas City, MO, USA.
| | - Andrea Bernal-Rivera
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Grupo de Investigación en Ecología Animal, Departamento de Biología, Universidad del Valle, Cali, Colombia
| | - Valentina Peña
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Sofia M C Robb
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Yongfu Wang
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Dai Tsuchiya
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Oscar E Murillo-García
- Grupo de Investigación en Ecología Animal, Departamento de Biología, Universidad del Valle, Cali, Colombia.
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO, USA.
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
45
|
Gonzalez E, Lee MD, Tierney BT, Lipieta N, Flores P, Mishra M, Beckett L, Finkelstein A, Mo A, Walton P, Karouia F, Barker R, Jansen RJ, Green SJ, Weging S, Kelliher J, Singh NK, Bezdan D, Galazska J, Brereton NJB. Spaceflight alters host-gut microbiota interactions. NPJ Biofilms Microbiomes 2024; 10:71. [PMID: 39209868 PMCID: PMC11362537 DOI: 10.1038/s41522-024-00545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The ISS rodent habitat has provided crucial insights into the impact of spaceflight on mammals, inducing symptoms characteristic of liver disease, insulin resistance, osteopenia, and myopathy. Although these physiological responses can involve the microbiome on Earth, host-microbiota interactions during spaceflight are still being elucidated. We explore murine gut microbiota and host gene expression in the colon and liver after 29 and 56 days of spaceflight using multiomics. Metagenomics revealed significant changes in 44 microbiome species, including relative reductions in bile acid and butyrate metabolising bacteria like Extibacter muris and Dysosmobacter welbionis. Functional prediction indicate over-representation of fatty acid and bile acid metabolism, extracellular matrix interactions, and antibiotic resistance genes. Host gene expression described corresponding changes to bile acid and energy metabolism, and immune suppression. These changes imply that interactions at the host-gut microbiome interface contribute to spaceflight pathology and that these interactions might critically influence human health and long-duration spaceflight feasibility.
Collapse
Affiliation(s)
- E Gonzalez
- Microbiome Unit, Canadian Centre for Computational Genomics, Department of Human Genetics, McGill University, Montréal, Canada
- Centre for Microbiome Research, McGill University, Montréal, Canada
| | - M D Lee
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - B T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - N Lipieta
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - P Flores
- BioServe Space Technologies, University of Colorado Boulder, Boulder, CO, USA
| | - M Mishra
- Grossman School of Medicine, New York University, New York, USA
| | - L Beckett
- University of Nottingham, Nottingham, NG7 2RD, UK
| | - A Finkelstein
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - A Mo
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - P Walton
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - F Karouia
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Centre for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - R Barker
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Yuri GmbH, Wiesentalstr. 40, 88074, Meckenbeuren, Germany
- University of Wisconsin-Madison, Madison, WI, USA
| | - R J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics, Phenomics, and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
| | - S J Green
- Genomics and Microbiome Core Facility, Rush University Medical Centre, 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - S Weging
- Institute of Computer Science, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - J Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - N K Singh
- Department of Industrial Relations, Division of Occupational Safety and Health, Oakland, USA
| | - D Bezdan
- University of Wisconsin-Madison, Madison, WI, USA
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - J Galazska
- Space Biosciences Research Branch, NASA Ames Research Centre, Moffett Field, CA, USA
| | - N J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
46
|
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
|
47
|
Palmer CS, Perdios C, Abdel-Mohsen M, Mudd J, Datta PK, Maness NJ, Lehmicke G, Golden N, Hellmers L, Coyne C, Moore Green K, Midkiff C, Williams K, Tiburcio R, Fahlberg M, Boykin K, Kenway C, Russell-Lodrigue K, Birnbaum A, Bohm R, Blair R, Dufour JP, Fischer T, Saied AA, Rappaport J. Non-human primate model of long-COVID identifies immune associates of hyperglycemia. Nat Commun 2024; 15:6664. [PMID: 39164284 PMCID: PMC11335872 DOI: 10.1038/s41467-024-50339-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 07/08/2024] [Indexed: 08/22/2024] Open
Abstract
Hyperglycemia, and exacerbation of pre-existing deficits in glucose metabolism, are manifestations of the post-acute sequelae of SARS-CoV-2. Our understanding of metabolic decline after acute COVID-19 remains unclear due to the lack of animal models. Here, we report a non-human primate model of metabolic post-acute sequelae of SARS-CoV-2 using SARS-CoV-2 infected African green monkeys. Using this model, we identify a dysregulated blood chemokine signature during acute COVID-19 that correlates with elevated and persistent hyperglycemia four months post-infection. Hyperglycemia also correlates with liver glycogen levels, but there is no evidence of substantial long-term SARS-CoV-2 replication in the liver and pancreas. Finally, we report a favorable glycemic effect of the SARS-CoV-2 mRNA vaccine, administered on day 4 post-infection. Together, these data suggest that the African green monkey model exhibits important similarities to humans and can be utilized to assess therapeutic candidates to combat COVID-related metabolic defects.
Collapse
Affiliation(s)
- Clovis S Palmer
- Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| | - Chrysostomos Perdios
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Joseph Mudd
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Prasun K Datta
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Linh Hellmers
- Tulane National Primate Research Center, Covington, LA, USA
| | - Carol Coyne
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Cecily Midkiff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Rafael Tiburcio
- Division of Experimental Medicine, Department of Medicine, University of San Francisco, CA, USA
| | | | - Kyndal Boykin
- Tulane National Primate Research Center, Covington, LA, USA
| | - Carys Kenway
- Tulane National Primate Research Center, Covington, LA, USA
| | - Kasi Russell-Lodrigue
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Rudolf Bohm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Robert Blair
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jason P Dufour
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tracy Fischer
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ahmad A Saied
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
48
|
Gautier-Stein A, Vily-Petit J, Rajas F, Mithieux G. Intestinal gluconeogenesis: A translator of nutritional information needed for glycemic and emotional balance. Biochimie 2024; 223:206-214. [PMID: 38040189 DOI: 10.1016/j.biochi.2023.11.012] [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: 07/25/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
At the interface between the outside world and the self, the intestine is the first organ receiving nutritional information. One intestinal function, gluconeogenesis, is activated by various nutrients, particularly diets enriched in fiber or protein, and thus results in glucose production in the portal vein in the post-absorptive period. The detection of portal glucose induces a nervous signal controlling the activity of the central nuclei involved in the regulation of metabolism and emotional behavior. Induction of intestinal gluconeogenesis is necessary for the beneficial effects of fiber or protein-enriched diets on metabolism and emotional behavior. Through its ability to translate nutritional information from the diet to the brain's regulatory centers, intestinal gluconeogenesis plays an essential role in maintaining physiological balance.
Collapse
Affiliation(s)
- Amandine Gautier-Stein
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France.
| | - Justine Vily-Petit
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| | - Fabienne Rajas
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| | - Gilles Mithieux
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| |
Collapse
|
49
|
Rossi A, Simeoli C, Pivonello R, Salerno M, Rosano C, Brunetti B, Strisciuglio P, Colao A, Parenti G, Melis D, Derks TGJ. Endocrine involvement in hepatic glycogen storage diseases: pathophysiology and implications for care. Rev Endocr Metab Disord 2024; 25:707-725. [PMID: 38556561 PMCID: PMC11294274 DOI: 10.1007/s11154-024-09880-2] [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] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Hepatic glycogen storage diseases constitute a group of disorders due to defects in the enzymes and transporters involved in glycogen breakdown and synthesis in the liver. Although hypoglycemia and hepatomegaly are the primary manifestations of (most of) hepatic GSDs, involvement of the endocrine system has been reported at multiple levels in individuals with hepatic GSDs. While some endocrine abnormalities (e.g., hypothalamic‑pituitary axis dysfunction in GSD I) can be direct consequence of the genetic defect itself, others (e.g., osteopenia in GSD Ib, insulin-resistance in GSD I and GSD III) may be triggered by the (dietary/medical) treatment. Being aware of the endocrine abnormalities occurring in hepatic GSDs is essential (1) to provide optimized medical care to this group of individuals and (2) to drive research aiming at understanding the disease pathophysiology. In this review, a thorough description of the endocrine manifestations in individuals with hepatic GSDs is presented, including pathophysiological and clinical implications.
Collapse
Affiliation(s)
- Alessandro Rossi
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy.
| | - Chiara Simeoli
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia ed Andrologia, University of Naples "Federico II", Naples, Italy
| | - Rosario Pivonello
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia ed Andrologia, University of Naples "Federico II", Naples, Italy
| | - Mariacarolina Salerno
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Carmen Rosano
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Barbara Brunetti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Section of Pediatrics, University of Salerno, Baronissi, Italy
| | - Pietro Strisciuglio
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Annamaria Colao
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia ed Andrologia, University of Naples "Federico II", Naples, Italy
| | - Giancarlo Parenti
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Section of Pediatrics, University of Salerno, Baronissi, Italy
| | - Terry G J Derks
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
50
|
Caspi I, Tremmel DM, Pulecio J, Yang D, Liu D, Yan J, Odorico JS, Huangfu D. Glucose Transporters Are Key Components of the Human Glucostat. Diabetes 2024; 73:1336-1351. [PMID: 38775784 PMCID: PMC11262048 DOI: 10.2337/db23-0508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 04/16/2024] [Indexed: 07/21/2024]
Abstract
Mouse models are extensively used in metabolic studies. However, inherent differences between the species, notably their blood glucose levels, hampered data translation into clinical settings. In this study, we confirmed GLUT1 to be the predominantly expressed glucose transporter in both adult and fetal human β-cells. In comparison, GLUT2 is detected in a small yet significant subpopulation of adult β-cells and is expressed to a greater extent in fetal β-cells. Notably, GLUT1/2 expression in INS+ cells from human stem cell-derived islet-like clusters (SC-islets) exhibited a closer resemblance to that observed in fetal islets. Transplantation of primary human islets or SC-islets, but not murine islets, lowered murine blood glucose to the human glycemic range, emphasizing the critical role of β-cells in establishing species-specific glycemia. We further demonstrate the functional requirements of GLUT1 and GLUT2 in glucose uptake and insulin secretion through chemically inhibiting GLUT1 in primary islets and SC-islets and genetically disrupting GLUT2 in SC-islets. Finally, we developed a mathematical model to predict changes in glucose uptake and insulin secretion as a function of GLUT1/2 expression. Collectively, our findings illustrate the crucial roles of GLUTs in human β-cells, and identify them as key components in establishing species-specific glycemic set points. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Inbal Caspi
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY
- Developmental Biology Program, Sloan Kettering Institute, New York, NY
| | - Daniel M. Tremmel
- Transplantation Division, Department of Surgery, University of Wisconsin-Madison, Madison, WI
| | - Julian Pulecio
- Developmental Biology Program, Sloan Kettering Institute, New York, NY
| | - Dapeng Yang
- Developmental Biology Program, Sloan Kettering Institute, New York, NY
| | - Dingyu Liu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jielin Yan
- Developmental Biology Program, Sloan Kettering Institute, New York, NY
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jon S. Odorico
- Transplantation Division, Department of Surgery, University of Wisconsin-Madison, Madison, WI
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY
| |
Collapse
|