1
|
Zhi S, Wang J, Wang Y, Li Y, Zhao M, Yang L, Qin C, Yan X, Nie G. Molecular characterization of AMP-activated protein kinase (AMPK) α1/α2 from Cyprinus carpio and its roles in glucolipid metabolism and immune response. Int J Biol Macromol 2025; 303:140736. [PMID: 39920952 DOI: 10.1016/j.ijbiomac.2025.140736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/26/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
AMPKα1 and AMPKα2, key kinases in regulating energy homeostasis, have not been previously cloned or characterized in common carp (Cyprinus carpio). This study identified the open reading frame (ORF) sequences of ampkα1 (1722 bp, encoding 573 amino acids) and ampkα2 (1659 bp, encoding 552 amino acids) through homologous cloning. Sequence alignment and phylogenetic analysis showed a high similarity of both genes with fish homologs. Expression analysis revealed that ampkα1 and ampkα2 are widely expressed across tissues in carp, with ampkα1 highly expressed in the gonads and ampkα2 in the heart. Fasting significantly reduced ampkα1 expression in the heart, adipose tissue, and foregut but increased it in the hindgut and white muscle. Similarly, ampkα2 expression decreased in the hypothalamus and muscle during fasting, with an increase in the midgut. Glucose tolerance tests showed dynamic regulation of ampkα1 and ampkα2, with initial downregulation followed by upregulation in the hepatopancreas, red muscle, and brain. High-glucose and high-fat diets significantly increased ampkα1 and ampkα2 expression in multiple tissues. Insulin and glucagon treatment induced time-dependent changes in both genes in hepatocytes, while Aeromonas hydrophila infection, LPS, and Poly (I:C) stimulation upregulated ampkα1 and ampkα2 in immune-related tissues. Knockdown of ampkα2, but not ampkα1, reduced glut1b mRNA levels, while both knockdowns of ampkα1 and ampkα2 promoted the expression of gsk3β, pygm, acc, fas, srebp, cs, and pro-inflammatory cytokines, suggesting their involvement in metabolic and immune regulation in carp.
Collapse
Affiliation(s)
- Shaoyang Zhi
- Aquatic Animal Nutrition and Feed Research Laboratory, College of Fisheries, Henan Normal University, Xinxiang 453007, PR China
| | - Junli Wang
- College of Life Science, Henan Normal University, Xinxiang 453007, PR China.
| | - Yiran Wang
- College of Life Science, Henan Normal University, Xinxiang 453007, PR China
| | - Yijie Li
- Aquatic Animal Nutrition and Feed Research Laboratory, College of Fisheries, Henan Normal University, Xinxiang 453007, PR China
| | - Mengjuan Zhao
- Aquatic Animal Nutrition and Feed Research Laboratory, College of Fisheries, Henan Normal University, Xinxiang 453007, PR China
| | - Liping Yang
- Aquatic Animal Nutrition and Feed Research Laboratory, College of Fisheries, Henan Normal University, Xinxiang 453007, PR China
| | - Chaobin Qin
- Aquatic Animal Nutrition and Feed Research Laboratory, College of Fisheries, Henan Normal University, Xinxiang 453007, PR China
| | - Xiao Yan
- Aquatic Animal Nutrition and Feed Research Laboratory, College of Fisheries, Henan Normal University, Xinxiang 453007, PR China
| | - Guoxing Nie
- Aquatic Animal Nutrition and Feed Research Laboratory, College of Fisheries, Henan Normal University, Xinxiang 453007, PR China.
| |
Collapse
|
2
|
Kielbowski K, Bratborska AW, Bakinowska E, Pawlik A. Sirtuins as therapeutic targets in diabetes. Expert Opin Ther Targets 2025:1-19. [PMID: 40116767 DOI: 10.1080/14728222.2025.2482563] [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: 08/28/2024] [Revised: 03/01/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION Sirtuins (SIRTs) are NAD+-dependent deacetylases that mediate post-translational modifications of proteins. Seven members of the SIRT family have been identified in mammals. Importantly, SIRTs interact with numerous metabolic and inflammatory pathways. Thus, researchers have investigated their role in metabolic and inflammatory disorders. AREAS COVERED In this review, we comprehensively discuss the involvement of SIRTs in the processes of pancreatic β-cell dysfunction, glucose tolerance, insulin secretion, lipid metabolism, and adipocyte functions. In addition, we describe the current evidence regarding modulation of the expression and activity of SIRTs in diabetes, diabetic complications, and obesity. EXPERT OPINION The development of specific SIRT activators and inhibitors that exhibit high selectivity toward specific SIRT isoforms remains a major challenge. This involves the need to elucidate the physiological pathways involving SIRTs, as well as their important role in the development of metabolic disorders. Molecular modeling techniques will be helpful to develop new compounds that modulate the activity of SIRTs, which may contribute to the preparation of new drugs that selectively target specific SIRTs. SIRTs hold promise as potential targets in metabolic disease, but there is much to learn about specific modulators and the final answers will await clinical trials.
Collapse
Affiliation(s)
- Kajetan Kielbowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
3
|
Pang ZD, Wang Y, Han MZ, She G, Sun X, Bai RY, Lai BC, Zhang Y, Du XJ, Shyy JYJ, Deng XL. AMPK phosphorylation of K Ca2.3 alleviates angiotensin II-induced endothelial dysfunction. Biochem Pharmacol 2025; 236:116880. [PMID: 40112931 DOI: 10.1016/j.bcp.2025.116880] [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: 07/24/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
The endothelial small-conductance calcium-activated potassium channels (KCa2.3) are indispensable for endothelium-dependent hyperpolarization (EDH) response, mainly in resistance arteries. We recently demonstrated in diet-induced obese mice that adenosine monophosphate-activated protein kinase (AMPK) upregulates endothelial KCa2.3 expression and improves endothelial function. However, the molecular mechanism of regulation of KCa2.3 by AMPK remains less explored. Using techniques of bioinformatics, molecular biology and wire myograph system, we examined KCa2.3 phosphorylation by AMPK in human umbilical vein endothelial cells (HUVECs), human embryonic kidney 293 (HEK-293T) cells and second-order mesenteric resistance arteries from angiotensin II-induced hypertensive mice. In HUVECs, treatment with activators of AMPK (AICAR, metformin, and MK-8722) significantly increased phosphorylation of KCa2.3 Thr106 (human), which was antagonized by AMPK inhibitor compound C. In HEK-293T cells, KCa2.3 current was enhanced by AMPK activation or phosphomimetic mutant KCa2.3 (T106D), which was abolished after de-phosphomimetic mutant (T106A) or deletion of KCa2.3 of Thr106 site (T106Del). In mice with angiotensin II infusion, 2-week treatment with AICAR or overexpressing phosphomimetic mutant KCa2.3 Thr107D (mouse) restored KCa2.3-mediated EDH-dependent relaxation in mesenteric resistance arteries together with reversal of early phase hypertension. Our study demonstrates for the first time that AMPK activation mediates KCa2.3 phosphorylation in endothelial cells with enhanced channel activity. This effect ameliorates endothelial dysfunction of mesenteric resistance arteries and alleviates angiotensin II-induced early phase hypertension in mice.
Collapse
Affiliation(s)
- Zheng-Da Pang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Yan Wang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Meng-Zhuan Han
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Gang She
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Xia Sun
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Ru-Yue Bai
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Bao-Chang Lai
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Yi Zhang
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - Xiao-Jun Du
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613, USA.
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, Cardiovascular Research Centre, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061 Shaanxi, China.
| |
Collapse
|
4
|
Widmer A, Lillegard K, Wood K, Robles M, Fan R, Ye F, Koethe JR, Silver HJ. Consumption of tree nuts as snacks stimulates changes in plasma fatty acid profiles and adipose tissue gene expression in young adults at risk for metabolic syndrome. Clin Nutr 2025; 48:25-34. [PMID: 40117963 DOI: 10.1016/j.clnu.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/10/2025] [Accepted: 03/01/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND AND AIMS The prevalence of metabolic syndrome has been increasing in young adults, concomitant with the occurrence of increased abdominal adiposity. We previously reported that consuming tree nuts, as replacement for typical high-carbohydrate snacks, reduces visceral fat and waist circumference in young adults who have one or more metabolic syndrome risk factors. We aimed to investigate the effects of tree nuts snack consumption on plasma and adipose tissue fatty acid profiles along with changes in the expression of adipose tissue genes involved in thermogenesis, glycemia, adipocyte signaling, lipolysis, and immunity. METHODS A randomized parallel-arm 16-week intervention trial was conducted in 84 adults aged 22-36 years. Participants in both groups were provided with caloric goals for weight maintenance, daily menus, and pre-portioned snacks at every other week visits with study registered dietitians. Changes in dietary fatty acid intakes, plasma and abdominal subcutaneous adipose tissue (SAT) triglycerides fatty acid profiles using gas-liquid chromatography, and the expression of 241 genes in abdominal SAT were evaluated. RESULTS Consuming tree nuts snacks increased mono- and polyunsaturated fatty acid intakes yielding a 9-fold greater dietary unsaturated to saturated fat ratio. The tree nuts snack group also had significantly greater improvements in plasma 16:1/16:0 ratio; plasma phospholipids oleic and gamma linolenic acid content; plasma diglycerides, triglycerides, and cholesterol esters oleic acid content; and total plasma monounsaturated fatty acids. While abdominal SAT only showed trends for increased oleic acid content and unsaturated to saturated fat ratio, the tree nuts snacks participants had altered expression of 13 genes in abdominal SAT that have roles in nutrient sensing, energy homeostasis, and vulnerability to obesity. CONCLUSIONS Replacing typical high-carbohydrate snacks with tree nuts results in more favorable dietary, plasma, and adipose tissue fatty acid profiles that could aid in preventing the development of excess adiposity and cardiometabolic disease states including metabolic syndrome. CLINICAL TRIAL REGISTRY This trial was registered at clinicaltrials.gov NCT03969264.
Collapse
Affiliation(s)
- Annaliese Widmer
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Kate Lillegard
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Kate Wood
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Michelle Robles
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA
| | - Run Fan
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, TN, USA; Vanderbilt University Medical Center, Department of Epidemiology, Nashville, TN, USA
| | - Fei Ye
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, TN, USA; Vanderbilt University Medical Center, Department of Epidemiology, Nashville, TN, USA
| | - John R Koethe
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, USA; Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA
| | - Heidi J Silver
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, USA; Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA.
| |
Collapse
|
5
|
Wöhlbrand L, Dörries M, Siani R, Medrano-Soto A, Schnaars V, Schumacher J, Hilbers C, Thies D, Kube M, Reinhardt R, Schloter M, Saier MH, Winklhofer M, Rabus R. Key role of Desulfobacteraceae in C/S cycles of marine sediments is based on congeneric catabolic-regulatory networks. SCIENCE ADVANCES 2025; 11:eads5631. [PMID: 40053579 PMCID: PMC11887813 DOI: 10.1126/sciadv.ads5631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Marine sediments are highly bioactive habitats, where sulfate-reducing bacteria contribute substantially to seabed carbon cycling by oxidizing ~77 Tmol Corg year-1. This remarkable activity is largely attributable to the deltaproteobacterial family Desulfobacteraceae of complete oxidizers (to CO2), which our biogeography focused meta-analysis verified as cosmopolitan. However, the catabolic/regulatory networks underlying this ecophysiological feat at the thermodynamic limit are essentially unknown. Integrating cultivation-based (80 conditions) proteogenomics of six representative Desulfobacteraceae spp., we identify molecular commonalities explaining the family's environmental relevance and success. Desulfobacteraceae genomes are specifically enriched in substrate uptake, degradation capacities, and regulatory functions including fine-tuned sulfate uptake. Conserved gene arrangements and shared regulatory patterns translate into strikingly similar (sub-)proteome profiles. From 319 proteins, we constructed a meta-network for catabolizing 35 substrates. Therefrom, we defined a Desulfobacteraceae characteristic gene subset, which we found prevalent in metagenomes of organic-rich, marine sediments. These genes are promising targets to advance our mechanistic understanding of Desulfobacteraceae-driven biogeochemical processes in marine sediments and beyond.
Collapse
Affiliation(s)
- Lars Wöhlbrand
- Institute for Chemistry and Biology of the Marine Environment (ICBM), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Marvin Dörries
- Institute for Chemistry and Biology of the Marine Environment (ICBM), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
- Helmholtz Institute for Functional Marine Biodiversity at the Carl von Ossietzky Universität Oldenburg (HIFMB), Oldenburg, Germany
| | - Roberto Siani
- Institute for Comparative Microbiome Analysis (COMI), Department of Environmental Sciences, Helmholtz Zentrum München, Oberschleißheim, Munich, Germany
- Chair for Environmental Microbiology, School of Life Sciences, Technical University Munich, Freising, Germany
| | - Arturo Medrano-Soto
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, San Diego, CA, USA
| | - Vanessa Schnaars
- Institute for Chemistry and Biology of the Marine Environment (ICBM), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Julian Schumacher
- Institute for Chemistry and Biology of the Marine Environment (ICBM), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Christina Hilbers
- Institute for Chemistry and Biology of the Marine Environment (ICBM), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Daniela Thies
- Max Planck Institute for Marine Microbiology, Bremen, Germany
| | - Michael Kube
- Integrative Infection Biology Crops-Livestocks, Faculty of Agricultural Sciences, University Hohenheim, Stuttgart, Germany
| | | | - Michael Schloter
- Institute for Comparative Microbiome Analysis (COMI), Department of Environmental Sciences, Helmholtz Zentrum München, Oberschleißheim, Munich, Germany
- Chair for Environmental Microbiology, School of Life Sciences, Technical University Munich, Freising, Germany
| | - Milton H. Saier
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, San Diego, CA, USA
| | - Michael Winklhofer
- Institute of Biology and Environmental Sciences (IBU), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Ralf Rabus
- Institute for Chemistry and Biology of the Marine Environment (ICBM), School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| |
Collapse
|
6
|
Lee JH, Jung IR, Tu-Sekine B, Jin S, Anokye-Danso F, Ahima RS, Kim SF. Loss of Skeletal Muscle Inositol Polyphosphate Multikinase Disrupts Glucose Regulation and Limits Exercise Capacity. Int J Mol Sci 2025; 26:2395. [PMID: 40141045 PMCID: PMC11942489 DOI: 10.3390/ijms26062395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Inositol phosphates are critical signaling messengers involved in a wide range of biological pathways, and inositol polyphosphate multikinase (IPMK) functions as a rate-limiting enzyme for inositol polyphosphate metabolism. IPMK has been implicated in cellular metabolism, but its function at the systemic level is still poorly understood. Since skeletal muscle is a major contributor to energy homeostasis, we have developed a mouse model in which skeletal muscle IPMK is specifically deleted and examined how a loss of IPMK affects whole-body metabolism. Here, we report that skeletal-muscle-specific IPMK knockout mice exhibited a ~12% increase in body weight compared to WT controls (p < 0.05). These mice also showed a significantly impaired glucose tolerance, as indicated by their ~50% higher blood glucose levels during GTT. Additionally, exercise capacity was reduced by ~45% in IPMK-MKO mice, demonstrating a decline in endurance. Moreover, these metabolic alterations were accompanied by a 2.5-fold increase in skeletal muscle triglyceride accumulation, suggesting impaired lipid metabolism. Further analysis revealed that IPMK-deficient myocytes exhibited 30% lower β-oxidation rates. Thus, our results suggest that IPMK mediates whole-body metabolism by regulating muscle metabolism and may be potentially targeted for the treatment of metabolic syndromes.
Collapse
Affiliation(s)
| | | | | | | | | | - Rexford S. Ahima
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, MD 21218, USA; (J.-H.L.); (I.-R.J.); (B.T.-S.); (F.A.-D.)
| | - Sangwon F. Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, MD 21218, USA; (J.-H.L.); (I.-R.J.); (B.T.-S.); (F.A.-D.)
| |
Collapse
|
7
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [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: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
8
|
Kujawowicz K, Mirończuk-Chodakowska I, Cyuńczyk M, Witkowska AM. Malnutrition Risk in Older Adults: Evaluating the Diagnostic Relevance of Serum Biomarkers: SIRT-1, CCK-8, Melatonin, and Total Antioxidant Capacity (TAC). Nutrients 2025; 17:726. [PMID: 40005054 PMCID: PMC11858257 DOI: 10.3390/nu17040726] [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: 01/29/2025] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Addressing the risk of malnutrition at an early stage is crucial to preventing its development, which can have a detrimental impact on physical and mental health status. This study investigates the potential role of biochemical biomarkers such as sirtuin 1 (SIRT-1), melatonin, cholecystokinin-8 (CCK-8), and total antioxidant capacity (TAC) in identifying the risk of malnutrition. Methods: This cross-sectional study assessed malnutrition risk in 153 community-dwelling older adults using the Mini Nutritional Assessment (MNA). Serum levels of SIRT-1, melatonin, and CCK-8 were analyzed with enzyme-linked immunosorbent assay (ELISA), and total antioxidant capacity (TAC) was measured using the ferric reducing ability of plasma (FRAP) method. Results: Serum levels of TAC and CCK-8 were significantly positively correlated with grip strength and visceral adipose tissue, with TAC levels also showing associations with appendicular skeletal muscle mass index (ASMI), total body water, total energy expenditure, fat-free mass index, and fat mass index (p < 0.001). CCK-8 emerged as a strong predictor of malnutrition risk (AUC = 0.58 in females, AUC = 0.64 in males), whereas SIRT-1 (AUC = 0.57 for both sexes), melatonin (AUC = 0.46 for females, AUC = 0.51 for males), and TAC (AUC = 0.42 for females, AUC = 0.54 for males) exhibited weaker predictive abilities. A multivariate model incorporating CCK-8 demonstrated excellent predictive accuracy (AUC = 0.84, 95% CI: 0.77-0.90) and indicated a potential association between elevated CCK-8 levels and a higher risk of malnutrition. Conclusions: In conclusion, this study highlights the effectiveness of a multi-parameter model incorporating CCK-8 as a reliable approach for assessing malnutrition risk in older adults, offering a comprehensive evaluation of the condition. However, further research is needed to confirm its applicability and accuracy in diverse elderly populations and clinical settings.
Collapse
Affiliation(s)
- Karolina Kujawowicz
- Department of Food Biotechnology, Medical University of Białystok, ul. Szpitalna 37, 15-285 Białystok, Poland; (I.M.-C.); (M.C.); (A.M.W.)
| | | | | | | |
Collapse
|
9
|
da Silva RS, de Paiva IHR, Mendonça IP, de Souza JRB, Lucena-Silva N, Peixoto CA. Anorexigenic and anti-inflammatory signaling pathways of semaglutide via the microbiota-gut--brain axis in obese mice. Inflammopharmacology 2025; 33:845-864. [PMID: 39586940 DOI: 10.1007/s10787-024-01603-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: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/27/2024]
Abstract
Our study focused on a mouse model of obesity induced by a high-fat diet (HFD). We administered Semaglutide intraperitoneally (Ozempic ®-0.05 mg/Kg-translational dose) every seven days for six weeks. HFD-fed mice had higher blood glucose, lipid profile, and insulin resistance. Moreover, mice fed HFD showed high gut levels of TLR4, NF-kB, TNF-α, IL-1β, and nitrotyrosine and low levels of occludin, indicating intestinal inflammation and permeability, culminating in higher serum levels of IL-1β and LPS. Treatment with semaglutide counteracted the dyslipidemia and insulin resistance, reducing gut and serum inflammatory markers. Structural changes in gut microbiome were determined by 16S rRNA sequencing. Semaglutide reduced the relative abundance of Firmicutes and augmented that of Bacteroidetes. Meanwhile, semaglutide dramatically changed the overall composition and promoted the growth of acetate-producing bacteria (Bacteroides acidifaciens and Blautia coccoides), increasing hypothalamic acetate levels. Semaglutide intervention increased the number of hypothalamic GLP-1R+ neurons that mediate endogenous action on feeding and energy. In addition, semaglutide treatment reversed the hypothalamic neuroinflammation HDF-induced decreasing TLR4/MyD88/NF-κB signaling and JNK and AMPK levels, improving the hypothalamic insulin resistance. Also, semaglutide modulated the intestinal microbiota, promoting the growth of acetate-producing bacteria, inducing high levels of hypothalamic acetate, and increasing GPR43+ /POMC+ neurons. In the ARC, acetate activated the GPR43 and its downstream PI3K-Akt pathway, which activates POMC neurons by repressing the FoxO-1. Thus, among the multifactorial effectors of hypothalamic energy homeostasis, possibly higher levels of acetate derived from the intestinal microbiota contribute to reducing food intake.
Collapse
Affiliation(s)
- Rodrigo Soares da Silva
- Laboratory of Ultrastructure, Laboratório de Ultraestrutura, Aggeu Magalhães Institute (IAM), FIOCRUZ, Av. Moraes Rego S/N, Recife, PE, CEP 50670-420, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - Igor Henrique Rodrigues de Paiva
- Laboratory of Ultrastructure, Laboratório de Ultraestrutura, Aggeu Magalhães Institute (IAM), FIOCRUZ, Av. Moraes Rego S/N, Recife, PE, CEP 50670-420, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - Ingrid Prata Mendonça
- Laboratory of Ultrastructure, Laboratório de Ultraestrutura, Aggeu Magalhães Institute (IAM), FIOCRUZ, Av. Moraes Rego S/N, Recife, PE, CEP 50670-420, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | | | - Norma Lucena-Silva
- Laboratory of Immunogenetics, Aggeu Magalhães Institute (IAM), Recife, PE, Brazil
| | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Laboratório de Ultraestrutura, Aggeu Magalhães Institute (IAM), FIOCRUZ, Av. Moraes Rego S/N, Recife, PE, CEP 50670-420, Brazil.
| |
Collapse
|
10
|
Freire-Agulleiro Ó, Estévez-Salguero Á, Ferreira V, Holleman CL, García-Currás J, González-García I, Nogueiras R, Tena-Sempere M, García-Cáceres C, Diéguez C, López M. SF1-specific deletion of the energy sensor AMPKγ2 induces obesity. Mol Metab 2025; 92:102091. [PMID: 39746605 PMCID: PMC11782900 DOI: 10.1016/j.molmet.2024.102091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE AMP-activated protein kinase (AMPK) is a heterotrimer complex consisting of a catalytic α subunit (α1, α2) with a serine/threonine kinase domain, and two regulatory subunits, β (β1, β2) and γ (γ1, γ2, γ3), encoded by different genes. In the hypothalamus, AMPK plays a crucial role in regulating energy balance, including feeding, energy expenditure, peripheral glucose and lipid metabolism. However, most research on hypothalamic AMPK has concentrated on the catalytic subunits AMPKα1 and AMPKα2, with little focus on the regulatory subunits. METHODS To fill this gap of knowledge, we investigated the effects of selectively deleting the regulatory isoform AMPKγ2, which is a primary "energy sensor", in steroidogenic factor 1 (SF1) neurons of the ventromedial hypothalamic nucleus (VMH). Complete metabolic phenotyping and molecular analyses in brown adipose tissue (BAT), white adipose tissue (WAT) and liver were carried out. RESULTS Our findings reveal that, in contrast to the obesity-protective effect of the genetic deletion of AMPKα subunits, the loss of AMPKγ2 in SF1 neurons leads to a sex-independent and feeding-independent obesity-prone phenotype due to decreased thermogenesis in brown adipose tissue (BAT) and reduced browning of WAT, resulting in lower energy expenditure. Additionally, SF1-Cre AMPKγ2 mice exhibit hepatic lipid accumulation, but surprisingly maintain normal glucose homeostasis. CONCLUSIONS Overall, these results highlight the distinct roles of AMPK subunits within the hypothalamus.
Collapse
Affiliation(s)
- Óscar Freire-Agulleiro
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Ánxela Estévez-Salguero
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Vitor Ferreira
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Cassie Lynn Holleman
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Julia García-Currás
- Biostatech Advice, Training and Innovation in Biostatistics, S.L, Ames, 15895, Spain
| | - Ismael González-García
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Rubén Nogueiras
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Universitario Reina Sofía, Córdoba, 14004, Spain
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336, Munich, Germany
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain.
| |
Collapse
|
11
|
Yoshimura Y, Matsui T, Kaneko N, Kobayashi I. Digestion and absorption of triacetin, a short-chain triacylglycerol. Lipids 2025. [PMID: 39891375 DOI: 10.1002/lipd.12433] [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: 08/17/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Triacylglycerol (TG) is categorized into long-, medium-, and short-chain TG (SCTG). While the digestion of long- and medium-chain TG is well established, the process for SCTG remains unclear. This study investigated SCTG digestion by administering 2 mmol of triacetin to rats and analyzing acetin, acetic acid, and glycerol levels in the portal blood and small intestine. Triacetin was fully degraded in the upper gastrointestinal tract and absorbed as acetic acid and glycerol. Glycerol influx into the liver promoted gluconeogenesis, while acetate activated AMPK, resulting in the suppression of fatty acid synthesis-related genes and the upregulation of fatty acid β-oxidation-related genes. These findings demonstrate that triacetin not only serves as a substrate for energy metabolism but also regulates hepatic gene expression, highlighting its dual role as both a metabolic substrate and signaling molecule. Triacetin thus shows potential as a dietary modulator for improving metabolic health.
Collapse
Affiliation(s)
| | - Tomoka Matsui
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| | - Nagisa Kaneko
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| | - Ikuha Kobayashi
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| |
Collapse
|
12
|
Ding Y, Yang H, Gao J, Tang C, Peng YY, Ma XM, Li S, Wang HY, Lu XM, Wang YT. Synaptic-mitochondrial transport: mechanisms in neural adaptation and degeneration. Mol Cell Biochem 2025:10.1007/s11010-025-05209-y. [PMID: 39841406 DOI: 10.1007/s11010-025-05209-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/04/2025] [Indexed: 01/23/2025]
Abstract
Synaptic plasticity is the basis for the proper functioning of the central nervous system. Synapses are the contact points between neurons and are crucial for information transmission, the structure and function of synapses change adaptively based on the different activities of neurons, thus affecting processes such as learning, memory, and neural development and repair. Synaptic activity requires a large amount of energy provided by mitochondria. Mitochondrial transport proteins regulate the positioning and movement of mitochondria to maintain normal energy metabolism. Recent studies have shown a close relationship between mitochondrial transport proteins and synaptic plasticity, providing a new direction for the study of adaptive changes in the central nervous system and new targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yang Ding
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huan Yang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jie Gao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Can Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yu-Yuan Peng
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Xin-Mei Ma
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
13
|
Wang Z, Zhu H, Xiong W. Metabolism and metabolomics in senescence, aging, and age-related diseases: a multiscale perspective. Front Med 2025:10.1007/s11684-024-1116-0. [PMID: 39821730 DOI: 10.1007/s11684-024-1116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
The pursuit of healthy aging has long rendered aging and senescence captivating. Age-related ailments, such as cardiovascular diseases, diabetes, and neurodegenerative disorders, pose significant threats to individuals. Recent studies have shed light on the intricate mechanisms encompassing genetics, epigenetics, transcriptomics, and metabolomics in the processes of senescence and aging, as well as the establishment of age-related pathologies. Amidst these underlying mechanisms governing aging and related pathology metabolism assumes a pivotal role that holds promise for intervention and therapeutics. The advancements in metabolomics techniques and analysis methods have significantly propelled the study of senescence and aging, particularly with the aid of multiscale metabolomics which has facilitated the discovery of metabolic markers and therapeutic potentials. This review provides an overview of senescence and aging, emphasizing the crucial role metabolism plays in the aging process as well as age-related diseases.
Collapse
Affiliation(s)
- Ziyi Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hongying Zhu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Wei Xiong
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| |
Collapse
|
14
|
Maharati A, Rajabloo Y, Moghbeli M. Molecular mechanisms of mTOR-mediated cisplatin response in tumor cells. Heliyon 2025; 11:e41483. [PMID: 39834411 PMCID: PMC11743095 DOI: 10.1016/j.heliyon.2024.e41483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025] Open
Abstract
Cisplatin (CDDP) is one of the main chemotherapeutic drugs that is widely used in many cancers. However, CDDP resistance is a frequent therapeutic challenge that reduces prognosis in cancer patients. Since, CDDP has noticeable side effects in normal tissues and organs, it is necessary to assess the molecular mechanisms associated with CDDP resistance to improve the therapeutic methods in cancer patients. Drug efflux, detoxifying systems, DNA repair mechanisms, and drug-induced apoptosis are involved in multidrug resistance in CDDP-resistant tumor cells. Mammalian target of rapamycin (mTOR), as a serine/threonine kinase has a pivotal role in various cellular mechanisms such as autophagy, metabolism, drug efflux, and cell proliferation. Although, mTOR is mainly activated by PI3K/AKT pathway, it can also be regulated by many other signaling pathways. PI3K/Akt/mTOR axis functions as a key modulator of drug resistance and unfavorable prognosis in different cancers. Regarding, the pivotal role of mTOR in CDDP response, in the present review we discussed the molecular mechanisms that regulate mTOR mediated CDDP response in tumor cells.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yasamin Rajabloo
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
16
|
Zhang J, Li N, Hu X. Metabolic Reprograming of Macrophages: A New Direction in Traditional Chinese Medicine for Treating Liver Failure. J Immunol Res 2024; 2024:5891381. [PMID: 39741958 PMCID: PMC11688140 DOI: 10.1155/jimr/5891381] [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: 12/19/2023] [Revised: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Acute liver failure (ALF) is a fulminant clinical syndrome that usually leads to multiple organ failure and high mortality. Macrophages play a crucial role in the initiation, development, and recovery of ALF. Targeting macrophages through immunotherapy holds significant promise as a therapeutic strategy. These cells exhibit remarkable plasticity, enabling them to differentiate into various subtypes based on changes in their surrounding microenvironment. M1-type macrophages are associated with a pro-inflammatory phenotype and primarily rely predominantly on glycolysis. In contrast, M2-type macrophages, which are characterized by anti-inflammatory phenotype, predominantly obtain their energy from oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO). Shifting macrophage metabolism from glycolysis to OXPHOS inhibits M1 macrophage activation and promotes M2 macrophage activation, thereby exerting anti-inflammatory and reparative effects. This study elucidates the relationship between macrophage activation and glucose metabolism reprograming from an immunometabolism perspective. A comprehensive literature review revealed that several signaling pathways may regulate macrophage polarization through energy metabolism, including phosphatidyl-inositol 3-kinase/protein kinase B (PI3K/AKT), mammalian target of rapamycin (mTOR)/hypoxia-inducible factor 1α (HIF-1α), nuclear factor-κB (NF-κB), and AMP-activated protein kinase (AMPK), which exhibit crosstalk with one another. Additionally, we systematically reviewed several traditional Chinese medicine (TCM) monomers that can modulate glucose metabolism reprograming and influence the polarization states of M1 and M2 macrophages. This review aimed to provide valuable insights that could contribute to the development of new therapies or drugs for ALF.
Collapse
Affiliation(s)
- Junli Zhang
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Na Li
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
17
|
Ramatchandirane M, Rajendran P, Athira MP, Suchiang K. Coniferaldehyde activates autophagy and enhances oxidative stress resistance and lifespan of Caenorhabditis elegans via par-4/aak-2/skn-1 pathway. Biogerontology 2024; 26:25. [PMID: 39674829 DOI: 10.1007/s10522-024-10163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Aging represents the gradual accumulation of alterations within an organism over time. The physical and chemical characteristics of our cells gradually change as we age, making it more difficult for our tissues and organs to self-regulate, regenerate, and maintain their structural and functional integrity. AMP- activated protein kinase (AMPK), a well-known sensor of cellular energy status acts as a central regulator of an integrated signalling network that control homeostasis, metabolism, stress resistance, cell survival and autophagy. Coniferaldehyde (CFA), a phenolic compound found in many edible plants, has multiple biological and pharmacological functions. Our findings demonstrated that 50 µM CFA could significantly activate autophagy and reduce oxidative stress, which enhanced the activity of antioxidant enzymes and increased resistance under oxidative stress. CFA treatment could efficiently decrease reactive oxygen species (ROS) levels and positively enhance the expression of antioxidant genes in Caenorhabditis elegans (C. elegans). On the other hand, CFA did not have any role in the lifespan extension of the several mutants linked to the AAK-2/AMPK pathway and it promotes SKN-1 (Skinhead-1) localization into the nucleus, which modulates downstream gene gst-4 (Glutathione S-transferase). In depth investigations revealed that CFA could lower oxidative stress and enhance the lifespan of C. elegans by activating the PAR-4/LKB-1-AAK-2/AMPK-SKN-1/NRF-2 pathway, with crucial involvement of bec-1 and lgg-1 genes for autophagy mediated lifespan extension. This study might contribute to understanding the interactions and mechanisms that allow natural compounds like CFA to treat age-related disorders among several species.
Collapse
Affiliation(s)
- Mahesh Ramatchandirane
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605014, India
| | - Ponsankaran Rajendran
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605014, India
| | - M P Athira
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605014, India
| | - Kitlangki Suchiang
- Department of Biochemistry, North Eastern Hill University, Shillong, Meghalaya, 793022, India.
| |
Collapse
|
18
|
Chen Y, Huang Y, Gan Q, Zhang W, Sun H, Zhu L, Wang W. Characterization of tea polysaccharides from Tieguanyin oolong tea and their hepatoprotective effects via AMP-activated protein kinase-mediated signaling pathways. J Food Sci 2024; 89:10064-10078. [PMID: 39636766 DOI: 10.1111/1750-3841.17575] [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: 08/30/2024] [Revised: 10/26/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
In the present study, we succeeded in extracting tea polysaccharide (TPS) from Tieguanyin oolong tea, and the TPS was characterized. TPS is an acidic heteropolysaccharide containing rhamnose, arabinose, galactose, glucose (Glc), xylose, mannose, galacturonic acid, and guluronic acid. We found that TPS supplementation partially reversed the elevated levels of serum alanine aminotransferase, total cholesterol, and low-density lipoprotein cholesterol in high-fat diet (HD)-induced nonalcoholic fatty liver disease (NAFLD) mice (p < 0.05), and hepatic steatosis and impaired Glc tolerance were also ameliorated. After HD intervention, the activity of Adenosine 5'-monophosphate-activated protein kinase (AMPK) and its downstream genes, including Sirtuin 1 (SIRT1), sterol regulatory element-binding protein-1c (SREBP1c), acetyl-coenzyme A carboxylase 1 (ACC1), and adipose triglyceride lipase (ATGL), was significantly inhibited (p < 0.05). TPS can increase the expression of these genes. The hepatoprotective effects of TPS in AMPK-/- mice almost completely disappeared. Moreover, the expression levels of SIRT1, SREBP1c, ACC1, and ATGL did not significantly change after TPS supplementation (p > 0.05). Therefore, our findings suggest that TPS protects the liver from hepatic glucolipid metabolism disorders in HD-induced NAFLD mice by activating AMPK-mediated signaling pathways.
Collapse
Affiliation(s)
- Yiqin Chen
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yanxin Huang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Department of Nutrition, The 95th Hospital of Putian, Putian, Fujian, China
| | - Qiaorong Gan
- Department of Hepatology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Wenhui Zhang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Han Sun
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Lingling Zhu
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenxiang Wang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
19
|
Smiles WJ, Ovens AJ, Oakhill JS, Kofler B. The metabolic sensor AMPK: Twelve enzymes in one. Mol Metab 2024; 90:102042. [PMID: 39362600 PMCID: PMC11752127 DOI: 10.1016/j.molmet.2024.102042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) is an evolutionarily conserved regulator of energy metabolism. AMPK is sensitive to acute perturbations to cellular energy status and leverages fundamental bioenergetic pathways to maintain cellular homeostasis. AMPK is a heterotrimer comprised of αβγ-subunits that in humans are encoded by seven individual genes (isoforms α1, α2, β1, β2, γ1, γ2 and γ3), permitting formation of at least 12 different complexes with personalised biochemical fingerprints and tissue expression patterns. While the canonical activation mechanisms of AMPK are well-defined, delineation of subtle, as well as substantial, differences in the regulation of heterogenous AMPK complexes remain poorly defined. SCOPE OF REVIEW Here, taking advantage of multidisciplinary findings, we dissect the many aspects of isoform-specific AMPK function and links to health and disease. These include, but are not limited to, allosteric activation by adenine nucleotides and small molecules, co-translational myristoylation and post-translational modifications (particularly phosphorylation), governance of subcellular localisation, and control of transcriptional networks. Finally, we delve into current debate over whether AMPK can form novel protein complexes (e.g., dimers lacking the α-subunit), altogether highlighting opportunities for future and impactful research. MAJOR CONCLUSIONS Baseline activity of α1-AMPK is higher than its α2 counterpart and is more sensitive to synergistic allosteric activation by metabolites and small molecules. α2 complexes however, show a greater response to energy stress (i.e., AMP production) and appear to be better substrates for LKB1 and mTORC1 upstream. These differences may explain to some extent why in certain cancers α1 is a tumour promoter and α2 a suppressor. β1-AMPK activity is toggled by a 'myristoyl-switch' mechanism that likely precedes a series of signalling events culminating in phosphorylation by ULK1 and sensitisation to small molecules or endogenous ligands like fatty acids. β2-AMPK, not entirely beholden to this myristoyl-switch, has a greater propensity to infiltrate the nucleus, which we suspect contributes to its oncogenicity in some cancers. Last, the unique N-terminal extensions of the γ2 and γ3 isoforms are major regulatory domains of AMPK. mTORC1 may directly phosphorylate this region in γ2, although whether this is inhibitory, especially in disease states, is unclear. Conversely, γ3 complexes might be preferentially regulated by mTORC1 in response to physical exercise.
Collapse
Affiliation(s)
- William J Smiles
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria; Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia.
| | - Ashley J Ovens
- Protein Engineering in Immunity & Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia; Department of Medicine, University of Melbourne, Parkville, Australia
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
20
|
Lim JW, Lee JH, Nejad JG, Lee HG. Effects of L-leucine and sodium acetate on milk protein synthesis under heat stress conditions in bovine mammary epithelial cells in vitro. J Therm Biol 2024; 126:103975. [PMID: 39571500 DOI: 10.1016/j.jtherbio.2024.103975] [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: 01/09/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 12/21/2024]
Abstract
It is widely known that heat stress (HS) has negative effects on dairy cows, such as a reduction in milk production and milk protein. However, there has been no research yet on the effects of HS at the bovine mammary epithelial cells (MAC-T) level and the function of L-leucine (LEU) and sodium acetate (ACE) in reducing HS. In this study, we evaluated the negative effects of HS at various temperatures on MAC-T and verified whether LEU and ACE are effective at reducing HS and increasing protein synthesis. An experiment was conducted by dividing MAC-T into three groups: 39 °C, 41 °C, and 43 °C. In the case of LEU and ACE supplementation experiments, the cells were supplemented with 0, 0.45, 0.9, 1.8, and 3.6 mM of LEU and ACE to reach the differentiation medium. It was observed that under HS at 41 °C, HSP70, BAX, and eIF4EBP1 gene expression were increased, whereas Bcl-2, eIF4E, and PRKAA1 gene expression were decreased. When 1.8 mM of LEU was added under HS at 41 °C, it suppressed apoptosis by reducing the gene expression of HSP70 and controlling the gene expression of apoptosis-related genes such as BAX and Bcl-2. Additionally, mTOR, P-mTOR, and β-casein proteins were increased. In the case of 0.9 mM of ACE, it was found to decrease the gene expression of HSP70 and BAX and increase the amount of β-casein protein synthesis. Simultaneous supplementation of LEU and ACE has been shown to reduce HS, inhibit apoptosis, and increase β-casein protein expression. In summary, HS at 41 °C began to have a negative effect on MAC-T, while LEU and ACE reduced HS and inhibited apoptosis, alleviating cell damage and effectively increasing β-casein protein synthesis. The results suggest that LEU and ACE have the potential to reduce HS and promote protein synthesis under HS conditions in MAC-T.
Collapse
Affiliation(s)
- Jung-Woo Lim
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Jun-Hee Lee
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Jalil Ghassemi Nejad
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Hong-Gu Lee
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
21
|
Li L, Sun J, Chen F, Xiong L, She L, Hao T, Zeng Y, Li L, Wang W, Zhao X, Liang G. Pedunculoside alleviates cognitive deficits and neuronal cell apoptosis by activating the AMPK signaling cascade. Chin Med 2024; 19:163. [PMID: 39574131 PMCID: PMC11583384 DOI: 10.1186/s13020-024-01033-6] [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: 06/19/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Mitochondrial dysfunction emerges as an early pathological hallmark of Alzheimer's disease (AD). The reduction in mitochondrial membrane potential and the elevation of reactive oxygen species (ROS) production are pivotal in the initiation of neuronal cell apoptosis. Pedunculoside(Ped), a novel triterpene saponin derived from the dried barks of Ilex rotunda Thunb, exhibits a potent anti-inflammatory effect. In the course of drug screening, we discovered that Ped offers significant protection against apoptosis induced by Aβ1-42. Nevertheless, the role and mechanism of Ped in AD are yet to be elucidated. METHODS Oxidative stress was evaluated by measuring mitochondrial membrane potential and intracellular ROS production. The expression of proteins associated with apoptosis was determined using western blot analysis and flow cytometry. In vivo, the pathological characteristics of AD were investigated through Western blot and tissue immunofluorescence techniques. Cognitive function was assessed using the Morris Water Maze and Novel Object Recognition tests. RESULTS We demonstrated that Ped decreased apoptosis in PC12 cells, reduced the generation of intracellular ROS, and restored mitochondrial membrane potential. Mechanistically, we found that the protective effect of Ped against Aβ-induced neurotoxicity was associated with activation of the AMPK/GSK-3β/Nrf2 signaling pathway. In vivo, Ped alleviated memory deficits and inhibited neuronal apoptosis, inflammation, and oxidative stress in the hippocampus of 3 × Tg AD mice, along with the activation of the AMPK signaling pathway. CONCLUSION The findings indicate that Ped exerts its neuroprotective effects against oxidative stress and apoptosis through the AMPK signaling cascade. The results demonstrate that Ped is a potential candidate for the treatment of AD.
Collapse
Affiliation(s)
- Liwei Li
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Jinfeng Sun
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, People's Republic of China
| | - Fan Chen
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Li Xiong
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Lingyu She
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, People's Republic of China
| | - Tang Hao
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Yuqing Zeng
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Luyao Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, 325035, Zhejiang, China
| | - Wei Wang
- Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Yongkang, 321399, Zhejiang, China
| | - Xia Zhao
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China.
| | - Guang Liang
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China.
| |
Collapse
|
22
|
Tsai KW, Liao JB, Tseng HW. Metformin regulates the proliferation and motility of melanoma cells by modulating the LINC00094/miR-1270 axis. Cancer Cell Int 2024; 24:384. [PMID: 39563323 PMCID: PMC11575040 DOI: 10.1186/s12935-024-03545-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Melanoma is an aggressive tumor with a high mortality rate. Metformin, a commonly prescribed diabetes medication, has shown promise in cancer prevention and treatment. Long noncoding RNAs (lncRNAs) are non-protein-coding RNA molecules that play a key role in tumor development by interacting with cellular chromatins. Despite the benefits of metformin, the anticancer mechanism underlying its effect on the regulation of lncRNAs in melanoma remains unclear. METHODS We investigated the lncRNA profiles of human melanoma cells with and without metformin treatment using a next-generation sequencing approach (NGS). Utilizing public databases, we analyzed the expression levels and clinical impacts of LINC00094 and miR-1270 in melanoma. The expression levels of LINC00094 and miR-1270 were verified in human cell lines and clinical samples by real-time PCR and in situ hybridization. The biological roles of LINC00094 and miR-1270 in cell growth, proliferation, cell cycle, apoptosis, and motility were studied using in vitro assays. RESULTS We identify a novel long noncoding RNA, namely LINC00094, whose expression considerably decreased in melanoma cells after metformin treatment. In situ hybridization analysis revealed substantially higher expression of LINC00094 in cutaneous melanoma tissue compared with adjacent normal epidermis and normal control tissues (P < 0.001). In nondiabetic patients with melanoma, the overall survival of high LINC00094 expression group was shorter than the low LINC00094 expression group with borderline statistical significance (log-rank test, P = 0.057). Coexpression analysis of LINC00094 indicated its involvement in the mitochondrial respiratory pathway, with its knockdown suppressing genes associated with mitochondrial oxidative phosphorylation, glycolysis, antioxidant production, and metabolite levels. Functional analysis revealed that silencing-LINC00094 inhibited the proliferation, colony formation, invasion, and migration of melanoma cells. Cell cycle analysis following LINC00094 knockdown revealed G1 phase arrest with reduced cell cycle protein expression. Combined TargetScan and reporter assays revealed a direct link between miR-1270 and LINC00094. Ectopic miR-1270 expression inhibited melanoma cell growth and motility while inducing apoptosis. Finally, through in silico analysis, we identified two miR-1270 target genes, CD276 and centromere protein M (CENPM), which may be involved in the biological functions of LINC00094. CONCLUSIONS Overall, LINC00094 expression may regulate melanoma cell growth and motility by modulating the expression of miR-1270, and targeting genes of CD276 and CENPM indicating its therapeutic potential in melanoma treatment.
Collapse
Affiliation(s)
- Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, New Taipei City, Taiwan
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Shu Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hui-Wen Tseng
- Department of Dermatology, Ministry of Health and Welfare Pingtung Hospital, Pingtung, Taiwan.
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- Department of Nursing, College of Nursing, Meiho University, Neipu, Pingtung, Taiwan.
| |
Collapse
|
23
|
Kiyimba F, Hartson SD, Mafi GG, Ramanathan R. Glycogen Supplementation in Vitro Promotes pH Decline in Dark-Cutting Beef by Reverting Muscle's Metabolome toward a Normal Postmortem Muscle State. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25275-25285. [PMID: 39496138 PMCID: PMC11565789 DOI: 10.1021/acs.jafc.4c06490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
Dysregulated muscle glycogen metabolism preslaughter contributes to aberrant postmortem muscle pH (>5.8) in dark-cutting beef phenotypes. However, the underlying mechanisms have remained elusive. Herein, we examine the glycogen dependent regulation of postmortem muscle pH decline and darkening in beef. We show that supplementation of glycogen in vitro restores postmortem pH decline in dark-cutting beef by reverting the metabolome toward a typical postmortem muscle state characterized by increased activities of enzymes glycogen phosphorylase and lactate dehydrogenase (p < 0.05) coupled with a pronounced abundance of glycolytic metabolites and reduced abundance of tricarboxylic acid cycle and amino acid metabolites. Furthermore, concurrent inhibition of mitochondrial respiration at complexes I, IV, and V with glycogen supplementation stimulates greater pH decline. Together, our findings show that supplementing glycogen at low concentrations (10 mM) can reprogram the dark-cutting beef muscle's metabolome toward typical postmortem state and promote muscle acidification. Thus, enhancing glycogen levels could represent a promising strategy for mitigating dark-cutting beef phenotypes and improving meat quality.
Collapse
Affiliation(s)
- Frank Kiyimba
- Department
of Animal and Food Sciences, Oklahoma State
University, Stillwater, Oklahoma 74078, United States
| | - Steven D. Hartson
- Department
of Biochemistry and Molecular Biology, Oklahoma
State University, Stillwater, Oklahoma 74078, United States
| | - Gretchen G. Mafi
- Department
of Animal and Food Sciences, Oklahoma State
University, Stillwater, Oklahoma 74078, United States
| | - Ranjith Ramanathan
- Department
of Animal and Food Sciences, Oklahoma State
University, Stillwater, Oklahoma 74078, United States
| |
Collapse
|
24
|
Yang M, Wu S, Dai Q, Qin W, Zhang Y, Lei Y, Song H, Zheng T, Guan M, Huang G, Liu X. Andrographolide prevents renal fibrosis via decelerating lipotoxicity-mediated premature senescence of tubular epithelial cells. Biochem Pharmacol 2024; 230:116615. [PMID: 39528075 DOI: 10.1016/j.bcp.2024.116615] [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: 05/07/2024] [Revised: 09/18/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Excessive lipid accumulation often occurs in the early stage of chronic kidney disease (CKD) which is prone to induce oxidative stress and mitochondrial damage, promoting the progression of kidney fibrosis. Andrographolide (AP), a multifunctional natural terpenoids derived from Andrographis paniculate, has been suggested to play beneficial roles in metabolic disorders-associated disease. Here, we reported that AP effectively counteracts tubule injury and interstitial fibrosis in mice fed with a long-term high-fat diet (HFD). AP treatment decreased HFD-induced lipid accumulation in kidney parenchyma and attenuated lipotoxicity-mediated oxidative stress and mitochondrial dysfunction, resulting in a marked decrease in tubular cell senescence. Importantly, AP inhibited senescence-associated secretory phenotype (SASP) secretion by senescent tubular cells, and in turn suppressed proliferation and activation of fibroblasts in a paracrine effect. Furthermore, we revealed that AP functions as an AMP-activated protein kinase (AMPK) activator to ameliorate renal lipid accumulation through coordinately modulating AMP-activated protein kinase AMPK target genes. By stimulation of AMPK activity, AP protects injured kidney against tubular cell senescence and fibroblast activation. These results suggest the potential therapeutic application of AP in the prevention and treatment of CKD, highlighting the promising drug strategy of targeting the lipotoxicity-mediated premature senescence in tubular cells.
Collapse
Affiliation(s)
- Meng Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Shengquan Wu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Qihui Dai
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Weihong Qin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yujie Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yiting Lei
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Haochang Song
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Tingting Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Min Guan
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China.
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China.
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
25
|
Ahmed ME, Akhter N, Fatima S, Ahmad S, Giri S, Hoda MN, Ahmad AS. Therapeutic utility of Perfluorocarbon Oxygent in limiting the severity of subarachnoid hemorrhage in mice. Sci Rep 2024; 14:26638. [PMID: 39496694 PMCID: PMC11535447 DOI: 10.1038/s41598-024-77321-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
Subarachnoid hemorrhage (SAH) is the deadliest form of hemorrhagic stroke; however, effective therapies are still lacking. Perfluorocarbons (PFCs) are lipid emulsion particles with great flexibility and their much smaller size as compared to red blood cells (RBCs) allows them to flow more efficiently within the blood circulation. Due to their ability to carry oxygen, a specific PFC-based emulsion, PFC-Oxygent, has been used as a blood substitute; however, its role in cerebral blood flow regulation is unknown. Adult C57BL/6 wildtype male mice were subjected to an endovascular perforation model of SAH followed by an intravenous (i.v.) injection of 9 ml/kg PFC-Oxygent or no treatment at 5 h after SAH. At 48 h after SAH, functional and anatomical outcomes were assessed. We found that SAH resulted in significant neurologic and motor deficits which were prevented by PFC-Oxygent treatment. We found that SAH-induced vasospasm, reduced RBC deformability, and augmented endothelial dysfunction were also restricted by PFC-Oxygent treatment. Moreover, mitochondrial activity and fusion proteins were also markedly decreased as assessed by oxidative phosphorylation (OXPHOS) after SAH. Interestingly, PFC-Oxygent treatment brought the mitochondrial activity close to the basal level. Moreover, SAH attenuated the level of phosphorylated AMP-activated protein kinase (pAMPK), whereas PFC treatment improved pAMPK levels. These data show the beneficial effects of PFC-Oxygent in limiting the severity of SAH. Further studies are needed to fully understand the mechanism through which PFC-Oxygent exerts its beneficial effects in limiting SAH severity.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neurology, Henry Ford Health, 2799 W Grand Blvd, Detroit, MI, 48202, USA
| | - Naseem Akhter
- Department of Neurology, Henry Ford Health, 2799 W Grand Blvd, Detroit, MI, 48202, USA
| | - Sumbul Fatima
- Department of Neurology, Henry Ford Health, 2799 W Grand Blvd, Detroit, MI, 48202, USA
| | - Saif Ahmad
- Department of Neurosurgery and Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, 2799 W Grand Blvd, Detroit, MI, 48202, USA
| | - Md Nasrul Hoda
- Department of Neurology, Henry Ford Health, 2799 W Grand Blvd, Detroit, MI, 48202, USA
| | | |
Collapse
|
26
|
Hu H, Lu X, He Y, Li J, Wang S, Luo Z, Wang Y, Wei J, Huang H, Duan C, Sun N. Sestrin2 in POMC neurons modulates energy balance and obesity related metabolic disorders via mTOR signaling. J Nutr Biochem 2024; 133:109703. [PMID: 39025457 DOI: 10.1016/j.jnutbio.2024.109703] [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: 10/17/2023] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 07/20/2024]
Abstract
Sestrin2 is a highly conserved protein that can be induced under various stress conditions. Researches have revealed that the signaling pathway of the mammalian target of rapamycin (mTOR) is essential in modulating both glucose and lipid metabolism. However, the precise involvement of Sestrin2 in the hypothalamus, particularly in pro-opiomelanocortin (POMC) neurons, in control of energy homeostasis remains uncertain. In this study, we aimed to investigate the functional role of Sestrin2 in hypothalamic POMC neurons in regulation of energy balance, as well as revealing the underlying mechanisms. Therefore, cre-dependent AAV virus encoding or silencing Sestrin2 was injected into the hypothalamic ARC of pomc-cre transgenic mice. The results demonstrated that Sestrin2 overexpression in POMC neurons ameliorated high-fat diet (HFD)-induced obesity and increased energy expenditure. Conversely, Sestrin2 deficiency in POMC neurons predisposed mice to HFD induced obesity. Additionally, the thermogenesis of brown adipose tissue and lipolysis of inguinal white adipose tissue were both enhanced by the increased sympathetic nerve innervation in Sestrin2 overexpressed mice. Further exploration revealed that Sestrin2 overexpression inhibited the mTOR signaling pathway in hypothalamic POMC neurons, which may account for the alleviation of systematic metabolic disturbance induced by HFD in these mice. Collectively, our findings demonstrate that Sestrin2 in POMC neurons plays a pivotal role in maintaining energy balance in a context of HFD-induced obesity by inhibiting the mTOR pathway, providing new insights into how hypothalamic neurons respond to nutritional signals to protect against obesity-associated metabolic dysfunction.
Collapse
Affiliation(s)
- Huiling Hu
- Department of Clinical Laboratory, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoxia Lu
- Department of Clinical Laboratory, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuqing He
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Li
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Shoujie Wang
- Center for Precision Medicine, Platform of Metabolomics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhijun Luo
- Emergency Department, The Seventh Affiliated Hospital, Southern Medical University, Foshan, China
| | - Ying Wang
- Department of Clinical Laboratory, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Wei
- Department of Clinical Laboratory, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hao Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Chaohui Duan
- Department of Clinical Laboratory, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Nannan Sun
- Department of Obstetrics and Gynecology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
27
|
Naseem S, Sun L, Qiu J. Stress granules in atherosclerosis: Insights and therapeutic opportunities. Curr Probl Cardiol 2024; 49:102760. [PMID: 39059785 DOI: 10.1016/j.cpcardiol.2024.102760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Atherosclerosis, a complex inflammatory and metabolic disorder, is the underlying cause of several life-threatening cardiovascular diseases. Stress granules (SG) are biomolecular condensates composed of proteins and mRNA that form in response to stress. Recent studies suggest a potential link between SG and atherosclerosis development. However, there remain gaps in understanding SG role in atherosclerosis development. Here we provide a thorough analysis of the role of SG in atherosclerosis, covering cellular stresses stimulation, core components, and regulatory genes in SG formation. Furthermore, we explore atherosclerosis induced factors such as inflammation, low or oscillatory shear stress (OSS), and oxidative stress (OS) may impact SG formation and then the development of atherosclerotic lesions. We have assessed how changes in SG dynamics impact pro-atherogenic processes like endothelial dysfunction, lipid metabolism, and immune cell recruitment in atherosclerosis. In summary, this review emphasizes the complex interplay between SG and atherosclerosis that could open innovative directions for targeted therapeutic strategies in preventing or treating atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Sahar Naseem
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lijuan Sun
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
28
|
Yang Y, Zhao Y, Lei H. Alleviating effect of Lactobacillus rhamnosus SDSP202418 on exercise-induced fatigue in mice. Front Microbiol 2024; 15:1420872. [PMID: 39391603 PMCID: PMC11464290 DOI: 10.3389/fmicb.2024.1420872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
In this study, the effects of Lactobacillus rhamnosus SDSP202418 isolated from shrimp paste on the exercise performance of fatigued mice were analyzed, and the potential action mechanism was revealed. L. rhamnosus SDSP202418 significantly improved the exhaustion time of the mice and regulated the biochemical indices (lactate dehydrogenase, nitrogen, and uric acid) of the fatigued mice to resist fatigue. L. rhamnosus SDSP202418 also upregulated the mRNA expression of slow muscle fibers and downregulated the mRNA expression of fast muscle fibers in the exercise mice by activating the AMPK/PGC-1α pathway in the fatigued mice. It also increased the contents of antioxidant enzymes (superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH)) in the liver and muscle. These enzymes removed and repaired oxidative free radicals to achieve antifatigue. In addition, L. rhamnosus SDSP202418 can change the gut microbial structure and modulate the abundance and balance of fatigue-related gut microbiota, which in turn exerts antifatigue effects. L. rhamnosus SDSP202418 is a functional food component that relieves fatigue after exercise.
Collapse
Affiliation(s)
- Yang Yang
- College of Physical Education, Chengdu Sport University, Chengdu, China
| | - Yuanji Zhao
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Huan Lei
- College of Physical Education, Chengdu Sport University, Chengdu, China
| |
Collapse
|
29
|
Cheng Z, Liu B, Liu X. Circadian gene signatures in the progression of obesity based on machine learning and Mendelian randomization analysis. Front Nutr 2024; 11:1407265. [PMID: 39351493 PMCID: PMC11439728 DOI: 10.3389/fnut.2024.1407265] [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: 03/26/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Objective Obesity, a global health concern, is associated with a spectrum of chronic diseases and cancers. Our research sheds light on the regulatory role of circadian genes in obesity progression, providing insight into the immune landscape of obese patients, and introducing new avenues for therapeutic interventions. Methods Expression files of multiple datasets were retrieved from the GEO database. By 80 machine-learning algorithm combinations and Mendelian randomization analysis, we discovered the key circadian genes contributing to and protecting against obesity. Subsequently, an immune infiltration analysis was conducted to examine the alterations in immune cell types and their abundance in the body and to investigate the relationships between circadian genes and immune cells. Furthermore, we delved into the molecular mechanisms of key genes implicated in obesity. Results Our study identified three key circadian genes (BHLHE40, PPP1CB, and CSNK1E) associated with obesity. BHLHE40 was found to promote obesity through various pathways, while PPP1CB and CSNK1E counteracted lipid metabolism disorders, and modulated cytokines, immune receptors, T cells, and monocytes. Conclusion In conclusion, the key circadian genes (BHLHE40, CSNK1E, and PPP1CB) may serve as novel biomarkers for understanding obesity pathogenesis and have significant correlations with infiltrating immune cells, thus providing potential new targets for obese prevention and treatment.
Collapse
Affiliation(s)
- Zhi’ang Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Binghong Liu
- College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Xiaoyong Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Department of Ophthalmology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| |
Collapse
|
30
|
Rehman IU, Park JS, Choe K, Park HY, Park TJ, Kim MO. Overview of a novel osmotin abolishes abnormal metabolic-associated adiponectin mechanism in Alzheimer's disease: Peripheral and CNS insights. Ageing Res Rev 2024; 100:102447. [PMID: 39111409 DOI: 10.1016/j.arr.2024.102447] [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: 07/19/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disease that affects millions of people worldwide. It is caused by abnormalities in cholinergic neurons, oxidative stress, and inflammatory cascades. The illness is accompanied by personality changes, memory issues, and dementia. Metabolic signaling pathways help with fundamental processes like DNA replication and RNA transcription. Being adaptable is essential for both surviving and treating illness. The body's metabolic signaling depends on adipokines, including adiponectin (APN) and other adipokines secreted by adipose tissues. Energy homeostasis is balanced by adipokines, and nutrients. Overconsumption of nutrients messes with irregular signaling of adipokines, such as APN in both peripheral and brain which leads to neurodegeneration, such as AD. Despite the failure of traditional treatments like memantine and cholinesterase inhibitors, natural plant bioactive substances like Osmotin (OSM) have been given a focus as potential therapeutics due to their antioxidant properties, better blood brain barrier (BBB) permeability, excellent cell viability, and especially nanoparticle approaches. The review highlights the published preclinical literature regarding the role of OSM in AD pathology while there is a need for more research to investigate the hidden therapeutic potential of OSM which may open a new gateway and further strengthen its healing role in the pathogenesis of neurodegeneration, especially AD.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands.
| | - Hyun Young Park
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands; Department of Pediatrics, Maastricht University Medical Center (MUMC+), Maastricht 6202 AZ, the Netherlands.
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, United Kingdom.
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Alz-Dementia Korea Co., Jinju 52828, Republic of Korea.
| |
Collapse
|
31
|
Li J, Xiao F, Wang S, Fan X, He Z, Yan T, Zhang J, Yang M, Yang D. LncRNAs are involved in regulating ageing and age-related disease through the adenosine monophosphate-activated protein kinase signalling pathway. Genes Dis 2024; 11:101042. [PMID: 38966041 PMCID: PMC11222807 DOI: 10.1016/j.gendis.2023.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 06/15/2023] [Indexed: 07/06/2024] Open
Abstract
A long noncoding RNA (lncRNA) is longer than 200 bp. It regulates various biological processes mainly by interacting with DNA, RNA, or protein in multiple kinds of biological processes. Adenosine monophosphate-activated protein kinase (AMPK) is activated during nutrient starvation, especially glucose starvation and oxygen deficiency (hypoxia), and exposure to toxins that inhibit mitochondrial respiratory chain complex function. AMPK is an energy switch in organisms that controls cell growth and multiple cellular processes, including lipid and glucose metabolism, thereby maintaining intracellular energy homeostasis by activating catabolism and inhibiting anabolism. The AMPK signalling pathway consists of AMPK and its upstream and downstream targets. AMPK upstream targets include proteins such as the transforming growth factor β-activated kinase 1 (TAK1), liver kinase B1 (LKB1), and calcium/calmodulin-dependent protein kinase β (CaMKKβ), and its downstream targets include proteins such as the mechanistic/mammalian target of rapamycin (mTOR) complex 1 (mTORC1), hepatocyte nuclear factor 4α (HNF4α), and silencing information regulatory 1 (SIRT1). In general, proteins function relatively independently and cooperate. In this article, a review of the currently known lncRNAs involved in the AMPK signalling pathway is presented and insights into the regulatory mechanisms involved in human ageing and age-related diseases are provided.
Collapse
Affiliation(s)
- Jiamei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Feng Xiao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Siqi Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiaolan Fan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zhi He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Taiming Yan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jia Zhang
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610017, China
| | - Mingyao Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Deying Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
32
|
Li Z, Zhao Q, Liu X, Zhou X, Wang Y, Zhao M, Wu F, Zhao G, Guo X. Capsaicin combined with cisplatin inhibits TGF-β1-induced EMT and TSCC cells migration via the Claudin-1/PI3K/AKT/mTOR signaling pathway. Cancer Cell Int 2024; 24:300. [PMID: 39198820 PMCID: PMC11360848 DOI: 10.1186/s12935-024-03485-0] [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: 04/20/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Tongue squamous cell carcinoma (TSCC) is one of the most common malignant tumors among oral cancers, and its treatment is based on radio-chemotherapy and surgery, which always produces more serious side effects and sequelae. Traditional medicine can compensate for the shortcomings of modern medical treatments and play a better therapeutic role. Currently, active ingredients derived from plants are attracting the attention of researchers and clinical professionals. We examined capsaicin (CAP), an active ingredient isolated from Capsicum annuum (family Solanaceae), and explored the effect of CAP combined with cisplatin (DDP) on epithelial-mesenchymal transition (EMT) and TSCC cells migration. Our results demonstrated that Transforming growth factor-β1(TGF-β1) induced EMT and promoted cell migration in TSCC cells. CAP combined with DDP inhibits non-TGF-β1-induced or TGF-β1-induced EMT and migration. Mechanistically, the inhibition of non-TGF-β1-induced EMT and migration by CAP combined with DDP was mediated by the AMPK/mTOR pathway, whereas TGF-β1-induced EMT and migration were regulated by the Claudin-1/PI3K/AKT/mTOR pathway. A nude lung metastasis mouse model was established for in vivo validation. These results support our hypothesis that the combination of CAP and DDP inhibits TSCC metastasis. These data set the stage for further studies aimed at validating CAP as an effective active ingredient for enhancing chemotherapy efficacy and reducing the dosage and toxicity of chemotherapeutic drugs, ultimately paving the way for translational research and clinical trials for TSCC eradication.
Collapse
Affiliation(s)
- Zhuang Li
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Qiwei Zhao
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Xiayang Liu
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Xinyue Zhou
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Yu Wang
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Min Zhao
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Fenghua Wu
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Gang Zhao
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
| | - Xiaohong Guo
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China.
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China.
| |
Collapse
|
33
|
Lee JH, Jung IR, Tu-Sekine B, Jin S, Anokye-Danso F, Ahima RS, Kim SF. Genetic Deletion of Skeletal Muscle Inositol Polyphosphate Multikinase Disrupts Glucose Homeostasis and Impairs Exercise Tolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605526. [PMID: 39131310 PMCID: PMC11312436 DOI: 10.1101/2024.07.28.605526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Inositol phosphates are critical signaling messengers involved in a wide range of biological pathways in which inositol polyphosphate multikinase (IPMK) functions as a rate-limiting enzyme for inositol polyphosphate metabolism. IPMK has been implicated in cellular metabolism, but its function at the systemic level is still poorly understood. Since skeletal muscle is a major contributor to energy homeostasis, we have developed a mouse model in which skeletal muscle IPMK is specifically deleted and examined how a loss of IPMK affects whole-body metabolism. Here, we report that mice in which IPMK knockout is deleted, specifically in the skeletal muscle, displayed an increased body weight, disrupted glucose tolerance, and reduced exercise tolerance under the normal diet. Moreover, these changes were associated with an increased accumulation of triglyceride in skeletal muscle. Furthermore, we have confirmed that a loss of IPMK led to reduced beta-oxidation, increased triglyceride accumulation, and impaired insulin response in IPMK-deficient muscle cells. Thus, our results suggest that IPMK mediates the whole-body metabolism via regulating muscle metabolism and may be potentially targeted for the treatment of metabolic syndromes.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Ik-Rak Jung
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Becky Tu-Sekine
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Sunghee Jin
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Frederick Anokye-Danso
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Rexford S Ahima
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| | - Sangwon F Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA. 21224
| |
Collapse
|
34
|
Ramesh T, Shahid M. Bacoside-A repressed the differentiation and lipid accumulation of 3T3-L1 preadipocytes by modulating the expression of adipogenic genes. Biotechnol Appl Biochem 2024; 71:741-752. [PMID: 38419375 DOI: 10.1002/bab.2573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Obesity is one of the more complicated diseases, it can induce numerous life-threatening diseases mainly diabetes mellitus, cardiovascular disease, hypertension, and certain cancers. In this study, we assessed the efficacy of bacoside-A (a dammarane-type triterpenoid saponin derived from the plant Bacopa monniera Linn.) on the adipogenesis of 3T3-L1 preadipocytes. Results of this study illustrated that bacoside-A decreased the differentiation of 3T3-L1 cell, as evidenced by diminution of lipid droplets, which contains triglycerides and other lipids. During the differentiation process, transcription factors, which are mainly participating in adipogenesis such us CCAAT/enhancer-binding protein α (C/EBPα) and C/EBPβ, peroxisome proliferator-activated receptor-γ (PPARγ), and sterol regulatory element-binding protein-1c (SREBP-1c), expressions were significantly suppressed by bacoside-A. In addition, bacoside-A showed a potent reduction in genes precise to adipocytes such as lipoprotein lipase (LPL), fatty acid synthase (FAS), adipocyte fatty acid-binding protein (FABP4), and leptin expressions. Further, bacoside-A stimulated the phosphorylation of acetyl CoA carboxylase (ACC) and AMP-activated protein kinase (AMPK). These results demonstrated that bacoside-A has anti-adipogenic effects by regulating the transcription factors involved in adipocyte differentiation. Therefore, bacoside-A might be considered as a potent therapeutic agent for alleviating obesity and hyperlipidemia.
Collapse
Affiliation(s)
- Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| |
Collapse
|
35
|
Romero-Herrera I, Nogales F, Gallego-López MDC, Díaz-Castro J, Carreras O, Ojeda ML. Selenium supplementation via modulation of selenoproteins ameliorates binge drinking-induced oxidative, energetic, metabolic, and endocrine imbalance in adolescent rats' skeletal muscle. Food Funct 2024; 15:7988-8007. [PMID: 38984595 DOI: 10.1039/d4fo01354a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Adolescence is characterized by increased vulnerability to addiction and ethanol (EtOH) toxicity, particularly through binge drinking (BD), a favored acute EtOH-ingestion pattern among teenagers. BD, highly pro-oxidant, induces oxidative stress (OS), affecting skeletal muscle (SKM), where selenium (Se), an antioxidant element and catalytic center of selenoproteins, is stored, among other tissues. Investigating the effects of Se supplementation on SKM after BD exposure holds therapeutic promise. For this, we randomised 32 adolescent Wistar rats into 4 groups, exposed or not to intermittent i.p. BD [BD and control (C)] (3 g EtOH per kg per day), and supplemented with selenite [BDSe and CSe] (0.4 ppm). In SKM, we examined the oxidative balance, energy status (AMPK, SIRT-1), protein turnover (IRS-1, Akt1, mTOR, IGF-1, NF-κB p65, MAFbx, ULK1, pelF2α), serum myokines (myostatin, IL-6, FGF21, irisin, BDNF, IL-15, fractalkine, FSTL-1, FABP-3), and selenoproteins (GPx1, GPx4, SelM, SelP). In the pancreas, we studied the oxidative balance and SIRT-1 expression. Selenite supplementation mitigated BD-induced OS by enhancing the expression of selenoproteins, which restored oxidative balance, notably stimulating protein synthesis and normalizing the myokine profile, leading to improved SKM mass growth and metabolism, and reduced inflammation and apoptosis (caspase-3). Selenite restoration of SelP's receptor LRP1 expression, reduced by BD, outlines the crucial role of SKM in the SelP cycle, linking Se levels to SKM development. Furthermore, Se attenuated pancreatic OS, preserving insulin secretion. Se supplementation shows potential for alleviating SKM damage from BD, with additional beneficial endocrine effects on the pancreas, adipose tissue, liver, heart and brain that position it as a broad-spectrum treatment for adolescent alcohol consumption, preventing metabolic diseases in adulthood.
Collapse
Affiliation(s)
- Inés Romero-Herrera
- Department of Physiology, Faculty of Pharmacy, University of Seville, C/Professor García González 2, 41012-Seville, Spain.
| | - Fátima Nogales
- Department of Physiology, Faculty of Pharmacy, University of Seville, C/Professor García González 2, 41012-Seville, Spain.
| | - María Del Carmen Gallego-López
- Department of Physiology, Faculty of Pharmacy, University of Seville, C/Professor García González 2, 41012-Seville, Spain.
| | - Javier Díaz-Castro
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Avenida del Conocimiento s/n, 18071-Armilla, Granada, Spain.
- Department of Physiology, University of Granada, Granada, Spain
| | - Olimpia Carreras
- Department of Physiology, Faculty of Pharmacy, University of Seville, C/Professor García González 2, 41012-Seville, Spain.
| | - María Luisa Ojeda
- Department of Physiology, Faculty of Pharmacy, University of Seville, C/Professor García González 2, 41012-Seville, Spain.
| |
Collapse
|
36
|
Muscarà C, Speciale A, Molonia MS, Salamone FL, Saija A, Cimino F. Intestinal epithelial differentiation and barrier function is promoted in vitro by a Cynara cardunculus L. leaf extract through AMPK pathway activation. Nat Prod Res 2024:1-11. [PMID: 39058646 DOI: 10.1080/14786419.2024.2384080] [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: 03/16/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Gut epithelial barrier perturbation leads to leaky gut syndrome and permeation of substances activating immune response. Polyphenols can improve intestinal barrier function and represent candidates for preventing development of leaky gut. Herein, we evaluated in vitro the molecular mechanisms involved in the protective effects of a polyphenol-rich extract from leaves of Cynara cardunculus L. (CCLE) on intestinal barrier function and integrity on Caco-2 human epithelial cells. Treatment with CCLE from seeding until complete differentiation improved intestinal function by increasing trans-epithelial electrical resistance (TEER), reducing paracellular permeability to fluorescein, and promoting faster recovery of tight junctions (TJ) assembly in the Ca2+ switch assay. CCLE stimulated epithelial cell differentiation inducing alkaline phosphatase activity and TJ proteins. These CCLE-induced effects were attributed to activation of AMP-activated protein kinase (AMPK) pathway. Our data support the use of Cynara cardunculus L. leaves, an agricultural co-product rich in bioactive polyphenols, for the health of intestinal epithelium.
Collapse
Affiliation(s)
- Claudia Muscarà
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Antonio Speciale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Maria Sofia Molonia
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Federica Lina Salamone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Antonella Saija
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Francesco Cimino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
37
|
Aodah AH, Alkholifi FK, Alharthy KM, Devi S, Foudah AI, Yusufoglu HS, Alam A. Effects of kaempherol-3-rhamnoside on metabolic enzymes and AMPK in the liver tissue of STZ-induced diabetes in mice. Sci Rep 2024; 14:16167. [PMID: 39003280 PMCID: PMC11246446 DOI: 10.1038/s41598-024-66426-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: 02/12/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by persistent hyperglycemia. It involves disturbances in carbohydrate, fat, and protein metabolism due to defects in insulin secretion, insulin action, or both. Novel therapeutic approaches are continuously being explored to enhance metabolic control and prevent complications associated with the disease. This study investigates the therapeutic potential of kaempherol-3-rhamnoside, a flavonoid, in managing diabetes by modulating the AMP-activated protein kinase (AMPK) pathway and improving metabolic enzyme activities in streptozotocin (STZ) -induced diabetic mice. Diabetic mice were treated with varying doses of kaempherol-3-rhamnoside and/or insulin over a 28-day period. Glycolytic and gluconeogenesis enzyme activities in the liver, fasting blood glucose levels, serum insulin levels, lipid profiles and oxidative stress markers were assessed. Treatment with kaempherol-3-rhamnoside significantly improved glycolytic enzyme activities, reduced fasting blood glucose, and enhanced insulin levels compared to diabetic controls. The compound also normalized lipid profiles and reduced oxidative stress in the liver, suggesting its potential in reversing diabetic dyslipidemia and oxidative damage. Furthermore, kaempherol-3-rhamnoside activated the AMPK pathway, indicating a mechanism through which it could exert its effects. Kaempherol-3-rhamnoside exhibits promising antidiabetic properties, potentially through AMPK pathway activation and metabolic enzyme modulation. These findings support its potential use as an adjunct therapy for diabetes management. Further clinical studies are warranted to validate these results in human subjects.
Collapse
Affiliation(s)
- Alhussain H Aodah
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Faisal K Alkholifi
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Khalid M Alharthy
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Ahmed I Foudah
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Hasan S Yusufoglu
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Dentistry and Pharmacy, Buraydah Private Colleges, 51418, Buraydah, Saudi Arabia
| | - Aftab Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia.
| |
Collapse
|
38
|
Song C, Jung D, Kendi AT, Rho JK, Kim EJ, Horn I, Curran GL, Ghattamaneni S, Shim JY, Kang PS, Kang D, Thakkar JB, Dewan S, Lowe VJ, Lee SB. Metformin Prevents Tumor Cell Growth and Invasion of Human Hormone Receptor-Positive Breast Cancer (HR+ BC) Cells via FOXA1 Inhibition. Int J Mol Sci 2024; 25:7494. [PMID: 39000600 PMCID: PMC11242876 DOI: 10.3390/ijms25137494] [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: 06/05/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Women with type 2 diabetes (T2D) have a higher risk of being diagnosed with breast cancer and have worse survival than non-diabetic women if they do develop breast cancer. However, more research is needed to elucidate the biological underpinnings of these relationships. Here, we found that forkhead box A1 (FOXA1), a forkhead family transcription factor, and metformin (1,1-dimethylbiguanide hydrochloride), a medication used to treat T2D, may impact hormone-receptor-positive (HR+) breast cancer (BC) tumor cell growth and metastasis. Indeed, fourteen diabetes-associated genes are highly expressed in only three HR+ breast cancer cell lines but not the other subtypes utilizing a 53,805 gene database obtained from NCBI GEO. Among the diabetes-related genes, FOXA1, MTA3, PAK4, FGFR3, and KIF22 were highly expressed in HR+ breast cancer from 4032 breast cancer patient tissue samples using the Breast Cancer Gene Expression Omnibus. Notably, elevated FOXA1 expression correlated with poorer overall survival in patients with estrogen-receptor-positive/progesterone-receptor-positive (ER+/PR+) breast cancer. Furthermore, experiments demonstrated that loss of the FOXA1 gene inhibited tumor proliferation and invasion in vitro using MCF-7 and T47D HR+ breast cancer cell lines. Metformin, an anti-diabetic medication, significantly suppressed tumor cell growth in MCF-7 cells. Additionally, either metformin treatment or FOXA1 gene deletion enhanced tamoxifen-induced tumor growth inhibition in HR+ breast cancer cell lines within an ex vivo three-dimensional (3D) organoid model. Therefore, the diabetes-related medicine metformin and FOXA1 gene inhibition might be a new treatment for patients with HR+ breast cancer when combined with tamoxifen, an endocrine therapy.
Collapse
Affiliation(s)
- Christine Song
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
- Harvard University, Cambridge, MA 02138, USA
| | - Dawa Jung
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Ayse Tuba Kendi
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Jin Kyung Rho
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - Eun-Joo Kim
- Department of Molecular Biology, Dankook University, Cheonan 31116, Chungcheongnam, Republic of Korea;
| | - Ian Horn
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Geoffry L. Curran
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Sujala Ghattamaneni
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Ji Yeon Shim
- College of Nursing, Dankook University, Cheonan 31116, Chungcheongnam, Republic of Korea;
| | - Pil Soo Kang
- U&Hang Clinic, Asan 31514, Chungcheongnam, Republic of Korea;
| | - Daehun Kang
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Jay B. Thakkar
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Sannidhi Dewan
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Val J. Lowe
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
| | - Seung Baek Lee
- Division of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (C.S.); (D.J.); (A.T.K.); (I.H.); (G.L.C.); (S.G.); (D.K.); (J.B.T.); (S.D.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
39
|
Dieguez HH, Romeo HE, Alaimo A, Bernal Aguirre NA, Calanni JS, Adán Aréan JS, Alvarez S, Sciurano R, Rosenstein RE, Dorfman D. Mitochondrial quality control in non-exudative age-related macular degeneration: From molecular mechanisms to structural and functional recovery. Free Radic Biol Med 2024; 219:17-30. [PMID: 38579938 DOI: 10.1016/j.freeradbiomed.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
Non-exudative age-related macular degeneration (NE-AMD) is the leading blindness cause in the elderly. Clinical and experimental evidence supports that early alterations in macular retinal pigment epithelium (RPE) mitochondria play a key role in NE-AMD-induced damage. Mitochondrial dynamics (biogenesis, fusion, fission, and mitophagy), which is under the central control of AMP-activated kinase (AMPK), in turn, determines mitochondrial quality. We have developed a NE-AMD model in C57BL/6J mice induced by unilateral superior cervical ganglionectomy (SCGx), which progressively reproduces the disease hallmarks circumscribed to the temporal region of the RPE/outer retina that exhibits several characteristics of the human macula. In this work we have studied RPE mitochondrial structure, dynamics, function, and AMPK role on these parameters' regulation at the nasal and temporal RPE from control eyes and at an early stage of experimental NE-AMD (i.e., 4 weeks post-SCGx). Although RPE mitochondrial mass was preserved, their function, which was higher at the temporal than at the nasal RPE in control eyes, was significantly decreased at 4 weeks post-SCGx at the same region. Mitochondria were bigger, more elongated, and with denser cristae at the temporal RPE from control eyes. Exclusively at the temporal RPE, SCGx severely affected mitochondrial morphology and dynamics, together with the levels of phosphorylated AMPK (p-AMPK). AMPK activation with metformin restored RPE p-AMPK levels, and mitochondrial dynamics, structure, and function at 4 weeks post-SCGx, as well as visual function and RPE/outer retina structure at 10 weeks post-SCGx. These results demonstrate a key role of the temporal RPE mitochondrial homeostasis as an early target for NE-AMD-induced damage, and that pharmacological AMPK activation could preserve mitochondrial morphology, dynamics, and function, and, consequently, avoid the functional and structural damage induced by NE-AMD.
Collapse
Affiliation(s)
- Hernán H Dieguez
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Horacio E Romeo
- School of Engineering and Agrarian Sciences, Pontifical Catholic University of Argentina, BIOMED/UCA/CONICET, Buenos Aires, Argentina
| | - Agustina Alaimo
- Interdisciplinary Laboratory of Cellular Dynamics and Nanotools, Department of Biological Chemistry, Faculty of Exact and Natural Sciences/IQUIBICEN, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Nathaly A Bernal Aguirre
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Juan S Calanni
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Juan S Adán Aréan
- Department of Analytical Chemistry and Physicochemistry, School of Pharmacy and Biochemistry/IBIMOL, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Silvia Alvarez
- Department of Analytical Chemistry and Physicochemistry, School of Pharmacy and Biochemistry/IBIMOL, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Roberta Sciurano
- Department of Cellular Biology, Histology, Embryology and Genetics, School of Medicine/INBIOMED, UBA/CONICET, Buenos Aires, Argentina
| | - Ruth E Rosenstein
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Biological Chemistry, Faculty of Exact and Natural Sciences/IQUIBICEN, University of Buenos Aires, Buenos Aires, Argentina; Department of Human Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Damián Dorfman
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina.
| |
Collapse
|
40
|
HamaSalih RM. Effects of Semaglutide in Doxorubicin-Induced Cardiac Toxicity in Wistar Albino Rats. Cancer Manag Res 2024; 16:731-740. [PMID: 38952352 PMCID: PMC11216551 DOI: 10.2147/cmar.s468453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024] Open
Abstract
Background Doxorubicin (DOX) is used to treat various types of cancers. However, its use is restricted by cardiotoxicity, a leading cause of morbidity and mortality. Glucagon-like peptide 1 receptor agonists (GLP-1 RAs) may be associated with cardioprotective properties. Purpose This study aims to determine the protective effects of different semaglutide (SEM) doses on DOX-induced cardiotoxicity in a rat model. Methodology Thirty-five female Wistar rats were divided into five groups. The first group received distilled water as a negative control (NC); the positive control (PC) group received distilled water plus DOX; the third group (SL) received a low dose of SEM (0.06 mg/kg) plus DOX; the fourth group (SM) received a moderate dose of SEM (0.12 mg/kg) plus DOX; and the fifth group (SH) received a high dose of SEM (0.24 mg/kg) plus DOX. Blood samples were collected on day 8 to assess serum troponin, lactate dehydrogenase (LDH), creatine phosphokinase (CPK), total lipid profile, and vascular cell adhesion molecule 1 (VCAM-1). Cardiac tissue was sent for histopathological analysis. Results DOX increased the total cholesterol (TC), low-density lipoprotein (LDL), triglyceride (TG), LDH, and CKP levels. Moderate and high doses of semaglutide significantly reduced serum cholesterol levels (*p = 0.0199), (**p = 0.0077), respectively. A significant reduction (***p = 0.0013) in total body weight after treatment with SEM was observed in the SL group and a highly significant reduction (****p < 0.0001) was observed in the SM and SH groups. SEM at all doses reduced CPK levels. The SL group showed a significant reduction in troponin level (*p=0.0344). Serum LDH levels were reduced by all three SEM doses. The histopathological findings support the biochemical results. Conclusion Semaglutide may possess cardioprotective properties against DOX-induced cardiotoxicity in a rat model by decreasing serum biochemical markers of cardiotoxicity.
Collapse
Affiliation(s)
- Raz Muhammed HamaSalih
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Sulaimani, Slemani, Kurdistan Region, Iraq
| |
Collapse
|
41
|
Sapkota S, Briski KP. Sex-Dimorphic Effects of Hypoglycemia on Metabolic Sensor mRNA Expression in Ventromedial Hypothalamic Nucleus-Dorsomedial Division (VMNdm) Growth Hormone-Releasing Hormone Neurons. ACS Chem Neurosci 2024; 15:2350-2358. [PMID: 38757688 DOI: 10.1021/acschemneuro.4c00206] [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] [Indexed: 05/18/2024] Open
Abstract
Growth hormone-releasing hormone (Ghrh) neurons in the dorsomedial ventromedial hypothalamic nucleus (VMNdm) express the metabolic transcription factor steroidogenic factor-1 and hypoglycemia-sensitive neurochemicals of diverse chemical structures, transmission modes, and temporal signaling profiles. Ghrh imposes neuromodulatory control of coexpressed transmitters. Multiple metabolic sensory mechanisms are employed in the brain, including screening of the critical nutrient glucose or the energy currency ATP. Here, combinatory laser-catapult-microdissection/single-cell multiplex qPCR tools were used to investigate whether these neurons possess molecular machinery for monitoring cellular metabolic status and if these biomarkers exhibit sex-specific sensitivity to insulin-induced hypoglycemia. Data show that hypoglycemia up- (male) or downregulated (female) Ghrh neuron glucokinase (Gck) mRNA; Ghrh gene silencing decreased baseline and hypoglycemic patterns of Gck gene expression in each sex. Ghrh neuron glucokinase regulatory protein (Gckr) transcript levels were respectively diminished or augmented in hypoglycemic male vs female rats; this mRNA profile was decreased by Ghrh siRNA in both sexes. Gene transcripts encoding catalytic alpha subunits of the energy monitor 5-AMP-activated protein kinase (AMPK), i.e., Prkaa1 and 2, were increased by hypoglycemia in males, yet only the former mRNA was hypoglycemia-sensitive in females. Ghrh siRNA downregulated baseline and hypoglycemia-associated Prkaa subunit mRNAs in males but elicited divergent changes in Prkaa2 transcripts in eu- vs hypoglycemic females. Results provide unique evidence that VMNdm Ghrh neurons express the characterized metabolic sensor biomarkers glucokinase and AMPK and that the corresponding gene profiles exhibit distinctive sex-dimorphic transcriptional responses to hypoglycemia. Data further document Ghrh neuromodulation of baseline and hypoglycemic transcription patterns of these metabolic gene profiles.
Collapse
Affiliation(s)
- Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, United States
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, United States
| |
Collapse
|
42
|
Adamidis N, Papalexis P, Adamidis S. Exploring the Link Between Metabolic Syndrome and Cellulite. Cureus 2024; 16:e63464. [PMID: 38947139 PMCID: PMC11214470 DOI: 10.7759/cureus.63464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2024] [Indexed: 07/02/2024] Open
Abstract
Metabolic syndrome (MetS) encompasses a cluster of metabolic abnormalities, including insulin resistance, hypertension, abdominal obesity, and dyslipidemia, increasing cardiovascular disease and type 2 diabetes risks. Cellulite, a cosmetic condition marked by dimpled skin, predominantly affects women and shares risk factors with MetS, such as obesity and hormonal imbalances. This review examines the potential link between MetS and cellulite, focusing on shared pathophysiological pathways and implications for clinical practice and future research. Common factors such as inflammation, hormonal changes, and adipose tissue dysfunction are explored. The review highlights the importance of longitudinal studies to track cellulite progression in MetS patients, biomarker identification for early detection, intervention trials to assess therapeutic efficacy, mechanistic studies to elucidate underlying pathways and the impact of comorbidities on cellulite development. Understanding these relationships can enhance prevention, diagnosis, and treatment strategies for both MetS and cellulite, addressing significant public health and cosmetic concerns.
Collapse
Affiliation(s)
| | - Petros Papalexis
- Endocrinology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | | |
Collapse
|
43
|
Xiao Y, Jing D, Zhou G, Tang Z, Peng C, Kuang Y, Zhu W, Chen X, Liu H, Shen M. Adenosine 5'monophosphate-activated protein kinase activation reduces the risks of psoriasis and its comorbidities: a Mendelian randomization study in the UK Biobank. Rheumatology (Oxford) 2024; 63:1664-1671. [PMID: 37672020 DOI: 10.1093/rheumatology/kead462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/25/2023] [Accepted: 08/08/2023] [Indexed: 09/07/2023] Open
Abstract
OBJECTIVE Whether metformin and its adenosine 5'monophosphate-activated protein kinase (AMPK) activation protect from psoriasis risk is unconcluded. We investigated the effect of AMPK, a pharmacological target of metformin, on the risk of psoriasis and its comorbidities and mortality among participants in the UK Biobank (UKB). METHODS To avoid immortal time biases in pharmacoepidemiologic studies, Mendelian randomization was used to infer the AMPK pathway-dependent effects. The cut-off age for distinguishing early-onset/late-onset psoriasis (EOP/LOP) was set at 60 years, based on the incident psoriasis peak in UKB. A genetic instrument comprising 44 single-nucleotide polymorphisms associated with glycated haemoglobin (HbA1c), serving as a proxy for AMPK genetic risk score (negatively associated with AMPK activation), was employed as previously reported in the literature. Log-binomial models were used to estimate the effect size of AMPK regarding relative risk (RR) and 95% CI. RESULTS A total of 407 159 participants were analysed, including 9126 EOP and 3324 LOP. The AMPK genetic risk score was associated with a 12.4% increase in the risk of LOP in men (RR = 1.124, 95% CI: 1.022-1.236). This association was not significant for EOP or women. AMPK genetic risk score exhibited an elevated risk of ischemic heart disease (RR = 1.217, 95% CI 1.062-1.395) in male psoriasis patients. CONCLUSIONS AMPK activation may protect against LOPs and associated ischemic heart disease in men. A sex-specific, comorbidity-targeted intervention for psoriasis is needed.
Collapse
Affiliation(s)
- Yi Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Danrong Jing
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Guowei Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenwei Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Yehong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Minxue Shen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
44
|
Gao WY, Chen PY, Hsu HJ, Liou JW, Wu CL, Wu MJ, Yen JH. Xanthohumol, a prenylated chalcone, regulates lipid metabolism by modulating the LXRα/RXR-ANGPTL3-LPL axis in hepatic cell lines and high-fat diet-fed zebrafish models. Biomed Pharmacother 2024; 174:116598. [PMID: 38615609 DOI: 10.1016/j.biopha.2024.116598] [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: 01/18/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024] Open
Abstract
Angiopoietin-like 3 (ANGPTL3) acts as an inhibitor of lipoprotein lipase (LPL), impeding the breakdown of triglyceride-rich lipoproteins (TGRLs) in circulation. Targeting ANGPTL3 is considered a novel strategy for improving dyslipidemia and atherosclerotic cardiovascular diseases (ASCVD). Hops (Humulus lupulus L.) contain several bioactive prenylflavonoids, including xanthohumol (Xan), isoxanthohumol (Isoxan), 6-prenylnaringenin (6-PN), and 8-prenylnaringenin (8-PN), with the potential to manage lipid metabolism. The aim of this study was to investigate the lipid-lowering effects of Xan, the effective prenylated chalcone in attenuating ANGPTL3 transcriptional activity, both in vitro using hepatic cells and in vivo using zebrafish models, along with exploring the underlying mechanisms. Xan (10 and 20 μM) significantly reduced ANGPTL3 mRNA and protein expression in HepG2 and Huh7 cells, leading to a marked decrease in secreted ANGPTL3 proteins via hepatic cells. In animal studies, orally administered Xan significantly alleviated plasma triglyceride (TG) and cholesterol levels in zebrafish fed a high-fat diet. Furthermore, it reduced hepatic ANGPTL3 protein levels and increased LPL activity in zebrafish models, indicating its potential to modulate lipid profiles in circulation. Furthermore, molecular docking results predicted that Xan exhibits a higher binding affinity to interact with liver X receptor α (LXRα) and retinoic acid X receptor (RXR) than their respective agonists, T0901317 and 9-Cis-retinoic acid (9-Cis-RA). We observed that Xan suppressed hepatic ANGPTL3 expression by antagonizing the LXRα/RXR-mediated transcription. These findings suggest that Xan ameliorates dyslipidemia by modulating the LXRα/RXR-ANGPTL3-LPL axis. Xan represents a novel potential inhibitor of ANGPTL3 for the prevention or treatment of ASCVD.
Collapse
Affiliation(s)
- Wan-Yun Gao
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan
| | - Pei-Yi Chen
- Laboratory of Medical Genetics, Genetic Counseling Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970374, Taiwan; Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970374, Taiwan
| | - Hao-Jen Hsu
- Department of Biomedical Science and Engineering, Tzu Chi University, Hualien 970374, Taiwan
| | - Je-Wen Liou
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Chia-Ling Wu
- Laboratory of Medical Genetics, Genetic Counseling Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970374, Taiwan
| | - Ming-Jiuan Wu
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717301, Taiwan
| | - Jui-Hung Yen
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan; Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 970374, Taiwan.
| |
Collapse
|
45
|
Bavaresco A, Mazzeo P, Lazzara M, Barbot M. Adipose tissue in cortisol excess: What Cushing's syndrome can teach us? Biochem Pharmacol 2024; 223:116137. [PMID: 38494065 DOI: 10.1016/j.bcp.2024.116137] [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: 10/31/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Endogenous Cushing's syndrome (CS) is a rare condition due to prolonged exposure to elevated circulating cortisol levels that features its typical phenotype characterised by moon face, proximal myopathy, easy bruising, hirsutism in females and a centripetal distribution of body fat. Given the direct and indirect effects of hypercortisolism, CS is a severe disease burdened by increased cardio-metabolic morbidity and mortality in which visceral adiposity plays a leading role. Although not commonly found in clinical setting, endogenous CS is definitely underestimated leading to delayed diagnosis with consequent increased rate of complications and reduced likelihood of their reversal after disease control. Most of all, CS is a unique model for systemic impairment induced by exogenous glucocorticoid therapy that is commonly prescribed for a number of chronic conditions in a relevant proportion of the worldwide population. In this review we aim to summarise on one side, the mechanisms behind visceral adiposity and lipid metabolism impairment in CS during active disease and after remission and on the other explore the potential role of cortisol in promoting adipose tissue accumulation.
Collapse
Affiliation(s)
- Alessandro Bavaresco
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy
| | - Pierluigi Mazzeo
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy
| | - Martina Lazzara
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy
| | - Mattia Barbot
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy.
| |
Collapse
|
46
|
Senavirathna T, Shafaei A, Lareu R, Balmer L. Unlocking the Therapeutic Potential of Ellagic Acid for Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Antioxidants (Basel) 2024; 13:485. [PMID: 38671932 PMCID: PMC11047720 DOI: 10.3390/antiox13040485] [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: 03/05/2024] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity is in epidemic proportions in many parts of the world, contributing to increasing rates of non-alcoholic fatty liver disease (NAFLD). NAFLD represents a range of conditions from the initial stage of fatty liver to non-alcoholic steatohepatitis (NASH), which can progress to severe fibrosis, through to hepatocellular carcinoma. There currently exists no treatment for the long-term management of NAFLD/NASH, however, dietary interventions have been investigated for the treatment of NASH, including several polyphenolic compounds. Ellagic acid is one such polyphenolic compound. Nutraceutical food abundant in ellagic acid undergoes initial hydrolysis to free ellagic acid within the stomach and small intestine. The proposed mechanism of action of ellagic acid extends beyond its initial therapeutic potential, as it is further broken down by the gut microbiome into urolithin. Both ellagic acid and urolithin have been found to alleviate oxidative stress, inflammation, and fibrosis, which are associated with NAFLD/NASH. While progress has been made in understanding the pharmacological and biological activity of ellagic acid and its involvement in NAFLD/NASH, it has yet to be fully elucidated. Thus, the aim of this review is to summarise the currently available literature elucidating the therapeutic potential of ellagic acid and its microbial-derived metabolite urolithin in NAFLD/NASH.
Collapse
Affiliation(s)
- Tharani Senavirathna
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia;
| | - Armaghan Shafaei
- Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Perth, WA 6027, Australia;
| | - Ricky Lareu
- Curtin Medical School and Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA 6845, Australia
| | - Lois Balmer
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia;
| |
Collapse
|
47
|
Galal MA, Al-Rimawi M, Hajeer A, Dahman H, Alouch S, Aljada A. Metformin: A Dual-Role Player in Cancer Treatment and Prevention. Int J Mol Sci 2024; 25:4083. [PMID: 38612893 PMCID: PMC11012626 DOI: 10.3390/ijms25074083] [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: 03/06/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer continues to pose a significant global health challenge, as evidenced by the increasing incidence rates and high mortality rates, despite the advancements made in chemotherapy. The emergence of chemoresistance further complicates the effectiveness of treatment. However, there is growing interest in the potential of metformin, a commonly prescribed drug for type 2 diabetes mellitus (T2DM), as an adjuvant chemotherapy agent in cancer treatment. Although the precise mechanism of action of metformin in cancer therapy is not fully understood, it has been found to have pleiotropic effects, including the modulation of metabolic pathways, reduction in inflammation, and the regulation of cellular proliferation. This comprehensive review examines the anticancer properties of metformin, drawing insights from various studies conducted in vitro and in vivo, as well as from clinical trials and observational research. This review discusses the mechanisms of action involving both insulin-dependent and independent pathways, shedding light on the potential of metformin as a therapeutic agent for different types of cancer. Despite promising findings, there are challenges that need to be addressed, such as conflicting outcomes in clinical trials, considerations regarding dosing, and the development of resistance. These challenges highlight the importance of further research to fully harness the therapeutic potential of metformin in cancer treatment. The aims of this review are to provide a contemporary understanding of the role of metformin in cancer therapy and identify areas for future exploration in the pursuit of effective anticancer strategies.
Collapse
Affiliation(s)
- Mariam Ahmed Galal
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Mohammed Al-Rimawi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | | | - Huda Dahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Samhar Alouch
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| |
Collapse
|
48
|
Wang Y, Shi Y, Peng X, Li T, Liang C, Wang W, Zhou M, Yang J, Cheng J, Zhang Z, Hou L. Biochemotaxis-Oriented Engineering Bacteria Expressing GLP-1 Enhance Diabetes Therapy by Regulating the Balance of Immune. Adv Healthc Mater 2024; 13:e2303958. [PMID: 38253022 DOI: 10.1002/adhm.202303958] [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: 11/12/2023] [Indexed: 01/24/2024]
Abstract
Glucagon like peptide-1 (GLP-1) is an effective hypoglycemic drug that can repair the pancreas β cells and promote insulin secretion. However, GLP-1 has poor stability and lacks of target ability, which makes it difficult to reach the site of action to exert its efficacy. Here, GLP-1-expressing plasmids are introduced into the Escherichia coli Nissle 1917 (EcN) and a lipid membrane is formed through simple self-assembly on its surface, resulting in an oral delivery system (LEG) capable of resisting the harsh environment of the gastrointestinal tract. The system utilizes the chemotactic properties of probiotics to achieve efficient enrichment at the pancreatic site, and protects islet β cells from destruction by regulating the balance of immune cells. More interestingly, LEG not only continuously produces GLP-1 to restore pancreatic islet β cell function and secrete insulin to control blood sugar levels, but also regulates the intestinal flora and increases the richness and diversity of probiotics. In mice diabetes models, oral administration of LEG only once every other day has good biosafety and compliance, and achieves long-term control of blood glucose. Therefore, this strategy not only provides an oral delivery platform for pancreatic targeting, but also opens up new avenues for reversing diabetes.
Collapse
Affiliation(s)
- Yifei Wang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yupeng Shi
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xueyuan Peng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Tongtong Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chenglin Liang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenhao Wang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mengyang Zhou
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiali Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
49
|
Zhu Z, Ren W, Li S, Gao L, Zhi K. Functional significance of O-linked N-acetylglucosamine protein modification in regulating autophagy. Pharmacol Res 2024; 202:107120. [PMID: 38417774 DOI: 10.1016/j.phrs.2024.107120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/16/2024] [Accepted: 02/24/2024] [Indexed: 03/01/2024]
Abstract
Autophagy is a core molecular pathway that preserves cellular and organismal homeostasis. Being susceptible to nutrient availability and stress, eukaryotic cells recycle or degrade internal components via membrane transport pathways to provide sustainable biological molecules and energy sources. The dysregulation of this highly conserved physiological process has been strongly linked to human disease. Post-translational modification, a mechanism that regulates protein function, plays a crucial role in autophagy regulation. O-linked N-acetylglucosamine protein modification (O-GlcNAcylation), a monosaccharide post-translational modification of intracellular proteins, is essential in nutritional and stress regulatory mechanisms. O-GlcNAcylation has emerged as an essential regulatory mechanism of autophagy. It regulates autophagy throughout its lifetime by targeting the core components of the autophagy regulatory network. This review provides an overview of the O-GlcNAcylation of autophagy-associated proteins and their regulation and function in the autophagy pathway. Therefore, this article may contribute to further understanding of the role of O-GlcNAc-regulated autophagy and provide new perspectives for the treatment of human diseases.
Collapse
Affiliation(s)
- Zhuang Zhu
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao 266555, China; School of Stomatology, Qingdao University, Qingdao 266003, China; Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao 266555, China.
| | - Wenhao Ren
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao 266555, China; Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao 266555, China.
| | - Shaoming Li
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao 266555, China; School of Stomatology, Qingdao University, Qingdao 266003, China; Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao 266555, China.
| | - Ling Gao
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao 266555, China; School of Stomatology, Qingdao University, Qingdao 266003, China; Key Lab of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266003, China; Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao 266555, China.
| | - Keqian Zhi
- Department of Oral and Maxillofacial Reconstruction, the Affiliated Hospital of Qingdao University, Qingdao 266555, China; School of Stomatology, Qingdao University, Qingdao 266003, China; Key Lab of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266003, China; Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao 266555, China.
| |
Collapse
|
50
|
Yang X, Yang J, Lyu M, Li Y, Liu A, Shen B. The α subunit of AMP-activated protein kinase is critical for the metabolic success and tachyzoite proliferation of Toxoplasma gondii. Microb Biotechnol 2024; 17:e14455. [PMID: 38635138 PMCID: PMC11025617 DOI: 10.1111/1751-7915.14455] [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: 01/10/2024] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 04/19/2024] Open
Abstract
Toxoplasma gondii is a zoonotic parasite infecting humans and nearly all warm-blooded animals. Successful parasitism in diverse hosts at various developmental stages requires the parasites to fine tune their metabolism according to environmental cues and the parasite's needs. By manipulating the β and γ subunits, we have previously shown that AMP-activated protein kinase (AMPK) has critical roles in regulating the metabolic and developmental programmes. However, the biological functions of the α catalytic subunit have not been established. T. gondii encodes a canonical AMPKα, as well as a KIN kinase whose kinase domain has high sequence similarities to those of classic AMPKα proteins. Here, we found that TgKIN is dispensable for tachyzoite growth, whereas TgAMPKα is essential. Depletion of TgAMPKα expression resulted in decreased ATP levels and reduced metabolic flux in glycolysis and the tricarboxylic acid cycle, confirming that TgAMPK is involved in metabolic regulation and energy homeostasis in the parasite. Sequential truncations at the C-terminus found an α-helix that is key for the function of TgAMPKα. The amino acid sequences of this α-helix are not conserved among various AMPKα proteins, likely because it is involved in interactions with TgAMPKβ, which only have limited sequence similarities to AMPKβ in other eukaryotes. The essential role of the less conserved C-terminus of TgAMPKα provides opportunities for parasite specific drug designs targeting TgAMPKα.
Collapse
Affiliation(s)
- Xuke Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei ProvinceChina
- Research Center for Infectious Diseases, Department of Pathogen Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Jichao Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei ProvinceChina
| | - Mengyu Lyu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei ProvinceChina
| | - Yaqiong Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei ProvinceChina
| | - Anqi Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei ProvinceChina
| | - Bang Shen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei ProvinceChina
- Hubei Hongshan LaboratoryWuhanHubei ProvinceChina
- Key Laboratory of Preventive Medicine in Hubei ProvinceHuazhong Agricultural UniversityWuhanHubei ProvinceChina
- Shenzhen Institute of Nutrition and HealthHuazhong Agricultural UniversityShenzhenGuangdong ProvinceChina
| |
Collapse
|