1
|
Li H, Zhuang P, Liu X, Li Y, Ao Y, Tian Y, Jia W, Zhang Y, Jiao J. Marine N-3 Fatty Acids Mitigate Hyperglycemia in Prediabetes by Improving Muscular Glucose Transporter 4 Translocation and Glucose Homeostasis. RESEARCH (WASHINGTON, D.C.) 2025; 8:0683. [PMID: 40302785 PMCID: PMC12038161 DOI: 10.34133/research.0683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025]
Abstract
Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) have been proposed to benefit cardiometabolic health. However, the relationship between the intake of DHA and EPA and type 2 diabetes (T2D) risk remains equivocal, and the effects of DHA and EPA on skeletal muscle, the primary organ for glucose metabolism, merit further investigation. Here, we show that habitual fish oil supplementation was associated with a 9% lower T2D risk and significantly interacted with variants at GLUT4 in a prospective cohort of 48,358 people with prediabetes. Muscular metabolome analysis in the animal study revealed that DHA and EPA altered branched-chain amino acids, creatine, and glucose oxidation-related metabolites, concurrently with elevated muscular glycogen synthase and pyruvate dehydrogenase contents that promoted glucose disposal. Further myotube investigation revealed that DHA and EPA promoted muscular GLUT4 translocation by elevating Rab GTPases and target-SNARE expression. Together, DHA and EPA supplementation provides a promising approach for T2D prevention through targeting muscular glucose homeostasis, including enhancing GLUT4 translocation, glycogen synthesis, and aerobic glycolysis.
Collapse
Affiliation(s)
- Haoyu Li
- Department of Endocrinology, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory of Agri-food Reources and High-value Utilization, College of Biosystems Engineering and Food Science,
Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Xiaohui Liu
- Department of Endocrinology, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
- Department of Nutrition, School of Public Health,
Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yin Li
- Department of Nutrition, School of Public Health,
Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yang Ao
- Department of Nutrition, School of Public Health,
Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yimei Tian
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory of Agri-food Reources and High-value Utilization, College of Biosystems Engineering and Food Science,
Zhejiang University, Hangzhou 310058, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Wei Jia
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory of Agri-food Reources and High-value Utilization, College of Biosystems Engineering and Food Science,
Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory of Agri-food Reources and High-value Utilization, College of Biosystems Engineering and Food Science,
Zhejiang University, Hangzhou 310058, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
- Department of Nutrition, School of Public Health,
Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
2
|
Tang K, Tang Z, Niu M, Kuang Z, Xue W, Wang X, Liu X, Yu Y, Jeong S, Ma Y, Wu A, Kim BYS, Jiang W, Yang Z, Li C. Allosteric targeted drug delivery for enhanced blood-brain barrier penetration via mimicking transmembrane domain interactions. Nat Commun 2025; 16:3410. [PMID: 40210849 PMCID: PMC11986143 DOI: 10.1038/s41467-025-58746-x] [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: 07/01/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Current strategies for active targeting in the brain are entirely based on the effective interaction of the ligand with the orthosteric sites of specific receptors on the blood-brain barrier (BBB), which is highly susceptible to various pathophysiological factors and limits the efficacy of drug delivery. Here, we propose an allosteric targeted drug delivery strategy that targets classical BBB transmembrane receptors by designing peptide ligands that specifically bind to their transmembrane domains. This strategy prevents competitive interference from endogenous ligands and antibodies by using the insulin receptor and integrin αv as model targets, respectively, and can effectively overcome pseudotargets or target loss caused by shedding or mutating the extracellular domain of target receptors. Moreover, these ligands can be spontaneously embedded in the phospholipid layer of lipid carriers using a plug-and-play approach without chemical modification, with excellent tunability and immunocompatibility. Overall, this allosteric targeted drug delivery strategy can be applied to multiple receptor targets and drug carriers and offers promising therapeutic benefits in brain diseases.
Collapse
Affiliation(s)
- Kaicheng Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Miaomiao Niu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zuyin Kuang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xinyu Wang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Yang Yu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Seongdong Jeong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Annette Wu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China.
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Li H, Xiao F, Zhou C, Zhu T, Wang S. Metabolic Adaptations and Therapies in Cardiac Hypoxia: Mechanisms and Clinical Implications/ Potential Strategies. JACC Basic Transl Sci 2025:S2452-302X(24)00458-3. [PMID: 40265246 DOI: 10.1016/j.jacbts.2024.12.008] [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: 09/10/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 04/24/2025]
Abstract
Cardiac hypoxia triggers a cascade of responses and functional changes in myocardial and non-myocardial cells, profoundly affecting cellular metabolism, oxygen-sensing mechanisms, and immune responses. Myocardial cells, being the primary cell type in cardiac tissue, undergo significant alterations in energy metabolism, including glycolysis, fatty acid metabolism, ketone body utilization, and branched-chain amino acid metabolism, to maintain cardiac function under hypoxic conditions. Non-myocardial cells, such as fibroblasts, endothelial cells, and immune cells, although fewer in number, play crucial roles in regulating cardiac homeostasis, maintaining structural integrity, and responding to injury. This review discusses the metabolic reprogramming of immune cells, particularly macrophages, during ischemia-reperfusion injury and explores various therapeutic strategies that modulate these metabolic pathways to protect the heart during hypoxia. Understanding these interactions provides valuable insights and potential therapeutic targets for heart disease treatment.
Collapse
Affiliation(s)
- Huili Li
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fei Xiao
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenghui Zhou
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012, West China Hospital, Sichuan University, Chengdu, China.
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Linzhi People's Hospital, Linzhi, Tibet, China.
| |
Collapse
|
4
|
Zhang D, Wang Y, Shi Y, Panya A, Pandith H, Inta A, Yang X. Identified prenylated polyphenols from Mesua ferrea L. and their stimulatory effect on glucose uptake in 3 T3-L1 adipocytes. Fitoterapia 2025; 182:106468. [PMID: 40054702 DOI: 10.1016/j.fitote.2025.106468] [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: 08/12/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Mesua ferrea L. has traditionally utilized in folk medicine for its antidiabetic properties, and contemporary pharmacological studies have confirmed its hypoglycemic activity. While, the specific components responsible for these effects have not yet been fully elucidated. In this study, we employed a bioactivity-guided fractionation approach to isolate 22 prenylated polyphenols from M. ferrea leaves, including two novel 4-phenylcoumarins, mesuol A (1) and mesuol B (2), along with 20 previously identified compounds. The majority of these compounds, including 13 4-phenylcoumarins and two xanthones, exhibited significant stimulatory effect on glucose uptake in 3 T3-L1 adipocytes. Notably, at a concentration of 2 μM, isomesuol (14), disparinol D (17), and isodisparinol A (19) exhibited glucose uptake stimulatory effect that were either superior or equivalent to that of insulin (positive control). The structure-activity relationship analysis revealed that cyclization of 4-phenylcoumarins to form a furan ring markedly diminished their glucose uptake stimulatory effects, thereby reducing their hypoglycemic potential. In contrast, non-cyclized and pyran ring-cyclized 4-phenylcoumarins demonstrated stronger glucose uptake-stimulatory activities. These findings highlight the non-cyclized and pyran ring-cyclized 4-phenylcoumarins as promising leads for the development of anti-diabetic agents, with M. ferrea leaves serving as a valuable source of these bioactive compounds.
Collapse
Affiliation(s)
- Dongdong Zhang
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Nay Pyi Taw 05282, Myanmar; Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Yuehu Wang
- Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Yinxian Shi
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Nay Pyi Taw 05282, Myanmar; Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Aussara Panya
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Hataichanok Pandith
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Angkhana Inta
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Xuefei Yang
- Southeast Asia Biodiversity Research Institute, Chinese Academy of Sciences, Nay Pyi Taw 05282, Myanmar; Key Laboratory of Economic Plants and Biotechnology, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; Yunnan International Joint Laboratory of Southeast Asia Biodiversity Conservation, Xishuangbanna 666303, China.
| |
Collapse
|
5
|
Venkatesh D, Sarkar S, Kandasamy T, Ghosh SS. In-silico identification and validation of Silibinin as a dual inhibitor for ENO1 and GLUT4 to curtail EMT signaling and TNBC progression. Comput Biol Chem 2025; 115:108312. [PMID: 39689434 DOI: 10.1016/j.compbiolchem.2024.108312] [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: 07/20/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
The aberrant metabolic reprogramming endows TNBC cells with sufficient ATP and lactate required for survival and metastasis. Hence, the intervention of the metabolic network represents a promising avenue to alleviate the Warburg effect in TNBC cells to impair their invasive and metastatic potential. Multitudinous in-silico analysis identified Enolase1 (ENO1) and the surface transporter protein, GLUT4 to be the potential targets for the abrogation of the metabolic network. The expression profiles of ENO1 and GLUT4 genes showed anomalous expression in various cancers, including breast cancer. Subsequently, the functional and physiological interactions of the target proteins were analyzed from the protein-protein interaction network. The pathway enrichment analysis identified the prime cancer signaling pathways in which these proteins are involved. Further, docking results bestowed Silibinin as the concurrent inhibitor of ENO1 and GLUT4. Moreover, the stable interaction of Silibinin with both proteins deciphered the binding free energies values of -48.86 and -104.31 KJ/mol from MMPBSA analysis and MD simulation, respectively. Furthermore, the cell viability, ROS assay, and live-dead imaging underscored the pronounced cytotoxicity of Silibinin, illuminating its capacity to incur apoptosis within TNBC cells. Additionally, glycolysis assay and gene expression analysis demonstrated the silibinin-mediated inhibition of the glycolysis pathway. Eventually, a lipidomic reprogramming towards fatty acid metabolism was established from the elevated lipid droplet accumulation, exogenous fatty acid uptake and de-novo lipogenesis. Nevertheless, repression of EMT and Wnt pathway progression by Silibinin was perceived from the gene expression studies. Overall, the current study highlights the tweaking of intricate signaling crosstalk between glycolysis and the Wnt pathway in TNBC cells through inhibiting ENO1 and GLUT4.
Collapse
Affiliation(s)
- Dheepika Venkatesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Shilpi Sarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India.
| |
Collapse
|
6
|
Khamis MM, Moselhy SS, Rihan S. Role of trans-resveratrol in ameliorating biochemical and molecular alterations in obese rats induced by a high fructose/fat diet. Sci Rep 2025; 15:7879. [PMID: 40050385 PMCID: PMC11885455 DOI: 10.1038/s41598-025-91027-7] [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: 10/01/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
We evaluated the effect of trans-resveratrol (RSV) in ameliorating biochemical and molecular alterations in obese Wister male rats fed on high-fat/high-fructose-fed. Male Wister rats were divided into eight groups and fed with either a standard diet (control), high fructose (HF), high fat (HFAT), or a high- fructose high- fat (HF/HFAT) diet and supplemented with RSV (30 mg/kg/day) for 4 weeks. The food intake, body weight, glycemic parameters, lipid profile, oxidative stress were assessed. SIRT1 gene expression, PGC-1α, cyto-c and GLUT-4 were evaluated by qRT-PCR in adipose tissue of normal and obese rats. The body weight gain, serum fasting glucose, insulin, and HOMA-IR values were significantly higher in the HF and HF/HFAT groups than in the HFAT and control groups. Hyperlipidemia was observed in high calorie diets fed rats compared to control group. The levels of total cholesterol, triglycerides and LDL-c were significantly elevated while HDL- c was significantly decreased in HF & HF/HFAT groups compared to HFAT group. The levels of serum malondialdhyde (MDA) and superoxide dismutase (SOD) activity in adipose tissue were elevated in all groups compared to control group, particularly in the groups that were kept on a high fructose diets (HF, HF/HFAT). SIRT-1, PGC-1α, Cyto-c, and GLUT-4 genes levels were significantly down regulated in HF, HFAT & HF/HFAT groups compared to control group. Supplementation of T-RSV restored the alteration in carbohydrates-lipid metabolism as well as oxidative stress and upregulation of SIRT-1, PGC-1α, Cyto-c, and GLUT-4 genes. RSV is a promising treatment in the management of pathologic consequences of obesity from high-calorie diet consumption via molecular alteration of target genes.
Collapse
Affiliation(s)
- Marwa Maher Khamis
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Said Salama Moselhy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Shaimaa Rihan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
7
|
Lee WD, Weilandt DR, Liang L, MacArthur MR, Jaiswal N, Ong O, Mann CG, Chu Q, Hunter CJ, Ryseck RP, Lu W, Oschmann AM, Cowan AJ, TeSlaa TA, Bartman CR, Jang C, Baur JA, Titchenell PM, Rabinowitz JD. Lactate homeostasis is maintained through regulation of glycolysis and lipolysis. Cell Metab 2025; 37:758-771.e8. [PMID: 39889702 PMCID: PMC11926601 DOI: 10.1016/j.cmet.2024.12.009] [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: 04/30/2024] [Revised: 09/19/2024] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
Lactate is among the highest flux circulating metabolites. It is made by glycolysis and cleared by both tricarboxylic acid (TCA) cycle oxidation and gluconeogenesis. Severe lactate elevations are life-threatening, and modest elevations predict future diabetes. How lactate homeostasis is maintained, however, remains poorly understood. Here, we identify, in mice, homeostatic circuits regulating lactate production and consumption. Insulin induces lactate production by upregulating glycolysis. We find that hyperlactatemia inhibits insulin-induced glycolysis, thereby suppressing excess lactate production. Unexpectedly, insulin also promotes lactate TCA cycle oxidation. The mechanism involves lowering circulating fatty acids, which compete with lactate for mitochondrial oxidation. Similarly, lactate can promote its own consumption by lowering circulating fatty acids via the adipocyte-expressed G-protein-coupled receptor hydroxycarboxylic acid receptor 1 (HCAR1). Quantitative modeling suggests that these mechanisms suffice to produce lactate homeostasis, with robustness to noise and perturbation of individual regulatory mechanisms. Thus, through regulation of glycolysis and lipolysis, lactate homeostasis is maintained.
Collapse
Affiliation(s)
- Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Daniel R Weilandt
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Lingfan Liang
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Natasha Jaiswal
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Olivia Ong
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Charlotte G Mann
- Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Qingwei Chu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig J Hunter
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Rolf-Peter Ryseck
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Anna M Oschmann
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Alexis J Cowan
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Tara A TeSlaa
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Caroline R Bartman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
8
|
Quan C, Jiang X. The molecular mechanism underlying the human glucose facilitators inhibition. VITAMINS AND HORMONES 2025; 128:49-92. [PMID: 40097253 DOI: 10.1016/bs.vh.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Glucose is the primary energy substrate and an essential precursor for cellular metabolism. Maintaining glucose homeostasis necessitates the presence of glucose transporters, as the hydrophilic nature of glucose prevents its passage across the cell membrane. The GLUT family is a crucial group of glucose transporters that facilitate glucose diffusion along the transmembrane glucose concentration gradient. Dysfunction in GLUTs is associated with diseases, such as GLUT1 deficiency syndrome, Fanconi-Bickel syndrome, and type 2 diabetes. Furthermore, elevated expression of GLUTs fuels aerobic glycolysis, known as the Warburg effect, in various types of cancers, making GLUT isoforms possible targets for antineoplastic therapies. To date, 30 GLUT and homolog structures have been released on the Protein Data Bank (PDB), showcasing multiple conformational and ligand-binding states. These structures elucidate the molecular mechanisms underlying substrate recognition, the alternating access cycle, and transport inhibition. Here, we summarize the current knowledge of human GLUTs and their role in cancer, highlighting recent advances in the structural characterization of GLUTs. We also compare the inhibition mechanisms of exofacial and endofacial GLUT inhibitors, providing insights into the design and optimization of GLUT inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Cantao Quan
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China
| | - Xin Jiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China.
| |
Collapse
|
9
|
Yin Y, Nie W, Tang ZQ, Zhu SJ. Flavonoid-Rich Extracts from Chuju ( Asteraceae Chrysanthemum L.) Alleviate the Disturbance of Glycolipid Metabolism on Type 2 Diabetic Mice via Modulating the Gut Microbiota. Foods 2025; 14:765. [PMID: 40077469 PMCID: PMC11898795 DOI: 10.3390/foods14050765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) and its associated complications represent a significant public health issue affecting hundreds of millions of people globally; thus, measures to prevent T2DM are urgently needed. Chuju has been proven to possess antihyperglycemic activity. However, the bioactive ingredients in chuju that contribute to its antihyperglycemic activity, as well as the relationship between its antihyperglycemic activity and the gut microbiota, remain unclear. To understand the potential effects that it has on T2DM, the glycolipid metabolism and gut microbiota regulation of flavonoid-rich extracts from chuju (CJE) were investigated. The results showed that the top ten flavonoid compounds in CJE are Apigenin 6, 8-digalactoside, Apigenin 6-C-glucoside 8-C-arabinoside, Luteolin-4'-O-glucoside, Isoshaftoside, Scutellarin, Quercetin 3-O-malonylglucoside, Chrysoeriol 7-O-glucoside, Quercetin-3,4'-O-di-beta-glucoside, Luteolin 6-C-glucoside 8-C-arabinoside, and Homoorientin. Furthermore, CJE mitigated hyperglycemia and glycolipid metabolism by reducing the abundance of Faecalibaculum, Coriobacteriaceae, and Romboutsia and increasing the abundance of Alistipes. In addition, the results of Western blot analysis showed that CJE could enhance glycogen synthesis and glucose transport by up-regulating the phosphorylation of IRS1-PI3K-Akt and AMPK-GLUT4. Simultaneously, CJE could decrease gluconeogenesis by down-regulating the phosphorylation of FoxO1/GSK 3β. In conclusion, the findings of this study provide new evidence supporting the hypothesis that CJE can be used as part of a therapeutic approach for treating disturbances in glycolipid metabolism via regulating the gut microbiota and mediating the IRS1-PI3K-Akt-FoxO1/GSK 3β and AMPK-GLUT4 pathways.
Collapse
Affiliation(s)
- Yu Yin
- School of Life Sciences, Anhui University, Hefei 230601, China;
- School of Biological and Food Engineering, Chuzhou University, Chuzhou 239001, China;
| | - Wen Nie
- School of Biological and Food Engineering, Chuzhou University, Chuzhou 239001, China;
| | - Zheng-Quan Tang
- School of Life Sciences, Anhui University, Hefei 230601, China;
| | - Shuang-Jie Zhu
- School of Life Sciences, Anhui University, Hefei 230601, China;
- School of Biological and Food Engineering, Chuzhou University, Chuzhou 239001, China;
| |
Collapse
|
10
|
Chang YC, Jusko WJ. Comparing the Efficacy of Various Insulin Types: Pharmacokinetic and Pharmacodynamic Modeling of Glucose Clamp Effects in Healthy Volunteers. J Clin Pharmacol 2025. [PMID: 39982761 DOI: 10.1002/jcph.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/03/2025] [Indexed: 02/22/2025]
Abstract
This study compares the pharmacokinetics and efficacy of various subcutaneously (SC) dosed insulin analogs, including rapid-acting, intermediate-acting, long-acting, and regular human insulin, using mechanistic pharmacokinetic (PK) and pharmacodynamic (PD) models. These models were applied to data from euglycemic clamp studies in healthy volunteers, where insulin pharmacokinetics and its effects on glucose utilization were monitored. Data from published studies were digitized and modeled using MONOLIX (Version 2024). The PK model described insulin absorption via sequential first-order processes and linear elimination. The PD effects were captured using a model combination of biophase, indirect, and receptor down-regulation components. While PK parameters-especially absorption rates-varied between insulin types, a common set of nonlinear PD parameters was sought to account for dose-related differences in glucose utilization. The maximum glucose stimulation (S max ${{{\mathrm{S}}}_{{\mathrm{max}}}}$ ) was 163, and the insulin concentration for a half-maximal effect (S C 50 ${\mathrm{S}}{{{\mathrm{C}}}_{50}}$ ) were 1156 pmol/L for insulin lispro, regular human insulin, neutral protamine hagedorn (NPH) insulin, and insulin glargine; 674 pmol/L for insulin aspart; and 5335 pmol/L for insulin detemir. Insulin detemir showed similar overt effects as the other insulin types but with smaller clearances and lower potency. This mechanism-based glucose-insulin model demonstrated that most insulin analogs exhibit similar receptor- and transporter-related parameters. The model, with specific PK but unified PD parameters, may enable clinical optimization of insulin therapy by highlighting differences in pharmacokinetics and operating common intrinsic glucose utilization parameters.
Collapse
Affiliation(s)
- Yi Chien Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
11
|
Urbaniak E, Henry S, Lalowski M, Borowiak M. Molecular puzzle of insulin: structural assembly pathways and their role in diabetes. Front Cell Dev Biol 2025; 13:1502469. [PMID: 40052150 PMCID: PMC11882602 DOI: 10.3389/fcell.2025.1502469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Properly folded proteins are essential for virtually all cellular processes including enzyme catalysis, signal transduction, and structural support. The cells have evolved intricate mechanisms of control, such as the assistance of chaperones and proteostasis networks, to ensure that proteins mature and fold correctly and maintain their functional conformations. Here, we review the mechanisms governing the folding of key hormonal regulators or glucose homeostasis. The insulin synthesis in pancreatic β-cells begins with preproinsulin production. During translation, the insulin precursor involves components of the endoplasmic reticulum (ER) translocation machinery, which are essential for proper orientation, translocation, and cleavage of the signal peptide of preproinsulin. These steps are critical to initiate the correct folding of proinsulin. Proinsulin foldability is optimized in the ER, an environment evolved to support the folding process and the formation of disulfide bonds while minimizing misfolding. This environment is intricately linked to ER stress response pathways, which have both beneficial and potentially harmful effects on pancreatic β-cells. Proinsulin misfolding can result from excessive biosynthetic ER load, proinsulin gene mutations, or genetic predispositions affecting the ER folding environment. Misfolded proinsulin leads to deficient insulin production and contributes to diabetes pathogenesis. Understanding the mechanisms of protein folding is critical for addressing diabetes and other protein misfolding-related diseases.
Collapse
Affiliation(s)
- Edyta Urbaniak
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Sara Henry
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Maciej Lalowski
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
- Meilahti Clinical Proteomics Core Facility, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
- Center for Cell and Gene Therapy, Stem Cell and Regenerative Medicine Center, Baylor College of Medicine, Texas Children’s Hospital, Methodist Hospital, Houston, TX, United States
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
12
|
Hu C, Chen Y, Yin X, Xu R, Yin C, Wang C, Zhao Y. Pancreatic endocrine and exocrine signaling and crosstalk in physiological and pathological status. Signal Transduct Target Ther 2025; 10:39. [PMID: 39948335 PMCID: PMC11825823 DOI: 10.1038/s41392-024-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/20/2024] [Accepted: 12/03/2024] [Indexed: 02/16/2025] Open
Abstract
The pancreas, an organ with dual functions, regulates blood glucose levels through the endocrine system by secreting hormones such as insulin and glucagon. It also aids digestion through the exocrine system by secreting digestive enzymes. Complex interactions and signaling mechanisms between the endocrine and exocrine functions of the pancreas play a crucial role in maintaining metabolic homeostasis and overall health. Compelling evidence indicates direct and indirect crosstalk between the endocrine and exocrine parts, influencing the development of diseases affecting both. From a developmental perspective, the exocrine and endocrine parts share the same origin-the "tip-trunk" domain. In certain circumstances, pancreatic exocrine cells may transdifferentiate into endocrine-like cells, such as insulin-secreting cells. Additionally, several pancreatic diseases, including pancreatic cancer, pancreatitis, and diabetes, exhibit potential relevance to both endocrine and exocrine functions. Endocrine cells may communicate with exocrine cells directly through cytokines or indirectly by regulating the immune microenvironment. This crosstalk affects the onset and progression of these diseases. This review summarizes the history and milestones of findings related to the exocrine and endocrine pancreas, their embryonic development, phenotypic transformations, signaling roles in health and disease, the endocrine-exocrine crosstalk from the perspective of diseases, and potential therapeutic targets. Elucidating the regulatory mechanisms of pancreatic endocrine and exocrine signaling and provide novel insights for the understanding and treatment of diseases.
Collapse
Grants
- National High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
- cNational High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
Collapse
Affiliation(s)
- Chenglin Hu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chenxue Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chengcheng Wang
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
- Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
13
|
Lee K, Kim M. Evolutionary Insights into Irisin/FNDC5: Roles in Aging and Disease from Drosophila to Mammals. Biomolecules 2025; 15:261. [PMID: 40001564 PMCID: PMC11853655 DOI: 10.3390/biom15020261] [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: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The Irisin/FNDC5 protein family has emerged as a pivotal link between exercise and the prevention of age-associated diseases. Irisin is highly expressed during exercise from skeletal and cardiac muscle cells, playing a critical role in mediating systemic health benefits through its actions on various tissues. However, Irisin levels decline with age, correlating with a heightened incidence of diseases such as muscle weakness, cardiovascular disorders, and neurodegeneration. Notably, the administration of Irisin has shown significant potential in both preventing and treating these conditions. Recently, an Irisin/FNDC5 homolog was identified in an invertebrate Drosophila model, providing valuable insights into its conserved role in exercise physiology. Importantly, Irisin/FNDC5 has been demonstrated to regulate autophagy-a process essential for clearing excessive nutrients, toxic aggregates, and dysfunctional organelles-in both flies and mammals. Dysregulated autophagy is often implicated in age-related diseases, highlighting its relevance to Irisin/FNDC5's functions. These findings deepen our understanding of Irisin/FNDC5's roles and its potential as a therapeutic target for mitigating aging-related health decline. Further studies are needed to elucidate the precise mechanisms by which Irisin regulates autophagy and its broader impact on physiological aging and related diseases.
Collapse
Affiliation(s)
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
14
|
Choi E, Duan C, Bai XC. Regulation and function of insulin and insulin-like growth factor receptor signalling. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00826-3. [PMID: 39930003 DOI: 10.1038/s41580-025-00826-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 03/24/2025]
Abstract
Receptors of insulin and insulin-like growth factors (IGFs) are receptor tyrosine kinases whose signalling controls multiple aspects of animal physiology throughout life. In addition to regulating metabolism and growth, insulin-IGF receptor signalling has recently been linked to a variety of new, cell type-specific functions. In the last century, key questions have focused on how structural differences of insulin and IGFs affect receptor activation, and how insulin-IGF receptor signalling translates into pleiotropic biological functions. Technological advances such as cryo-electron microscopy have provided a detailed understanding of how native and engineered ligands activate insulin-IGF receptors. In this Review, we highlight recent structural and functional insights into the activation of insulin-IGF receptors, and summarize new agonists and antagonists developed for intervening in the activation of insulin-IGF receptor signalling. Furthermore, we discuss recently identified regulatory mechanisms beyond ligand-receptor interactions and functions of insulin-IGF receptor signalling in diseases.
Collapse
Affiliation(s)
- Eunhee Choi
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| | - Cunming Duan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| | - Xiao-Chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
15
|
Lv S, Zhu Z, Xiao H. Flavonoids and their metal complexes as potential agents for diabetes mellitus with future perspectives. Crit Rev Food Sci Nutr 2025:1-31. [PMID: 39902771 DOI: 10.1080/10408398.2025.2461238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a global health burden, with hyperglycemia as the main hallmark. This review commences with a concise overview of the intricate mechanisms underlying glucose uptake and utilization in organisms. Notably, we emphasize that T2DM management strategies pivot on delaying carbohydrate digestion, augmenting insulin secretion, and enhancing insulin sensitivity in target tissues. Unfortunately, the drugs currently available in the market for the treatment of T2DM have unpleasant side effects, spurring an urgent quest for safer and more efficacious alternatives. Flavonoids, emerging as a promising class of bioactive compounds derived from plants, offer a multi-faceted approach to diabetes treatment. Specifically, they potently inhibit enzymes such as α-amylase, α-glucosidase, dipeptidyl peptidase-4 (DPP-4), glycogen phosphorylase (GP) and protein-tyrosine phosphatase-1B (PTP1B). Through an in-depth analysis, this review not only summarizes these inhibitory actions but also establishes the structure-activity relationship (SAR), providing a blueprint for rational drug design. However, the clinical translation of flavonoids has been hampered by their suboptimal water solubility and bioavailability, attributable to the characteristic carbonyl and hydroxyl groups. Ingeniously, this chemical quirk has been harnessed to engineer metal chelates, which exhibit enhanced pharmacokinetic profiles. Herein, we offer an exhaustive overview of the latest advancements in flavonoid metal complexes research, spotlighting their potential as next-generation diabetes therapeutics. Available data are poised to galvanize the development of novel flavonoid derivatives, be it as potent drugs or functional foods, for combating T2DM.
Collapse
Affiliation(s)
- Shuang Lv
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Zhenbao Zhu
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, USA
| |
Collapse
|
16
|
Kobayashi H, Matsubara S, Yoshimoto C, Shigetomi H, Imanaka S. A Comprehensive Review of the Contribution of Mitochondrial DNA Mutations and Dysfunction in Polycystic Ovary Syndrome, Supported by Secondary Database Analysis. Int J Mol Sci 2025; 26:1172. [PMID: 39940939 PMCID: PMC11818232 DOI: 10.3390/ijms26031172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder affecting women of reproductive age characterized by a spectrum of clinical, metabolic, reproductive, and psychological abnormalities. This syndrome is associated with significant long-term health risks, necessitating elucidation of its pathophysiology, early diagnosis, and comprehensive management strategies. Several contributory factors in PCOS, including androgen excess and insulin resistance, collectively enhance oxidative stress, which subsequently leads to mitochondrial dysfunction. However, the precise mechanisms through which oxidative stress induces mitochondrial dysfunction remain incompletely understood. Comprehensive searches of electronic databases were conducted to identify relevant studies published up to 30 September 2024. Mitochondria, the primary sites of reactive oxygen species (ROS) generation, play critical roles in energy metabolism and cellular homeostasis. Oxidative stress can inflict damage on components, including lipids, proteins, and DNA. Damage to mitochondrial DNA (mtDNA), which lacks efficient repair mechanisms, may result in mutations that impair mitochondrial function. Dysfunctional mitochondrial activity further amplifies ROS production, thereby perpetuating oxidative stress. These disruptions are implicated in the complications associated with the syndrome. Advances in genetic analysis technologies, including next-generation sequencing, have identified point mutations and deletions in mtDNA, drawing significant attention to their association with oxidative stress. Emerging data from mtDNA mutation analyses challenge conventional paradigms and provide new insights into the role of oxidative stress in mitochondrial dysfunction. We are rethinking the pathogenesis of PCOS based on these database analyses. In conclusion, this review explores the intricate relationship between oxidative stress, mtDNA mutations, and mitochondrial dysfunction, offers an updated perspective on the pathophysiology of PCOS, and outlines directions for future research.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara 634-0813, Japan;
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
- Department of Medicine, Kei Oushin Clinic, 5-2-6 Naruo-cho, Nishinomiya 663-8184, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, 2-897-5 Shichijyonishi-machi, Nara 630-8581, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, 3-3-17 Kitatomigaoka-cho, Nara 634-0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara 634-0813, Japan;
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
| |
Collapse
|
17
|
Zhao P, Zhong S, Liao J, Tao J, Yao Y, Song P, Yang X. Caragana jubata ethanol extract ameliorates the symptoms of STZ-HFD-induced T2DM mice by PKC/GLUT4 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119171. [PMID: 39613004 DOI: 10.1016/j.jep.2024.119171] [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: 08/03/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Caragana jubata (Pall.) Poir., a traditional Tibetan medicinal plant in China, is renowned in Tibetan medicine for its hypoglycemic properties and long-standing use in treating diabetes. Despite its extensive clinical use, the mechanisms underlying its blood sugar-lowering effects still need to be explored. Our investigation contributes a new understanding of the hypoglycemic mechanism of C. jubata, validating its traditional medicinal application by demonstrating its ability to increase GLUT4 expression and glucose uptake, crucial elements in treating type 2 diabetes mellitus (T2DM). AIM OF THE STUDY This study investigated the potential anti-diabetic effects of C. jubata ethanol extract (CJEE) by upregulating GLUT4 expression and promoting its integration into the plasma membrane in L6 skeletal muscle cells and diabetic mice. Additionally, the research aimed to uncover the mechanisms involved, particularly focusing on the involvement of the PKC signaling pathway and Ca2⁺ release. MATERIALS AND METHODS The chemical composition of CJEE was evaluated using UPLC-Q-TOF/MS. Glucose uptake, GLUT4 expression, and plasma membrane fusion in L6 cells were assessed through a glucose oxidase kit, Western blotting, and laser confocal microscopy, respectively. The modulation of GLUT4 by Akt, AMPK, and PKC signaling pathways was investigated utilizing specific inhibitors. The impact of CJEE on intracellular Ca2⁺ concentration was determined with Fluo-4 dye. Additionally, an in vivo study was conducted on high-fat diet (HFD) and streptozotocin (STZ)-induced type 2 diabetic mice to evaluate the effects of CJEE on blood glucose levels, insulin resistance, lipid metabolism, and pancreatic function. RESULTS Chemical analysis of CJEE revealed 18 major constituents, primarily flavonoids. In L6 cells, CJEE was found to significantly enhance glucose uptake, increase GLUT4 expression, and facilitate its fusion with the plasma membrane. The study illustrated that CJEE predominantly activates the PKC pathway, with minimal involvement of the Akt pathway, emphasizing the critical role of Ca2⁺ release in GLUT4 regulation. Diabetic mice treated with CJEE exhibited decreased fasting blood glucose levels, enhanced oral glucose tolerance, reduced insulin resistance, and ameliorated lipid metabolism disorders. Additionally, CJEE elevated GLUT4 expression in insulin-sensitive tissues and alleviated pancreatic and hepatic lesions. CONCLUSIONS Our results demonstrated that the activation of the PKC pathway and release of Ca2⁺ by CJEE induce GLUT4 expression, promoting its fusion with the plasma membrane. Consequently, this process boosts glucose uptake and enhances insulin sensitivity, underscoring CJEE as a promising option for managing T2DM.
Collapse
Affiliation(s)
- Ping Zhao
- South-Central Minzu University, 182 Min-Zu Road, Wuhan, 430074, China
| | - Shunhua Zhong
- South-Central Minzu University, 182 Min-Zu Road, Wuhan, 430074, China
| | - Jingya Liao
- South-Central Minzu University, 182 Min-Zu Road, Wuhan, 430074, China
| | - Jingze Tao
- South-Central Minzu University, 182 Min-Zu Road, Wuhan, 430074, China
| | - Yanhong Yao
- South-Central Minzu University, 182 Min-Zu Road, Wuhan, 430074, China
| | - Ping Song
- College of Chemistry and Chemical Engineering, Qinghai Nationalities University, Xining, 810007, China.
| | - Xinzhou Yang
- South-Central Minzu University, 182 Min-Zu Road, Wuhan, 430074, China.
| |
Collapse
|
18
|
Chen X, Yao H, Lai J, Chen Y, Li X, Li S, Li L, He F. Endothelial versus Metabolic Insulin Resistance, A Descriptive Review. Curr Diabetes Rev 2025; 21:94-105. [PMID: 39676508 DOI: 10.2174/0115733998288601240327065724] [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: 11/20/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 12/17/2024]
Abstract
Cardiovascular complications are a primary focus in the clinical management of type 2 diabetes, as they are the leading causes of disability and mortality in individuals with diabetes. Insulin resistance and endothelial dysfunction commonly coexist in diabetic patients. An increasing body of research indicates a reciprocal and interconnected association between endothelial function and insulin resistance. Insulin resistance can manifest in two distinct forms: endothelial and metabolic, with the former predominantly affecting vascular endothelial cells and the latter primarily impacting peripheral cells. The understanding of endothelial insulin resistance is crucial in comprehending the pathophysiology of cardiovascular complications in type 2 diabetes. Hence, the objective of this study is to examine the correlations, interplays, and molecular pathways linking endothelial insulin resistance and metabolic insulin resistance, with the aim of offering novel insights and scholarly resources for the prevention and management of diabetic vascular complications.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Quality Control, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Huajie Yao
- Department of Quality Control, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
- Department of Pharmacy, Wuhan Polytechnic University, College of Life Science and Technology, Wuhan, China
| | - Jiaqi Lai
- Department of Quality Control, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Yanmei Chen
- Department of Quality Control, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Xiaodong Li
- Department of Quality Control, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Shanshan Li
- Department of Quality Control, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Ling Li
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Fazhong He
- Department of Quality Control, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| |
Collapse
|
19
|
Chaudhary N, Kiranmayee B. Non-receptor Type PTPases and their Role in Controlling Pathways Related to Diabetes and Liver Cancer Signalling. Curr Pharm Biotechnol 2025; 26:654-664. [PMID: 38424416 DOI: 10.2174/0113892010288624240213072415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
The role of non-receptor type Protein Tyrosine Phosphatase (PTPases) in controlling pathways related to diabetes and Hepatocellular Carcinoma (HCC) is significant. The insulin signal transduction pathway is regulated by the steady-state phosphorylation of tyrosyl residues of the insulin receptor and post-receptor substrates. PTPase has been shown to have a physiological role in the regulation of reversible tyrosine phosphorylation. There are several non-receptor type PTPases. PTPase containing the SH-2 domain (SHP-2) and the non-receptor type PTPase (PTP1B; encoded by the PTPN1 gene) are involved in negative regulation of the insulin signaling pathway, thereby indicating that the pathway can be made more efficient by the reduction in the activity of specific PTPases. Reduction in insulin resistance may be achieved by drugs targeting these specific enzymes. The modifications in the receptor and post-receptor events of insulin signal transduction give rise to insulin resistance, and a link between insulin-resistant states and HCC has been established. The cancer cells thrive on higher levels of energy and their growth gets encouraged since insulin-resistant states lead to greater glucose levels. Cancer, hyperglycemia, and hypoglycemia are highly linked through various pathways hence, clarifying the molecular mechanisms through which non-receptor type PTPase regulates the insulin signal transduction is necessary to find an effective target for cancer. Targeting the pathways related to PTPases; both receptor and non-receptor types, may lead to an effective candidate to fight against diabetes and HCC.
Collapse
Affiliation(s)
- Nidhee Chaudhary
- Centre for Biotechnology & Biochemical Engineering, Amity Institute Biotechnology, Amity University Uttar Pradesh, Sector-125, Expressway, Noida, 201313, Uttar Pradesh, India
| | - Bellam Kiranmayee
- Centre for Biotechnology & Biochemical Engineering, Amity Institute Biotechnology, Amity University Uttar Pradesh, Sector-125, Expressway, Noida, 201313, Uttar Pradesh, India
| |
Collapse
|
20
|
Shi X, Hu X, Fang X, Jia L, Wei F, Peng Y, Liu M, Gao A, Zhao K, Chen F, Hu X, Hong J, Ning G, Song Y, Wang J, Wang Y. A feeding-induced myokine modulates glucose homeostasis. Nat Metab 2025; 7:68-83. [PMID: 39747483 DOI: 10.1038/s42255-024-01175-9] [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: 01/26/2024] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Maintaining blood glucose homeostasis during fasting and feeding is crucial for the prevention of dysregulation that can lead to either hypo- or hyperglycaemia. Here we identified feimin, encoded by a gene with a previously unknown function (B230219D22Rik in mice, C5orf24 in humans), as a key modulator of glucose homeostasis. Feimin is secreted from skeletal muscle during feeding and binds to its receptor, receptor protein tyrosine kinase Mer (MERTK), promoting glucose uptake and inhibiting glucose production by activation of AKT. Administration of feimin and insulin synergistically improves blood glucose homeostasis in both normal and diabetic mice. Notably, a specific single nucleotide polymorphism (rs7604639, G>A) within the MERTK gene, causing an amino acid substitution (R466K) within the feimin-MERTK binding region, leads to reduced association with feimin and elevated postprandial blood glucose and insulin levels in humans. Our findings underscore a role of the feimin-MERTK signalling axis in glucose homeostasis, providing valuable insights into potential therapeutic avenues for diabetes.
Collapse
Affiliation(s)
- Xiaoliu Shi
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiao Hu
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xinlei Fang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Liangjie Jia
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Fangchao Wei
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Peng
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Menghao Liu
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Ke Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China
- Shandong Institute of Endocrine & Metabolic Disease, Jinan, China
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fengyi Chen
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaoli Hu
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yongfeng Song
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, China.
- Shandong Institute of Endocrine & Metabolic Disease, Jinan, China.
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China.
| | - Yiguo Wang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
21
|
Harrison AG, Yang D, Cahoon JG, Geng T, Cao Z, Karginov TA, Hu Y, Li X, Chiari CC, Qyang Y, Vella AT, Fan Z, Vanaja SK, Rathinam VA, Witczak CA, Bogan JS, Wang P. UBXN9 governs GLUT4-mediated spatial confinement of RIG-I-like receptors and signaling. Nat Immunol 2024; 25:2234-2246. [PMID: 39567760 PMCID: PMC12067455 DOI: 10.1038/s41590-024-02004-7] [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: 09/20/2023] [Accepted: 10/04/2024] [Indexed: 11/22/2024]
Abstract
The cytoplasmic RIG-I-like receptors (RLRs) recognize viral RNA and initiate innate antiviral immunity. RLR signaling also triggers glycolytic reprogramming through glucose transporters (GLUTs), whose role in antiviral immunity is elusive. Here, we unveil that insulin-responsive GLUT4 inhibits RLR signaling independently of glucose uptake in adipose and muscle tissues. At steady state, GLUT4 is trapped at the Golgi matrix by ubiquitin regulatory X domain 9 (UBXN9, TUG). Following RNA virus infection, GLUT4 is released and translocated to the cell surface where it spatially segregates a significant pool of cytosolic RLRs, preventing them from activating IFN-β responses. UBXN9 deletion prompts constitutive GLUT4 translocation, sequestration of RLRs and attenuation of antiviral immunity, whereas GLUT4 deletion heightens RLR signaling. Notably, reduced GLUT4 expression is uniquely associated with human inflammatory myopathies characterized by hyperactive interferon responses. Overall, our results demonstrate a noncanonical UBXN9-GLUT4 axis that controls antiviral immunity via plasma membrane tethering of cytosolic RLRs.
Collapse
Affiliation(s)
- Andrew G Harrison
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Duomeng Yang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA.
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, Guangdong, China.
| | - Jason G Cahoon
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Tingting Geng
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Ziming Cao
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Timofey A Karginov
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Youjia Hu
- Section of Endocrinology, Department of Internal Medicine, and Department of Cell Biology, and Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT, USA
| | - Xin Li
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Conner C Chiari
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Anthony T Vella
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Zhichao Fan
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Vijay A Rathinam
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Carol A Witczak
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jonathan S Bogan
- Section of Endocrinology, Department of Internal Medicine, and Department of Cell Biology, and Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT, USA
| | - Penghua Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
22
|
Kohlhoff G, Kirwan R, Mushtaq S. The effect of vitamin D supplementation on markers of insulin resistance in women with polycystic ovarian syndrome: a systematic review. Eur J Nutr 2024; 63:2859-2869. [PMID: 39276209 PMCID: PMC11519308 DOI: 10.1007/s00394-024-03489-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 08/26/2024] [Indexed: 09/16/2024]
Abstract
BACKGROUND Insulin resistance (IR) is a common pathology in women with polycystic ovarian syndrome (PCOS) involved in increased rates of cardiometabolic disease such as diabetes and cardiovascular disease. Low serum vitamin D is often associated with insulin resistance but there is no consensus on whether vitamin D supplementation can ameliorate markers of IR in PCOS. OBJECTIVES We assessed evidence on the effects of vitamin D supplementation (≥ 1000 IU/day), without the use of additional supplements or other pharmacological treatments known to affect IR, on markers of IR and glycemic control in women with PCOS. DESIGN A systematic search was conducted using PubMed, Medline and Web of Science databases from January 2000 up to November 2023. Randomized controlled trials that assessed the effects of vitamin D supplementation in women with PCOS, on fasting glucose, fasting insulin, glycated haemoglobin (HbA1c) or homeostatic model assessment for insulin resistance (HOMA-IR) were included. RESULTS 9 studies were identified. Study populations ranged from 28 to 180 participants, with mean ages ranging from 22 to 30 years. Daily vitamin D doses ranged from 1714-12,000 IU. Of the included studies, 3 reported statistically significant reductions in fasting glucose, 2 reported reductions in fasting insulin, 2 reported reductions in HOMA-IR, none reported reductions in HbA1c and 5 reported no differences in any of the relevant outcomes. CONCLUSIONS In conclusion, in RCTs of vitamin D supplementation in women with PCOS, the majority of studies do not report statistically significant improvements in fasting glucose, fasting insulin, HbA1c or HOMA-IR. However, as a minority of studies report some statistically significant results, further investigation may be warranted. REGISTRY PROSPERO ID: CRD42023486144.
Collapse
Affiliation(s)
- Georgia Kohlhoff
- Faculty of Medicine, Dentistry and Life Sciences, University of Chester, Chester, UK
| | - Richard Kirwan
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.
| | - Sohail Mushtaq
- Faculty of Medicine, Dentistry and Life Sciences, University of Chester, Chester, UK
| |
Collapse
|
23
|
Zhang L, Luo P, Li H, Pan Y, Zhang H, Si X, Chen W, Huang Y. Chicken GLUT4 function via enhancing mitochondrial oxidative phosphorylation and inhibiting ribosome pathway in skeletal muscle satellite cells. Poult Sci 2024; 103:104403. [PMID: 39515116 PMCID: PMC11584589 DOI: 10.1016/j.psj.2024.104403] [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: 08/19/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
Glucose Transporter 4 (GLUT4) is a crucial protein facilitating glucose uptake and metabolism across cell membranes in mammals. However, information on GLUT4 in birds has historically been limited. In this study, we investigated the dynamic expression profile of chicken GLUT4 using real-time quantitative PCR (RT-qPCR) and examined its potential effects and mechanisms via GLUT4 overexpression and RNA sequencing (RNA-seq) in chicken primary skeletal muscle satellite cells (CP-SMSCs). Our results demonstrated that chicken GLUT4 is differentially expressed across tissues, with predominant expression in skeletal muscles, and across developmental stages of CP-SMSCs, with notable upregulation during the phases of cell proliferation and early differentiation. Notably, 0.1 μM insulin for 60 min significantly elevated the expression of GLUT4 in CP-SMSCs (P < 0.05). GLUT4 overexpression in CP-SMSCs promoted cell proliferation, as evidenced by Cell Counting Kit-8 (CCK-8) (P < 0.05) and 5-Ethynyl-2'-Deoxyuridine (EDU) assays (P < 0.05), and enhanced glucose consumption following 0.1 μM insulin treatment (P < 0.05). However, it inhibited glucose consumption 12 h after the addition of 5 g/L glucose (P < 0.05). After overexpressing GLUT4, we identified 302 differentially expressed genes (DEGs) in CP-SMSCs, with 134 upregulated and 168 downregulated. These DEGs are primarily enriched in pathways such as oxidative phosphorylation, ribosome, cardiac muscle contraction, ATP metabolic processes, and mitochondrial protein complexes. Specifically, in the enriched oxidative phosphorylation pathway, the upregulated DEGs (12) encode mitochondrial proteins, while the downregulated DEGs (6) are nuclear genome-derived. The ribosomal pathway is predominantly inhibited, accompanying with the downregulation of the translocase of outer mitochondrial membrane 7 (TOMM7)/translocase of inner mitochondrial membrane 8 (TIMM8A) complex responsible for mitochondrial protein transport, and a reduction in 28S (LOC121106978) and 18S (LOC112533601) ribosomal rRNAs. In conclusion, chicken GLUT4 is dynamically modulated during development and acts as an insulin responder that significantly regulates cellular glucose uptake and cell proliferation. This regulation occurs mainly through enhancing the mitochondrial oxidative phosphorylation and inhibiting ribosomal pathway.
Collapse
Affiliation(s)
- Lin Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Pengna Luo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Huihong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Yuxian Pan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Huaiyong Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Xuemeng Si
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Wen Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China
| | - Yanqun Huang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China.
| |
Collapse
|
24
|
Grima-Terrén M, Campanario S, Ramírez-Pardo I, Cisneros A, Hong X, Perdiguero E, Serrano AL, Isern J, Muñoz-Cánoves P. Muscle aging and sarcopenia: The pathology, etiology, and most promising therapeutic targets. Mol Aspects Med 2024; 100:101319. [PMID: 39312874 DOI: 10.1016/j.mam.2024.101319] [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: 02/27/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Sarcopenia is a progressive muscle wasting disorder that severely impacts the quality of life of elderly individuals. Although the natural aging process primarily causes sarcopenia, it can develop in response to other conditions. Because muscle function is influenced by numerous changes that occur with age, the etiology of sarcopenia remains unclear. However, recent characterizations of the aging muscle transcriptional landscape, signaling pathway disruptions, fiber and extracellular matrix compositions, systemic metabolomic and inflammatory responses, mitochondrial function, and neurological inputs offer insights and hope for future treatments. This review will discuss age-related changes in healthy muscle and our current understanding of how this can deteriorate into sarcopenia. As our elderly population continues to grow, we must understand sarcopenia and find treatments that allow individuals to maintain independence and dignity throughout an extended lifespan.
Collapse
Affiliation(s)
- Mercedes Grima-Terrén
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Silvia Campanario
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Ignacio Ramírez-Pardo
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Andrés Cisneros
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Xiaotong Hong
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | | | - Antonio L Serrano
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Joan Isern
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Pura Muñoz-Cánoves
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain.
| |
Collapse
|
25
|
Tong A, Wang D, Jia N, Zheng Y, Qiu Y, Chen W, El-Seed HR, Zhao C. Algal Active Ingredients and Their Involvement in Managing Diabetic Mellitus. BIOLOGY 2024; 13:904. [PMID: 39596859 PMCID: PMC11591677 DOI: 10.3390/biology13110904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Diabetes mellitus (DM) is becoming increasingly prominent, posing a serious threat to human health. Its prevalence is rising every year, and often affects young people. In the past few decades, research on marine algae has been recognized as a major field of drug discovery. Seaweed active substances, including algal polysaccharides, algal polyphenols, algal unsaturated fatty acids, and algal dietary fiber, have unique biological activities. This article reviews the effects and mechanisms of the types, structures, and compositions of seaweed on inhibiting glucose and lipid metabolism disorders, with a focus on the inhibitory effect of active substances on blood glucose reduction. The aim is to provide a basis for the development of seaweed active substance hypoglycemic drugs.
Collapse
Affiliation(s)
- Aijun Tong
- College of Tea and Food Science, Wuyi University, Wuyishan 354300, China;
| | - Dengwei Wang
- Department of Chronic and Noncommunicable Disease Control and Prevention, Fujian Provincial Center for Disease Control and Prevention, Fuzhou 350012, China;
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China (W.C.)
| | - Nan Jia
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China (W.C.)
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ying Zheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yusong Qiu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Weichao Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China (W.C.)
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hesham R. El-Seed
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Chao Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China (W.C.)
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
26
|
Fu S, Gong X, Liang K, Ding K, Qiu L, Cen H, Du H. KLF3 impacts insulin sensitivity and glucose uptake in skeletal muscle. Am J Physiol Cell Physiol 2024; 327:C1219-C1235. [PMID: 39250818 DOI: 10.1152/ajpcell.00085.2024] [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: 02/04/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Skeletal muscle is one of the predominant sites involved in glucose disposal, accounting for ∼80% of postprandial glucose uptake, and plays a critical role in maintaining glycemic homeostasis. Dysregulation of energy metabolism in skeletal muscle is involved in developing insulin resistance and type 2 diabetes (T2D). Transcriptomic responses of skeletal muscle to exercise found that the expression of Klf3 was increased in T2D Goto-Kakizaki (GK) rats and decreased after exercise with improved hyperglycemia and insulin resistance, implying that Klf3 might be associated with insulin sensitivity and glucose metabolism. We also found that knockdown of Klf3 promoted basal and insulin-stimulated glucose uptake in L6 myotubes, whereas overexpression of Klf3 resulted in the opposite. Through pairwise comparisons of L6 myotubes transcriptome, we identified 2,256 and 1,988 differentially expressed genes in Klf3 knockdown and overexpression groups, respectively. In insulin signaling, the expression of Slc2a4, Akt2, Insr, and Sorbs1 was significantly increased by Klf3 knockdown and decreased with Klf3 overexpression; Ptprf and Fasn were markedly downregulated in Klf3 reduced group and upregulated in Klf3 overexpressed group. Moreover, downregulation of Klf3 promoted the expression of glucose transporter 4 (GLUT4) and protein kinase B (AKT) proteins, as well as the translocation of GLUT4 to the cell membrane in the basal situation, and enhanced insulin sensitivity, characterized by increased insulin-stimulated GLUT4 translocation and AKT, TBC1 domain family member 1 (TBC1D1) and TBC1 domain family member 4 (TBC1D4) phosphorylation, whereas overexpression of Klf3 showed contrary results. These results suggest that Klf3 affects glucose uptake and insulin sensitivity via insulin signal transduction and intracellular metabolism, offering a novel potential treatment strategy for T2D.NEW & NOTEWORTHY The knockdown of Klf3 increased glucose uptake and improved insulin sensitivity in L6 myotubes, whereas its overexpression had the opposite effect. To explore the underlying mechanisms, we evaluated the transcriptional profiles of L6 myotubes after Klf3 knockdown and overexpression and revealed that metabolism and insulin-related pathways were significantly impacted. Klf3 also influenced the expression or modification of glucose transporter 4 (GLUT4), protein kinase B (AKT), TBC1 domain family member 1 (TBC1D1), and TBC1 domain family member 4 (TBC1D4) in the insulin signaling pathway, affecting insulin sensitivity and glucose uptake.
Collapse
Affiliation(s)
- Shuying Fu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, People's Republic of China
- School of Life Sciences, Zhaoqing University, Zhaoqing, People's Republic of China
| | - Xiaocheng Gong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Keying Liang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Ke Ding
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Li Qiu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Huice Cen
- School of Life Sciences, Zhaoqing University, Zhaoqing, People's Republic of China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, People's Republic of China
| |
Collapse
|
27
|
Kato Y, Ariyoshi K, Nohara Y, Matsunaga N, Shimauchi T, Shindo N, Nishimura A, Mi X, Kim SG, Ide T, Kawanishi E, Ojida A, Nakashima N, Mori Y, Nishida M. Inhibition of dynamin-related protein 1-filamin interaction improves systemic glucose metabolism. Br J Pharmacol 2024; 181:4328-4347. [PMID: 38986570 DOI: 10.1111/bph.16487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 05/05/2024] [Accepted: 05/18/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Maintaining mitochondrial quality is attracting attention as a new strategy to treat diabetes and diabetic complications. We previously reported that mitochondrial hyperfission by forming a protein complex between dynamin-related protein (Drp) 1 and filamin, mediates chronic heart failure and cilnidipine, initially developed as an L/N-type Ca2+ channel blocker, improves heart failure by inhibiting Drp1-filamin protein complex. We investigated whether cilnidipine improves hyperglycaemia of various diabetic mice models. EXPERIMENTAL APPROACH Retrospective analysis focusing on haemoglobin A1c (HbA1c) was performed in hypertensive and hyperglycaemic patients taking cilnidipine and amlodipine. After developing diabetic mice by streptozotocin (STZ) treatment, an osmotic pump including drug was implanted intraperitoneally, followed by weekly measurements of blood glucose levels. Mitochondrial morphology was analysed by electron microscopy. A Ca2+ channel-insensitive cilnidipine derivative (1,4-dihydropyridine [DHP]) was synthesized and its pharmacological effect was evaluated using obese (ob/ob) mice fed with high-fat diet (HFD). KEY RESULTS In patients, cilnidipine was superior to amlodipine in HbA1c lowering effect. Cilnidipine treatment improved systemic hyperglycaemia and mitochondrial morphological abnormalities in STZ-exposed mice, without lowering blood pressure. Cilnidipine failed to improve hyperglycaemia of ob/ob mice, with suppressing insulin secretion. 1,4-DHP improved hyperglycaemia and mitochondria abnormality in ob/ob mice fed HFD. 1,4-DHP and cilnidipine improved basal oxygen consumption rate of HepG2 cells cultured under 25 mM glucose. CONCLUSION AND IMPLICATIONS Inhibition of Drp1-filamin protein complex formation becomes a new strategy for type 2 diabetes treatment.
Collapse
Affiliation(s)
- Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Ariyoshi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasunobu Nohara
- Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Naoya Matsunaga
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tsukasa Shimauchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Naoya Shindo
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Sang Geon Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang-si, South Korea
| | - Tomomi Ide
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Kawanishi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akio Ojida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Nakashima
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| |
Collapse
|
28
|
Chen W, Kahn CR. Insulin. Trends Endocrinol Metab 2024:S1043-2760(24)00253-4. [PMID: 39419674 DOI: 10.1016/j.tem.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Wenqiang Chen
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark.
| | - C Ronald Kahn
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
29
|
Zhong C, Li N, Wang S, Li D, Yang Z, Du L, Huang G, Li H, Yeung WS, He S, Ma S, Wang Z, Jiang H, Zhang H, Li Z, Wen X, Xue S, Tao X, Li H, Xie D, Zhang Y, Chen Z, Wang J, Yan J, Liang Z, Zhang Z, Zhong Z, Wu Z, Wan C, Liang C, Wang L, Yu S, Ma Y, Yu Y, Li F, Chen Y, Zhang B, Lyu A, Ren F, Zhou H, Liu J, Zhang G. Targeting osteoblastic 11β-HSD1 to combat high-fat diet-induced bone loss and obesity. Nat Commun 2024; 15:8588. [PMID: 39362888 PMCID: PMC11449908 DOI: 10.1038/s41467-024-52965-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
Excessive glucocorticoid (GC) action is linked to various metabolic disorders. Recent findings suggest that disrupting skeletal GC signaling prevents bone loss and alleviates metabolic disorders in high-fat diet (HFD)-fed obese mice, underpinning the neglected contribution of skeletal GC action to obesity and related bone loss. Here, we show that the elevated expression of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), the enzyme driving local GC activation, and GC signaling in osteoblasts, are associated with bone loss and obesity in HFD-fed male mice. Osteoblast-specific 11β-HSD1 knockout male mice exhibit resistance to HFD-induced bone loss and metabolic disorders. Mechanistically, elevated 11β-HSD1 restrains glucose uptake and osteogenic activity in osteoblast. Pharmacologically inhibiting osteoblastic 11β-HSD1 by using bone-targeted 11β-HSD1 inhibitor markedly promotes bone formation, ameliorates glucose handling and mitigated obesity in HFD-fed male mice. Taken together, our study demonstrates that osteoblastic 11β-HSD1 directly contributes to HFD-induced bone loss, glucose handling impairment and obesity.
Collapse
Affiliation(s)
- Chuanxin Zhong
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Nanxi Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dijie Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, College of Life Sciences, Guangxi Normal University, Gui Lin, China
| | - Zhihua Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guangxin Huang
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haitian Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wing Sze Yeung
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shan He
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuting Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zhuqian Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarui Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhanghao Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiaoxin Wen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Song Xue
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaohui Tao
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Haorui Li
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Duoli Xie
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yihao Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zefeng Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Junqin Wang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jianfeng Yan
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhengming Liang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhigang Zhong
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zeting Wu
- International Medical Service Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chao Wan
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sifan Yu
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yang Chen
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Baoting Zhang
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Hong Kong, China.
| | - Fuzeng Ren
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia.
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
30
|
Singh SSB, Patil KN. SIRT1/AMPK-mediated pathway: Ferulic acid from sugar beet pulp mitigating obesity-induced diabetes-linked complications and improving metabolic health. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159511. [PMID: 38761896 DOI: 10.1016/j.bbalip.2024.159511] [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: 08/08/2023] [Revised: 03/28/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Obesity-induced type 2 diabetes (T2D) increases the risk of metabolic syndrome due to the high calorie intake. The role of sugar beet pulp (SBP) in T2D and the mechanism of its action remain unclear, though it is abundant in phenolics and has antioxidant activity. In this study, we isolated and purified ferulic acid from SBP, referred to as SBP-E, and studied the underlying molecular mechanisms in the regulation of glucose and lipid metabolism developing high glucose/high fat diet-induced diabetic models in vitro and in vivo. SBP-E showed no cytotoxicity and reduced the oxidative stress by increasing glutathione (GSH) in human liver (HepG2) and rat skeletal muscle (L6) cells. It also decreased body weight gain, food intake, fasting blood glucose levels (FBGL), glucose intolerance, hepatic steatosis, and lipid accumulation. Additionally, SBP-E decreased the oxidative stress and improved the antioxidant enzyme levels in high-fat diet (HFD)-induced T2D mice. Further, SBP-E reduced plasma and liver advanced glycation end products (AGEs), malondialdehyde (MDA), and pro-inflammatory cytokines, and increased anti-inflammatory cytokines in HFD-fed mice. Importantly, SBP-E significantly elevated AMPK, glucose transporter, SIRT1 activity, and Nrf2 expression and decreased ACC activity and SREBP1 levels in diabetic models. Collectively, our study results suggest that SBP-E treatment can improve obesity-induced T2D by regulating glucose and lipid metabolism via SIRT1/AMPK signalling and the AMPK/SREBP1/ACC1 pathway.
Collapse
Affiliation(s)
- Sangeetha S B Singh
- Department of Microbiology and Fermentation Technology, Council of Scientific & Industrial Research-Central Food Technological Research Institute (CSIR-CFTRI), Mysuru 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - K Neelakanteshwar Patil
- Department of Microbiology and Fermentation Technology, Council of Scientific & Industrial Research-Central Food Technological Research Institute (CSIR-CFTRI), Mysuru 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
31
|
Greig J, Bates GT, Yin DI, Briant K, Simonetti B, Cullen PJ, Brodsky FM. CHC22 clathrin recruitment to the early secretory pathway requires two-site interaction with SNX5 and p115. EMBO J 2024; 43:4298-4323. [PMID: 39160272 PMCID: PMC11445476 DOI: 10.1038/s44318-024-00198-y] [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: 10/04/2023] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
The two clathrin isoforms, CHC17 and CHC22, mediate separate intracellular transport routes. CHC17 performs endocytosis and housekeeping membrane traffic in all cells. CHC22, expressed most highly in skeletal muscle, shuttles the glucose transporter GLUT4 from the ERGIC (endoplasmic-reticulum-to-Golgi intermediate compartment) directly to an intracellular GLUT4 storage compartment (GSC), from where GLUT4 can be mobilized to the plasma membrane by insulin. Here, molecular determinants distinguishing CHC22 from CHC17 trafficking are defined. We show that the C-terminal trimerization domain of CHC22 interacts with SNX5, which also binds the ERGIC tether p115. SNX5, and the functionally redundant SNX6, are required for CHC22 localization independently of their participation in the endosomal ESCPE-1 complex. In tandem, an isoform-specific patch in the CHC22 N-terminal domain separately mediates binding to p115. This dual mode of clathrin recruitment, involving interactions at both N- and C-termini of the heavy chain, is required for CHC22 targeting to ERGIC membranes to mediate the Golgi-bypass route for GLUT4 trafficking. Interference with either interaction inhibits GLUT4 targeting to the GSC, defining a bipartite mechanism regulating a key pathway in human glucose metabolism.
Collapse
Affiliation(s)
- Joshua Greig
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - George T Bates
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - Daowen I Yin
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - Kit Briant
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - Boris Simonetti
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Peter J Cullen
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Frances M Brodsky
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK.
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK.
| |
Collapse
|
32
|
Ruetz TJ, Pogson AN, Kashiwagi CM, Gagnon SD, Morton B, Sun ED, Na J, Yeo RW, Leeman DS, Morgens DW, Tsui CK, Li A, Bassik MC, Brunet A. CRISPR-Cas9 screens reveal regulators of ageing in neural stem cells. Nature 2024; 634:1150-1159. [PMID: 39358505 PMCID: PMC11525198 DOI: 10.1038/s41586-024-07972-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/20/2024] [Indexed: 10/04/2024]
Abstract
Ageing impairs the ability of neural stem cells (NSCs) to transition from quiescence to proliferation in the adult mammalian brain. Functional decline of NSCs results in the decreased production of new neurons and defective regeneration following injury during ageing1-4. Several genetic interventions have been found to ameliorate old brain function5-8, but systematic functional testing of genes in old NSCs-and more generally in old cells-has not been done. Here we develop in vitro and in vivo high-throughput CRISPR-Cas9 screening platforms to systematically uncover gene knockouts that boost NSC activation in old mice. Our genome-wide screens in primary cultures of young and old NSCs uncovered more than 300 gene knockouts that specifically restore the activation of old NSCs. The top gene knockouts are involved in cilium organization and glucose import. We also establish a scalable CRISPR-Cas9 screening platform in vivo, which identified 24 gene knockouts that boost NSC activation and the production of new neurons in old brains. Notably, the knockout of Slc2a4, which encodes the GLUT4 glucose transporter, is a top intervention that improves the function of old NSCs. Glucose uptake increases in NSCs during ageing, and transient glucose starvation restores the ability of old NSCs to activate. Thus, an increase in glucose uptake may contribute to the decline in NSC activation with age. Our work provides scalable platforms to systematically identify genetic interventions that boost the function of old NSCs, including in vivo, with important implications for countering regenerative decline during ageing.
Collapse
Affiliation(s)
- Tyson J Ruetz
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Angela N Pogson
- Department of Genetics, Stanford University, Stanford, CA, USA
- Developmental Biology Graduate Program, Stanford University, Stanford, CA, USA
| | | | | | - Bhek Morton
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Eric D Sun
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biomedical Informatics Graduate Program, Stanford University, Stanford, CA, USA
| | - Jeeyoon Na
- Department of Genetics, Stanford University, Stanford, CA, USA
- Stem Cell Biology & Regenerative Medicine Graduate Program, Stanford University, Stanford, CA, USA
| | - Robin W Yeo
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Dena S Leeman
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - David W Morgens
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - C Kimberly Tsui
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Amy Li
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
33
|
Ouyang W, Huang Z, Wan K, Nie T, Chen H, Yao H. RNA ac 4C modification in cancer: Unraveling multifaceted roles and promising therapeutic horizons. Cancer Lett 2024; 601:217159. [PMID: 39128536 DOI: 10.1016/j.canlet.2024.217159] [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: 03/21/2024] [Revised: 06/18/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
RNA modifications play a crucial role in cancer development, profoundly influencing various stages of the RNA lifecycle. These stages encompass nuclear processing, nuclear export, splicing, and translation in the cytoplasm. Among RNA modifications, RNA ac4C modification, also known as N4-acetylcytidine, stands out for its unique role in acetylation processes. Specific proteins regulate RNA ac4C modification, maintaining the dynamic and reversible nature of these changes. This review explores the molecular mechanisms and biological functions of RNA ac4C modification. It examines the intricate ways in which RNA ac4C modification influences the pathogenesis and progression of cancer. Additionally, the review provides an integrated overview of the current methodologies for detecting RNA ac4C modification. Exploring the potential applications of manipulating this modification suggests avenues for novel therapeutic strategies, potentially leading to more effective cancer treatments in the future.
Collapse
Affiliation(s)
- Wenhao Ouyang
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Zhenjun Huang
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Keyu Wan
- The First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Tiantian Nie
- The First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Haizhu Chen
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China.
| | - Herui Yao
- Department of Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China.
| |
Collapse
|
34
|
Sarker DK, Ray P, Dutta AK, Rouf R, Uddin SJ. Antidiabetic potential of fenugreek ( Trigonella foenum-graecum): A magic herb for diabetes mellitus. Food Sci Nutr 2024; 12:7108-7136. [PMID: 39479631 PMCID: PMC11521722 DOI: 10.1002/fsn3.4440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/13/2024] [Accepted: 08/17/2024] [Indexed: 11/02/2024] Open
Abstract
Fenugreek (Trigonella foenum-graecum) is a widely grown dietary herb in Asia, and its seeds are traditionally used for several diseases, including diabetes. The seeds and leaves possess a variety of compounds that play an important role in regulating their hypoglycemic effect. However, so far, no extensive systematic review exists on its antidiabetic effect, highlighting the molecular mechanisms and isolated compounds. The purpose of this review is to summarize the preclinical and clinical antidiabetic properties of fenugreek and its isolated compounds by focusing on underlying mechanisms. PubMed, Google Scholar, Science Direct, and Scopus databases were searched to retrieve articles until June, 2024. Preclinical studies demonstrated that the antidiabetic effect of fenugreek was mostly associated with enhanced glucose transporter type-4 (GLUT4) translocation and hexokinase activity, decreased glucose-6-phosphatase and fructose-1,6-bisphosphatase activities, inhibited α-amylase and maltase activities, protected β cells, and increased insulin release. Furthermore, few studies have reported its role as a glucagon-like peptide-1 (GLP-1) modulator, 5'-AMP-activated kinase (AMPK) activator, and dipeptidyl peptidase-IV (DPP-IV) inhibitor. Further clinical trials showed that fenugreek seeds improved blood glucose levels, insulin resistance, insulin sensitivity, and lipid profiles. This study highlights significant evidence of the antidiabetic effect of fenugreek and its isolated compounds; therefore, it could be a potential therapy for diabetes.
Collapse
Affiliation(s)
- Dipto Kumer Sarker
- Pharmacy Discipline, Life Science SchoolKhulna UniversityKhulnaBangladesh
| | - Pallobi Ray
- Pharmacy Discipline, Life Science SchoolKhulna UniversityKhulnaBangladesh
| | | | - Razina Rouf
- Department of Pharmacy, Faculty of Life ScienceBangabandhu Sheikh Mujibur Rahman Science & Technology UniversityGopalganjBangladesh
| | - Shaikh Jamal Uddin
- Pharmacy Discipline, Life Science SchoolKhulna UniversityKhulnaBangladesh
| |
Collapse
|
35
|
Glibetic N, Bowman S, Skaggs T, Weichhaus M. The Use of Patient-Derived Organoids in the Study of Molecular Metabolic Adaptation in Breast Cancer. Int J Mol Sci 2024; 25:10503. [PMID: 39408832 PMCID: PMC11477048 DOI: 10.3390/ijms251910503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Around 13% of women will likely develop breast cancer during their lifetime. Advances in cancer metabolism research have identified a range of metabolic reprogramming events, such as altered glucose and amino acid uptake, increased reliance on glycolysis, and interactions with the tumor microenvironment (TME), all of which present new opportunities for targeted therapies. However, studying these metabolic networks is challenging in traditional 2D cell cultures, which often fail to replicate the three-dimensional architecture and dynamic interactions of real tumors. To address this, organoid models have emerged as powerful tools. Tumor organoids are 3D cultures, often derived from patient tissue, that more accurately mimic the structural and functional properties of actual tumor tissues in vivo, offering a more realistic model for investigating cancer metabolism. This review explores the unique metabolic adaptations of breast cancer and discusses how organoid models can provide deeper insights into these processes. We evaluate the most advanced tools for studying cancer metabolism in three-dimensional culture models, including optical metabolic imaging (OMI), matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI), and recent advances in conventional techniques applied to 3D cultures. Finally, we explore the progress made in identifying and targeting potential therapeutic targets in breast cancer metabolism.
Collapse
Affiliation(s)
- Natalija Glibetic
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- The IDeA Networks of Biomedical Research Excellence (INBRE) Program, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
- United Nations CIFAL Honolulu Center, Chaminade University, Honolulu, HI 96816, USA
| | - Scott Bowman
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- Undergraduate Program in Biochemistry, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
| | - Tia Skaggs
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- Undergraduate Program in Biology, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
| | - Michael Weichhaus
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
| |
Collapse
|
36
|
Chen X, Li CG, Zhou X, Zhu M, Jin J, Wang P. A new perspective on the regulation of glucose and cholesterol transport by mitochondria-lysosome contact sites. Front Physiol 2024; 15:1431030. [PMID: 39290619 PMCID: PMC11405319 DOI: 10.3389/fphys.2024.1431030] [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: 05/13/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Mitochondria and lysosomes play a very important role in maintaining cellular homeostasis, and the dysfunction of these organelles is closely related to many diseases. Recent studies have revealed direct interactions between mitochondria and lysosomes, forming mitochondria-lysosome contact sites that regulate organelle network dynamics and mediate the transport of metabolites between them. Impaired function of these contact sites is not only linked to physiological processes such as glucose and cholesterol transport but also closely related to the pathological processes of metabolic diseases. Here, we highlight the recent progress in understanding the mitochondria-lysosome contact sites, elucidate their role in regulating metabolic homeostasis, and explore the potential implications of this pathway in metabolic disorders.
Collapse
Affiliation(s)
- Xiaolong Chen
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Minghua Zhu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jing Jin
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Ping Wang
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
37
|
Kirtipal N, Seo Y, Son J, Lee S. Systems Biology of Human Microbiome for the Prediction of Personal Glycaemic Response. Diabetes Metab J 2024; 48:821-836. [PMID: 39313228 PMCID: PMC11449821 DOI: 10.4093/dmj.2024.0382] [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: 07/13/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
The human gut microbiota is increasingly recognized as a pivotal factor in diabetes management, playing a significant role in the body's response to treatment. However, it is important to understand that long-term usage of medicines like metformin and other diabetic treatments can result in problems, gastrointestinal discomfort, and dysbiosis of the gut flora. Advanced sequencing technologies have improved our understanding of the gut microbiome's role in diabetes, uncovering complex interactions between microbial composition and metabolic health. We explore how the gut microbiota affects glucose metabolism and insulin sensitivity by examining a variety of -omics data, including genomics, transcriptomics, epigenomics, proteomics, metabolomics, and metagenomics. Machine learning algorithms and genome-scale modeling are now being applied to find microbiological biomarkers associated with diabetes risk, predicted disease progression, and guide customized therapy. This study holds promise for specialized diabetic therapy. Despite significant advances, some concerns remain unanswered, including understanding the complex relationship between diabetes etiology and gut microbiota, as well as developing user-friendly technological innovations. This mini-review explores the relationship between multiomics, precision medicine, and machine learning to improve our understanding of the gut microbiome's function in diabetes. In the era of precision medicine, the ultimate goal is to improve patient outcomes through personalized treatments.
Collapse
Affiliation(s)
- Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Youngchang Seo
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jangwon Son
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
38
|
Guo Y, Gu D, Okeke ES, Feng W, Chen Y, Mao G, Yang L, Wu X, Zhao T. Fenitrothion induces glucose metabolism disorders in rat liver BRL cells by inhibiting AMPKα and IRS1/PI3K/AKT signaling pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 204:106098. [PMID: 39277407 DOI: 10.1016/j.pestbp.2024.106098] [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: 06/05/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024]
Abstract
Fenitrothion (FNT) is a common organophosphorus pesticide that is widely used in both agricultural and domestic pest control. FNT has been frequently detected in various environmental media, including the human body, and is a notable contaminant. Epidemiological investigations have recently shown the implications of exposure to FNT in the incidence of various metabolic diseases, such as diabetes mellitus in humans, indicating that FNT may be a potential endocrine disruptor. However, the effects of FNT exposure on glucose homeostasis and their underlying mechanisms in model organisms remain largely unknown, which may limit our understanding of the health risks of FNT. In this study, FNT (4 5, 90, 180, and 4 50 μM) exposure model of rat hepatocytes (Buffalo Rat Liver, BRL cells) was established to investigate the effects and potential mechanisms of its toxicity on glucose metabolism. Several key processes of glucose metabolism were detected in this study. The results showed significantly increased glucose levels in the culture medium and decreased glycogen content in the FNT-exposed BRL cells. The results of quantitative real-time PCR and enzymology showed the abnormal expression of genes and activity/content of glucose metabolic enzymes involved in glucose metabolism, which might promote gluconeogenesis and inhibit glucose uptake, glycolysis, and glycogenesis. Furthermore, gluconeogenesis and glycolytic were carried out in the mitochondrial membrane. The abnormal of mitochondrial membrane potential may be a potential mechanism underlying FNT-induced glucose metabolism disorder. In addition, the mRNA and protein expression implicated that FNT may disrupt glucose metabolism by inhibiting the AMPKα and IRS1/PI3K/AKT signaling pathways. In conclusion, results provide in vitro evidence that FNT can cause glucose metabolism disorder, which emphasizes the potential health risks of exposure to FNT in inducing diabetes mellitus.
Collapse
Affiliation(s)
- Yuchao Guo
- School of the Environment, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Dandan Gu
- School of the Environment, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Emmanuel Sunday Okeke
- School of the Environment, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China; Department of Biochemistry, Faculty of Biological Science & Natural Science Unit, School of General Studies, University of Nigeria, Nsukka, Enugu State 410001, Nigeria
| | - Weiwei Feng
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, China
| | - Yao Chen
- School of the Environment, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Guanghua Mao
- School of the Environment, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Liuqing Yang
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, China
| | - Xiangyang Wu
- School of the Environment, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China.
| | - Ting Zhao
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, China.
| |
Collapse
|
39
|
Anwar A, Shukla S, Pathak P. Nitric oxide in modulating oxidative stress mediated skeletal muscle insulin resistance. Mol Biol Rep 2024; 51:944. [PMID: 39210004 DOI: 10.1007/s11033-024-09874-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Insulin resistance (IR) being the major cause behind different metabolic disorders, has attracted a lot of attention. Epidemiological data shows marked rise in the cases over a period of time. Nitric oxide (NO), produced from nitric oxide synthases (NOS), is involved in a variety of biological functions, alteration in which causes various disorders like hypertension, atherosclerosis, and angiogenesis-associated disorders. IR has been found to be a contributing factor, which is associated with abnormal NO signalling. Skeletal muscle is essential for metabolism, both for its role in glucose uptake and its importance in metabolic disease. In this article, we give an overview of the significance of NO in oxidative stress (OS) mediated IR, describing its role in different conditions that are associated with skeletal muscle IR. NO is found to be involved in the activation of insulin receptor downstream pathway, which suggests absence of NO could lead to reduced glucose uptake, and may ultimately result in IR.
Collapse
Affiliation(s)
- Aamir Anwar
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University (Lucknow Campus), Lucknow, Uttar Pradesh, 226010, India
| | - Shivang Shukla
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University (Lucknow Campus), Lucknow, Uttar Pradesh, 226010, India
| | - Priya Pathak
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University (Lucknow Campus), Lucknow, Uttar Pradesh, 226010, India.
| |
Collapse
|
40
|
Jia Y, Liu Y, Wang Y, Li J, Li G. Sialylation-induced stabilization of dynamic glycoprotein conformations unveiled by time-aligned parallel unfolding and glycan release mass spectrometry. Chem Sci 2024:d4sc03672g. [PMID: 39165727 PMCID: PMC11331314 DOI: 10.1039/d4sc03672g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
Sialylation, a critical post-translational modification, regulates glycoprotein structure and function by tuning their molecular heterogeneity. However, characterizing its subtle and dynamic conformational effects at the intact glycoprotein level remains challenging. We introduce a glycoform-resolved unfolding approach based on a high-throughput ion mobility-mass spectrometry (IM-MS) platform. This method integrates high-throughput unfolding with parallel fragmentation, enabling simultaneous analysis of sialylation patterns, stoichiometries, and their impact on conformational stability. Applying this approach to fetuin, we identified distinct sialylation patterns and their differential influence on protein conformation, namely sialylation-induced stabilization during early unfolding and increased flexibility in later unfolding stages. IM-MS-guided molecular dynamics simulations revealed that increased sialylation enhances the initial conformational stability, likely through enhanced electrostatic interactions and hydrogen bonding. These findings highlight the complex interplay between sialylation and protein dynamics and establish glycoform-resolved unfolding IM-MS as a powerful tool for characterizing glycoprotein conformational landscapes.
Collapse
Affiliation(s)
- Yifei Jia
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University Tianjin 300071 China
| | - Yichang Liu
- School of Pharmacy, Nantong University Nantong 226001 Jiangsu China
| | - Yamei Wang
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University Tianjin 300071 China
| | - Jinyu Li
- College of Chemistry, Fuzhou University Fuzhou 350108 Fujian China
| | - Gongyu Li
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University Tianjin 300071 China
- Haihe Laboratory of Sustainable Chemical Transformations Tianjin 300192 China
| |
Collapse
|
41
|
Taheri R, Mokhtari Y, Yousefi AM, Bashash D. The PI3K/Akt signaling axis and type 2 diabetes mellitus (T2DM): From mechanistic insights into possible therapeutic targets. Cell Biol Int 2024; 48:1049-1068. [PMID: 38812089 DOI: 10.1002/cbin.12189] [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: 07/07/2023] [Revised: 02/03/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is an immensely debilitating chronic disease that progressively undermines the well-being of various bodily organs and, indeed, most patients succumb to the disease due to post-T2DM complications. Although there is evidence supporting the activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway by insulin, which is essential in regulating glucose metabolism and insulin resistance, the significance of this pathway in T2DM has only been explored in a few studies. The current review aims to unravel the mechanisms by which different classes of PI3Ks control the metabolism of glucose; and also to discuss the original data obtained from international research laboratories on this topic. We also summarized the role of the PI3K/Akt signaling axis in target tissues spanning from the skeletal muscle to the adipose tissue and liver. Furthermore, inquiries regarding the impact of disrupting this axis on insulin function and the development of insulin resistance have been addressed. We also provide a general overview of the association of impaired PI3K/Akt signaling pathways in the pathogenesis of the most prevalent diabetes-related complications. The last section provides a special focus on the therapeutic potential of this axis by outlining the latest advances in active compounds that alleviate diabetes via modulation of the PI3K/Akt pathway. Finally, we comment on the future research aspects in which the field of T2DM therapies using PI3K modulators might be developed.
Collapse
Affiliation(s)
- Rana Taheri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yazdan Mokhtari
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Achter JS, Vega ET, Sorrentino A, Kahnert K, Galsgaard KD, Hernandez-Varas P, Wierer M, Holst JJ, Wojtaszewski JFP, Mills RW, Kjøbsted R, Lundby A. In-depth phosphoproteomic profiling of the insulin signaling response in heart tissue and cardiomyocytes unveils canonical and specialized regulation. Cardiovasc Diabetol 2024; 23:258. [PMID: 39026321 PMCID: PMC11264841 DOI: 10.1186/s12933-024-02338-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Insulin signaling regulates cardiac substrate utilization and is implicated in physiological adaptations of the heart. Alterations in the signaling response within the heart are believed to contribute to pathological conditions such as type-2 diabetes and heart failure. While extensively investigated in several metabolic organs using phosphoproteomic strategies, the signaling response elicited in cardiac tissue in general, and specifically in the specialized cardiomyocytes, has not yet been investigated to the same extent. METHODS Insulin or vehicle was administered to male C57BL6/JRj mice via intravenous injection into the vena cava. Ventricular tissue was extracted and subjected to quantitative phosphoproteomics analysis to evaluate the insulin signaling response. To delineate the cardiomyocyte-specific response and investigate the role of Tbc1d4 in insulin signal transduction, cardiomyocytes from the hearts of cardiac and skeletal muscle-specific Tbc1d4 knockout mice, as well as from wildtype littermates, were studied. The phosphoproteomic studies involved isobaric peptide labeling with Tandem Mass Tags (TMT), enrichment for phosphorylated peptides, fractionation via micro-flow reversed-phase liquid chromatography, and high-resolution mass spectrometry measurements. RESULTS We quantified 10,399 phosphorylated peptides from ventricular tissue and 12,739 from isolated cardiomyocytes, localizing to 3,232 and 3,128 unique proteins, respectively. In cardiac tissue, we identified 84 insulin-regulated phosphorylation events, including sites on the Insulin Receptor (InsrY1351, Y1175, Y1179, Y1180) itself as well as the Insulin receptor substrate protein 1 (Irs1S522, S526). Predicted kinases with increased activity in response to insulin stimulation included Rps6kb1, Akt1 and Mtor. Tbc1d4 emerged as a major phosphorylation target in cardiomyocytes. Despite limited impact on the global phosphorylation landscape, Tbc1d4 deficiency in cardiomyocytes attenuated insulin-induced Glut4 translocation and induced protein remodeling. We observed 15 proteins significantly regulated upon knockout of Tbc1d4. While Glut4 exhibited decreased protein abundance consequent to Tbc1d4-deficiency, Txnip levels were notably increased. Stimulation of wildtype cardiomyocytes with insulin led to the regulation of 262 significant phosphorylation events, predicted to be regulated by kinases such as Akt1, Mtor, Akt2, and Insr. In cardiomyocytes, the canonical insulin signaling response is elicited in addition to regulation on specialized cardiomyocyte proteins, such as Kcnj11Y12 and DspS2597. Details of all phosphorylation sites are provided. CONCLUSION We present a first global outline of the insulin-induced phosphorylation signaling response in heart tissue and in isolated adult cardiomyocytes, detailing the specific residues with changed phosphorylation abundances. Our study marks an important step towards understanding the role of insulin signaling in cardiac diseases linked to insulin resistance.
Collapse
Affiliation(s)
- Jonathan Samuel Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estefania Torres Vega
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Kahnert
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Hernandez-Varas
- Core Facility for Integrated Microscopy, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Wierer
- Proteomics Research Infrastructure, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Frank Pind Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Robert William Mills
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kjøbsted
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
43
|
Venugopal G, Dash R, Agrawal S, Ray S, Kumar Sahoo P, Ramadass B. A Novel Nutraceutical Supplement Lowers Postprandial Glucose and Insulin Levels upon a Carbohydrate-Rich Meal or Sucrose Drink Intake in Healthy Individuals-A Randomized, Placebo-Controlled, Crossover Feeding Study. Nutrients 2024; 16:2237. [PMID: 39064681 PMCID: PMC11280099 DOI: 10.3390/nu16142237] [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: 06/11/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Alkaloid- and polyphenol-rich white mulberry leaf and apple peel extracts have been shown to have potential glucose-lowering effects, benefitting the control of postprandial blood glucose levels. This study aimed to determine the effect of the combination of Malus domestica peel and Morus alba leaf extracts (GLUBLOCTM) on postprandial blood glucose and insulin-lowering effects in healthy adults after a carbohydrate-rich meal or sucrose drink intake. METHODS This study was designed as a randomized, crossover, single-blinded clinical trial. Out of 116 healthy participants, 85 subjects (aged 18-60 years) completed the day 1 and 5 crossover study. On day 1, subjects were supplemented with a placebo or GLUBLOCTM tablet 10 min before the carbohydrate-rich meal (300 g of tomato rice) or sucrose drink intake (75 g of sucrose dissolved in 300 mL water). On day 5, the treatments were crossed over, and the same diet was followed. Postprandial blood glucose and insulin levels were measured on days 1 and 5 (baseline 0, post-meal 30, 60, 90, and 120 min). Differences in iAUC, Cmax, and Tmax were determined between the placebo and GLUBLOCTM-treated cohorts. RESULTS Significant changes in total iAUC (0-120 min), Cmax, and Tmax of postprandial blood glucose and insulin levels were noticed upon GLUBLOCTM supplementation. The percentage reduction in the iAUC of blood glucose levels was 49.78% (iAUC0-60min) and 43.36% (iAUC0-120min), respectively, compared with the placebo in the sucrose drink intake study. Similarly, there was a 41.13% (iAUC0-60min) and 20.26% (iAUC0-120min) glucose-lowering effect compared with the placebo in the carbohydrate-rich meal intake study. CONCLUSIONS Premeal supplementation with GLUBLOCTM significantly reduced the postprandial surge in blood glucose and insulin levels after a carbohydrate-rich meal or sucrose drink intake over 120 min in healthy individuals. This study proves that GLUBLOCTM can manage steady postprandial blood glucose levels.
Collapse
Affiliation(s)
- Giriprasad Venugopal
- Center of Excellence for Clinical Microbiome Research, All India Institute of Medical Sciences Bhubaneswar, Bhubaneswar 751019, India; (G.V.); (R.D.)
| | - Rishikesh Dash
- Center of Excellence for Clinical Microbiome Research, All India Institute of Medical Sciences Bhubaneswar, Bhubaneswar 751019, India; (G.V.); (R.D.)
| | - Siwani Agrawal
- Department of Biochemistry, All India Institute of Medical Sciences, Bhubaneswar 751019, India;
| | - Sayantan Ray
- Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar 751019, India;
| | - Prasanta Kumar Sahoo
- Department of Ayurveda (AYUSH), All India Institute of Medical Sciences, Bhubaneswar 751019, India;
| | - Balamurugan Ramadass
- Center of Excellence for Clinical Microbiome Research, All India Institute of Medical Sciences Bhubaneswar, Bhubaneswar 751019, India; (G.V.); (R.D.)
- Department of Biochemistry, All India Institute of Medical Sciences, Bhubaneswar 751019, India;
| |
Collapse
|
44
|
Zhang X, Zheng W, Sun S, Du Y, Xu W, Sun Z, Liu F, Wang M, Zhao Z, Liu J, Liu Q. Cadmium contributes to cardiac metabolic disruption by activating endothelial HIF1A-GLUT1 axis. Cell Signal 2024; 119:111170. [PMID: 38604344 DOI: 10.1016/j.cellsig.2024.111170] [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: 01/21/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Cadmium (Cd) is an environmental risk factor of cardiovascular diseases. Researchers have found that Cd exposure causes energy metabolic disorders in the heart decades ago. However, the underlying molecular mechanisms are still elusive. In this study, male C57BL/6 J mice were exposed to cadmium chloride (CdCl2) through drinking water for 4 weeks. We found that exposure to CdCl2 increased glucose uptake and utilization, and disrupted normal metabolisms in the heart. In vitro studies showed that CdCl2 specifically increased endothelial glucose uptake without affecting cardiomyocytic glucose uptake and endothelial fatty acid uptake. The glucose transporter 1 (GLUT1) as well as its transcription factor HIF1A was significantly increased after CdCl2 treatment in endothelial cells. Further investigations found that CdCl2 treatment upregulated HIF1A expression by inhibiting its degradation through ubiquitin-proteasome pathway, thereby promoted its transcriptional activation of SLC2A1. Administration of HIF1A small molecule inhibitor echinomycin and A-485 reversed CdCl2-mediated increase of glucose uptake in endothelial cells. In accordance with this, intravenous injection of echinomycin effectively ameliorated CdCl2-mediated metabolic disruptions in the heart. Our study uncovered the molecular mechanisms of Cd in contributing cardiac metabolic disruption by inhibiting HIF1A degradation and increasing GLUT1 transcriptional expression. Inhibition of HIF1A could be a potential strategy to ameliorate Cd-mediated cardiac metabolic disorders and Cd-related cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Medical Physiology, School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, China; Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Wendan Zheng
- Department of Medical Physiology, School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, China; Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Shiyu Sun
- Department of Medical Physiology, School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, China; Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Yang Du
- Department of Personnel, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Wenjuan Xu
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering Laboratory for Health Management, Ji'nan, Shandong, China
| | - Zongguo Sun
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Fuhong Liu
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Manzhi Wang
- Department of Hematology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Zuohui Zhao
- Department of Pediatric Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Ju Liu
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Qiang Liu
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Laboratory of Translational Medicine in Microvascular Regulation, Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Ji'nan, Shandong, China.
| |
Collapse
|
45
|
Nakatsu Y, Matsunaga Y, Nakanishi M, Yamamotoya T, Sano T, Kanematsu T, Asano T. Prolyl isomerase Pin1 in skeletal muscles contributes to systemic energy metabolism and exercise capacity through regulating SERCA activity. Biochem Biophys Res Commun 2024; 715:150001. [PMID: 38676996 DOI: 10.1016/j.bbrc.2024.150001] [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: 04/04/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
The skeletal muscle is a pivotal organ involved in the regulation of both energy metabolism and exercise capacity. There is no doubt that exercise contributes to a healthy life through the consumption of excessive energy or the release of myokines. Skeletal muscles exhibit insulin sensitivity and can rapidly uptake blood glucose. In addition, they can undergo non-shivering thermogenesis through actions of both the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) and small peptide, sarcolipin, resulting in systemic energy metabolism. Accordingly, the maintenance of skeletal muscles is important for both metabolism and exercise. Prolyl isomerase Pin1 is an enzyme that converts the cis-trans form of proline residues and controls substrate function. We have previously reported that Pin1 plays important roles in insulin release, thermogenesis, and lipolysis. However, the roles of Pin1 in skeletal muscles remains unknown. To clarify this issue, we generated skeletal muscle-specific Pin1 knockout mice. Pin1 deficiency had no effects on muscle weights, morphology and ratio of fiber types. However, they showed exacerbated obesity or insulin resistance when fed with a high-fat diet. They also showed a lower ability to exercise than wild type mice did. We also found that Pin1 interacted with SERCA and elevated its activity, resulting in the upregulation of oxygen consumption. Overall, our study reveals that Pin1 in skeletal muscles contributes to both systemic energy metabolism and exercise capacity.
Collapse
Affiliation(s)
- Yusuke Nakatsu
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan.
| | - Yasuka Matsunaga
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mikako Nakanishi
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Takeshi Yamamotoya
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; Division of Diabetes and Metabolic Diseases, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchikami-cho, Itabashi-ku, 173-8610, Tokyo, Japan
| | - Tomomi Sano
- Department of Cell Biology, Aging Science, and Pharmacology, Faculty of Dental Science, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takashi Kanematsu
- Department of Cell Biology, Aging Science, and Pharmacology, Faculty of Dental Science, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoichiro Asano
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| |
Collapse
|
46
|
Jia Q, Zhang Y, Zhang B, An X. Reassessing type 2 diabetes in adolescents and its management strategies based on insulin resistance. Front Endocrinol (Lausanne) 2024; 15:1377918. [PMID: 38962677 PMCID: PMC11219588 DOI: 10.3389/fendo.2024.1377918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
With changes in lifestyle behaviors, including dietary structure and habits, the prevalence of Youth-onset Type 2 Diabetes Mellitus (YODM) has increased 2 to 3 times compared to 30 years ago. YODM patients experience complications earlier, progress faster, and exhibit more severe symptoms. However, limited and inconclusive direct evidence, coupled with poor patient compliance, poses challenges in the clinical management of YODM. Apart from the continuous decline in pancreatic β-cell function and quantity, tissue-specific insulin resistance (IR) is also a typical characteristic of YODM. The main mechanisms of IR in YODM involve different aspects such as obesity, dietary imbalance, abnormal substance metabolism, chronic inflammation, oxidative stress, and hormonal fluctuations during adolescence. For the comprehensive management of YODM, besides achieving good control of blood glucose levels, it may be necessary to apply the most appropriate methods considering the uniqueness of the patient population and the specifics of the disease. Early identification and detection of the disease are crucial. Precise screening of patients with well-functioning pancreatic insulin β-cells, primarily characterized by IR and obesity, represents the population most likely to achieve diabetes remission or reversal through lifestyle modifications, medications, or even surgical interventions. Additionally, considering potential emotional disorders or the impact of adolescent hormones in these patients, health education for patients and caregivers is essential to make them aware of the long-term benefits of well-controlled blood glucose. In conclusion, adopting comprehensive management measures to achieve diabetes remission or reversal is the ideal goal. Controlling high blood glucose, obesity, and other risk factors related to diabetes complications is the next priority to delay the occurrence and progression of complications. A comprehensive perspective on IR provides insights and references for identifying YODM and its management strategies.
Collapse
Affiliation(s)
- QianYou Jia
- Department of Pediatrics, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - YanMin Zhang
- Department of Pediatrics, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - BaoFeng Zhang
- Department of Pediatrics, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - XueDong An
- Department of Endocrinology & Diabetes Vascular Function Laboratory, Guang’anmen Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
47
|
Tokunaga W, Nagano N, Matsuda K, Nakazaki K, Shimizu S, Okuda K, Aoki R, Fuwa K, Murakami H, Morioka I. Efficacy of Human Recombinant Growth Hormone in Females of a Non-Obese Hyperglycemic Mouse Model after Birth with Low Birth Weight. Int J Mol Sci 2024; 25:6294. [PMID: 38928001 PMCID: PMC11203808 DOI: 10.3390/ijms25126294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
We examined whether the administration of growth hormone (GH) improves insulin resistance in females of a non-obese hyperglycemic mouse model after birth with low birth weight (LBW), given that GH is known to increase muscle mass. The intrauterine Ischemia group underwent uterine artery occlusion for 15 min on day 16.5 of gestation. At 4 weeks of age, female mice in the Ischemia group were divided into the GH-treated (Ischemia-GH) and non-GH-treated (Ischemia) groups. At 8 weeks of age, the glucose metabolism, muscle pathology, and metabolome of liver were assessed. The insulin resistance index improved in the Ischemia-GH group compared with the Ischemia group (p = 0.034). The percentage of type 1 muscle fibers was higher in the Ischemia-GH group than the Ischemia group (p < 0.001); the muscle fiber type was altered by GH. In the liver, oxidative stress factors were reduced, and ATP production was increased in the Ischemia-GH group compared to the Ischemia group (p = 0.014), indicating the improved mitochondrial function of liver. GH administration is effective in improving insulin resistance by increasing the content of type 1 muscle fibers and improving mitochondrial function of liver in our non-obese hyperglycemic mouse model after birth with LBW.
Collapse
Affiliation(s)
- Wataru Tokunaga
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | - Nobuhiko Nagano
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | - Kengo Matsuda
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | - Kimitaka Nakazaki
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | - Shoichi Shimizu
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | - Koh Okuda
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | - Ryoji Aoki
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | - Kazumasa Fuwa
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| | | | - Ichiro Morioka
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo 173-8610, Japan; (W.T.); (K.M.); (K.N.); (S.S.); (K.O.); (R.A.); (K.F.); (I.M.)
| |
Collapse
|
48
|
Rochowski MT, Jayathilake K, Balcerak JM, Tamil Selvan M, Gunasekara S, Rudd J, Miller C, Lacombe VA. Alterations of whole body glucose metabolism in a feline SARS-CoV-2 infection model. Am J Physiol Regul Integr Comp Physiol 2024; 326:R499-R506. [PMID: 38574344 PMCID: PMC11381005 DOI: 10.1152/ajpregu.00228.2023] [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: 10/04/2023] [Revised: 03/06/2024] [Accepted: 03/30/2024] [Indexed: 04/06/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been especially devastating to patients with comorbidities, including metabolic and cardiovascular diseases. Elevated blood glucose during SARS-CoV-2 infection increased mortality of patients with COVID-19, although the mechanisms are not well understood. It has been previously demonstrated that glucose transport and utilization is a crucial pathway for other highly infectious RNA viruses. Thus, we hypothesized that SARS-CoV-2 infection could lead to alterations in cellular and whole body glucose metabolism. Specific pathogen-free domestic cats were intratracheally inoculated with USA-WA1/2020 (wild-type) SARS-CoV-2 or vehicle-inoculated, then euthanized at 4- and 8-days postinoculation (dpi). Blood glucose and cortisol concentrations were elevated at 4 and 8 dpi. Blood ketones, insulin, and angiotensin II concentrations remained unchanged throughout the experimental timeline. SARS-CoV-2 RNA was detected in the lung and heart, without changes in angiotensin-converting enzyme 2 (ACE2) RNA expression. In the lung, SARS-CoV-2 infection increased glucose transporter 1 (GLUT1) protein levels at 4 and 8 dpi, whereas GLUT4 level was only upregulated at 8 dpi. In the heart, GLUT-1 and -4 protein levels remained unchanged. Furthermore, GLUT1 level was upregulated in the skeletal muscle at 8 dpi, and AMPK was activated in the hearts of infected cats. SARS-CoV-2 infection increased blood glucose concentration and pulmonary GLUT protein levels. These findings suggest that SARS-CoV-2 infection induces metabolic reprogramming primarily in the lung to support viral replication. Furthermore, this translational feline model mimicked human COVID-19 and could be used to explore novel therapeutic targets to treat metabolic disease during SARS-CoV-2 infection.NEW & NOTEWORTHY Our study on a feline model of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, mirroring human COVID-19, revealed alterations in whole body and cellular glucose metabolism. Infected cats developed mild hyperglycemia, increased protein levels of glucose transporters in the lung, and AMPK activation in the heart. These findings suggest that SARS-CoV-2 infection induces metabolic reprogramming in the cardiorespiratory system to support viral replication. Understanding these mechanisms could lead to novel antiviral therapeutic strategies.
Collapse
Affiliation(s)
- Matthew T Rochowski
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
- Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States
| | - Kaushalya Jayathilake
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - John-Michael Balcerak
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Miruthula Tamil Selvan
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Sachithra Gunasekara
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Jennifer Rudd
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Craig Miller
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Véronique A Lacombe
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, United States
- Harold Hamm Diabetes Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
49
|
Małkowska P. Positive Effects of Physical Activity on Insulin Signaling. Curr Issues Mol Biol 2024; 46:5467-5487. [PMID: 38920999 PMCID: PMC11202552 DOI: 10.3390/cimb46060327] [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: 04/23/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Physical activity is integral to metabolic health, particularly in addressing insulin resistance and related disorders such as type 2 diabetes mellitus (T2DM). Studies consistently demonstrate a strong association between physical activity levels and insulin sensitivity. Regular exercise interventions were shown to significantly improve glycemic control, highlighting exercise as a recommended therapeutic strategy for reducing insulin resistance. Physical inactivity is closely linked to islet cell insufficiency, exacerbating insulin resistance through various pathways including ER stress, mitochondrial dysfunction, oxidative stress, and inflammation. Conversely, physical training and exercise preserve and restore islet function, enhancing peripheral insulin sensitivity. Exercise interventions stimulate β-cell proliferation through increased circulating levels of growth factors, further emphasizing its role in maintaining pancreatic health and glucose metabolism. Furthermore, sedentary lifestyles contribute to elevated oxidative stress levels and ceramide production, impairing insulin signaling and glucose metabolism. Regular exercise induces anti-inflammatory responses, enhances antioxidant defenses, and promotes mitochondrial function, thereby improving insulin sensitivity and metabolic efficiency. Encouraging individuals to adopt active lifestyles and engage in regular exercise is crucial for preventing and managing insulin resistance and related metabolic disorders, ultimately promoting overall health and well-being.
Collapse
Affiliation(s)
- Paulina Małkowska
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland
| |
Collapse
|
50
|
Peng Q, Zeng W. The protective role of endothelial GLUT1 in ischemic stroke. Brain Behav 2024; 14:e3536. [PMID: 38747733 PMCID: PMC11095318 DOI: 10.1002/brb3.3536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVE To provide thorough insight on the protective role of endothelial glucose transporter 1 (GLUT1) in ischemic stroke. METHODS We comprehensively review the role of endothelial GLUT1 in ischemic stroke by narrating the findings concerning biological characteristics of GLUT1 in brain in depth, summarizing the changes of endothelial GLUT1 expression and activity during ischemic stroke, discussing how GLUT1 achieves its neuroprotective effect via maintaining endothelial function, and identifying some outstanding blind spots in current studies. RESULTS Endothelial GLUT1 maintains persistent high glucose and energy requirements of the brain by transporting glucose through the blood-brain barrier, which preserves endothelial function and is beneficial to stroke prognosis. CONCLUSION This review underscores the potential involvement of GLUT1 trafficking, activity modulation, and degradation, and we look forward to more clinical and animal studies to illuminate these mechanisms.
Collapse
Affiliation(s)
- Qiwei Peng
- Department of Critical Care Medicine, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology)Ministry of EducationWuhanChina
| | - Weiqi Zeng
- Department of NeurologyThe First People's Hospital of FoshanFoshanChina
| |
Collapse
|