1
|
Hossain L, Gomes KP, Safarpour S, Gibson SB. The microenvironment of secondary lymphedema. The key to finding effective treatments? Biochim Biophys Acta Mol Basis Dis 2025; 1871:167677. [PMID: 39828048 DOI: 10.1016/j.bbadis.2025.167677] [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: 10/15/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Lymphedema is characterized by the swelling of extremities due to the accumulation of interstitial fluids. It is a painful and devastating disease that increases the risk of infections and destroys patients' quality of life. Secondary lymphedema is caused by damage to the lymphatic system due to infections, obesity, surgery, and cancer treatments. This damage fails to be repaired and leads to fluid accumulation, tissue remodeling, inflammation, and ultimately fibrosis. The lymphedema microenvironment is altered by stress, immune dysfunction, and changes in metabolism. Stress in the microenvironment includes increased hypoxia and oxidative stress but how this contributes to lymphedema progression is unclear. The immune system plays a critical role in lymphedema through T cell helper type 2 (Th2) immune responses and the infiltration of macrophages into lymphedematous tissue. The inflammatory cytokines released by immune cells lead to tissue remodeling and fibrosis. There are also changes in metabolism in the lymphedema microenvironment with altered lipid oxidation, ketone body oxidation, and glycolysis. How these changes affect lymphedema and treatment interventions has been the focus of clinical trials. Lymphedema is also associated with cancer and obesity through damage to the lymphatic system. This review will illustrate microenvironmental changes in lymphedema and how this relates to cancer and obesity. In addition, we will discuss new therapeutic strategies to treat lymphedema. Finally, we will address the prospects of lymphedema research in the context of the microenvironment.
Collapse
Affiliation(s)
- Lazina Hossain
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Cross Cancer Institute, Alberta Health Services, Edmonton, Alberta, Canada
| | - Karina P Gomes
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Cross Cancer Institute, Alberta Health Services, Edmonton, Alberta, Canada
| | - Samaneh Safarpour
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Cross Cancer Institute, Alberta Health Services, Edmonton, Alberta, Canada
| | - Spencer B Gibson
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Cross Cancer Institute, Alberta Health Services, Edmonton, Alberta, Canada.
| |
Collapse
|
2
|
Selima RM, Saleem IA, Shawki MM, Darwish AA, Yehia MA, Mohamed EI. Effect of gold nanoparticles treatment on rats-induced obesity by evaluating body-composition directly and indirectly via bioelectric impedance analysis. Sci Rep 2025; 15:4942. [PMID: 39929902 PMCID: PMC11811030 DOI: 10.1038/s41598-025-87971-z] [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: 09/11/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
Obesity is a metabolic disease characterized by an imbalance between caloric intake and expenditure, leading to excess fat and increasing the risk of various health conditions. This study compares the anti-obesity effects of gold nanoparticles (AuNPs) to orlistat in an experimental model of induced obesity in Wistar Albino rats. In addition to negative and positive control rats, obese rats were treated with variable daily and weekly doses of AuNPs and daily orlistat for nine weeks. Bioelectric impedance analysis (BIA) and dissection techniques were used to indirectly and directly measure body-composition in all rat groups. Hepatic and renal function and ultrastructure were assessed by blood biochemical and histological examinations to detect treatment-related alterations. High doses of AuNPs reduced body fat, increased muscle mass, improved dyslipidemia, glycemia, and antioxidant effects in obese rats, and restored normal TG, FBG, and MDA levels by reducing obesity-related oxidative damage. Histological and ultrastructural examinations showed that these high doses repaired liver and kidney cells, and reduced fat accumulation and body weight compared to the standard treatment for obesity by orlistat.
Collapse
Affiliation(s)
- Rana M Selima
- Medical Biophysics Department, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, Alexandria, 5433005, Egypt
| | - Israa A Saleem
- Optometry Department, Technical Medical Institute, Erbil Polytechnic University, Erbil, Iraq
| | - Mamdouh M Shawki
- Medical Biophysics Department, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, Alexandria, 5433005, Egypt
| | - Amira A Darwish
- Medical Laboratory Technology Department, Faculty of Applied Health Sciences Technology, Pharos University, Alexandria, Egypt
| | - Mona A Yehia
- Histochemistry and Cell Biology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ehab I Mohamed
- Medical Biophysics Department, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, Alexandria, 5433005, Egypt.
| |
Collapse
|
3
|
Gutierrez MW, van Tilburg Bernardes E, Ren E, Kalbfleisch KN, Day M, Lameu EL, Glatthardt T, Mercer EM, Sharma S, Zhang H, Al-Azawy A, Chleilat F, Hirota SA, Reimer RA, Arrieta MC. Early-life gut mycobiome core species modulate metabolic health in mice. Nat Commun 2025; 16:1467. [PMID: 39922818 PMCID: PMC11807121 DOI: 10.1038/s41467-025-56743-8] [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: 02/12/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The gut microbiome causally contributes to obesity; however, the role of fungi remains understudied. We previously identified three core species of the infant gut mycobiome (Rhodotorula mucilaginosa, Malassezia restricta and Candida albicans) that correlated with body mass index, however their causal contributions to obesity development are unknown. Here we show the effects of early-life colonization by these fungal species on metabolic health in gnotobiotic mice fed standard (SD) or high-fat-high-sucrose (HFHS) diets. Each species resulted in bacterial microbiome compositional and functional differences. R. mucilaginosa and M. restricta increased adiposity in mice fed SD, while only R. mucilaginosa exacerbated metabolic disease. In contrast, C. albicans resulted in leanness and resistance to diet-induced obesity. Intestinal nutrient transporter expression was unaffected by the presence of fungi in jejunal enteroids, yet the immune landscape in white adipose tissue was distinctly impacted by each fungal species, suggesting that these phenotypes may be a result of fungal immune regulation. This work revealed that three common fungal colonizers have distinct causal influences on obesity and metabolic inflammation and justifies the consideration of fungi in microbiome research on host metabolism.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Erik van Tilburg Bernardes
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ellen Ren
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Kristen N Kalbfleisch
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Madeline Day
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ewandson Luiz Lameu
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Thaís Glatthardt
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Emily M Mercer
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Sunita Sharma
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Hong Zhang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ali Al-Azawy
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Faye Chleilat
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Simon A Hirota
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada.
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada.
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
4
|
Li J, Ge Z, Li C, Ran H, Zhang Y, Xiang Y. METRNL exerts cytoprotective effects on EPCs via regulation of the E2F1-TXNIP axis in obese limb ischemia. Cell Signal 2025; 126:111528. [PMID: 39603439 DOI: 10.1016/j.cellsig.2024.111528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/10/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Obesity increases cardiovascular disease risk by impairing angiogenesis, primarily through dysfunction of endothelial progenitor cells (EPCs). METRNL, a recently identified secreted protein, exhibits diverse biological activities. However, its impact on EPC function and its role in obesity-related microvascular dysfunction remain unclear. This study aims to investigate the effects of METRNL on EPC function and its potential therapeutic mechanisms for promoting angiogenesis. METHOD In vitro, human EPCs derived from peripheral and umbilical cord blood were treated with recombinant METRNL protein (rMETRNL) and exposed to palmitic acid (PA). EPC proliferation, migration, and tube formation were assessed. Apoptosis and pyroptosis levels were evaluated using Western blotting, flow cytometry, scanning electron microscopy (SEM), immunofluorescence (IF), and enzyme-linked immunosorbent assay (ELISA). RNA sequencing, ChIP, and dual-luciferase assays were performed to investigate the regulatory mechanisms. In vivo, an obese mouse model with hind limb ischemia received local injections of METRNL-overexpressing EPCs in the ischemic muscle. Blood flow recovery was monitored using laser Doppler flowmetry and CD31 immunofluorescence. RESULTS Replenishment of METNRL alleviated PA-induced apoptosis and pyroptosis of EPCs, while simultaneously enhancing their proliferation, migration, and tube formation. Mechanistically, RNA sequencing revealed that rMETRNL restoration downregulated E2F1 expression, and the protective effects of METRNL were partially reversed by E2F1 overexpression. Further, E2F1 was found to bind the TXNIP promoter region, promoting TXNIP transcription. Elevated TXNIP levels counteracted the beneficial effects of rMETRNL on EPC function in the presence of PA. In vivo, the transplantation of METRNL-overexpressing EPCs into the ischemic hind limbs of obese mice promoted angiogenesis, as evidenced by improved blood flow recovery and increased CD31 immunofluorescence in the ischemic tissues. CONCLUSION Our research emphasizes the potential of METRNL in reducing EPC cellular pyroptosis and promoting angiogenesis by inhibiting the E2F1-TXNIP signaling pathway. METRNL shows promise in treating obesity-related cardiovascular diseases through angiogenic therapy.
Collapse
Affiliation(s)
- Jing Li
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Shanghai 200000, China
| | - Zhuowang Ge
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Shanghai 200000, China
| | - Chengsi Li
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Shanghai 200000, China
| | - Hui Ran
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Shanghai 200000, China
| | - Yachen Zhang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Shanghai 200000, China.
| | - Yin Xiang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Shanghai 200000, China.
| |
Collapse
|
5
|
Lungruammit N, Pintana H, Pratchayasakul W, Songtrai S, Kaewsuwan S, Ittichaichareon J, Chattipakorn N, Chattipakorn SC. Cyclosorus terminans extract mitigates submandibular gland changes associated with high-fat diet consumption in male rats. Arch Oral Biol 2025; 170:106127. [PMID: 39561524 DOI: 10.1016/j.archoralbio.2024.106127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVES To investigate whether the prophylactic effect of Cyclosorus terminans extract mitigates metabolic impairment and submandibular gland changes, as indicated by increased aquaporin5 expression, decreased fibrosis, oxidative stress and inflammation, improved mitochondrial homeostasis/dynamics, and decreased cell death in the submandibular glands of high-fat diet (HFD)-feeding rats. METHODS Thirty-two male Wistar rats were assigned to either a normal diet (ND) as control rats (n=8) or a HFD (n=24) for 12 weeks. The HFD-treated rats were divided into 3 subgroups to receive either: 1) vehicle (HDV), 2) Cyclosorus terminans at a dose of 100 mg/kg/d (HF100), or 3) Cyclosorus terminans at a dose of 200 mg/kg/d (HF200). At week 13, metabolic parameters, systemic oxidative stress, and submandibular gland parameters were assessed. RESULTS Twelve weeks of HFD-feeding rats induced obese-insulin resistance and submandibular gland changes. Both HF100- and HF200-treated groups improved metabolic parameters and prevented gland changes by reducing fibrosis (TGF-β and p-38), malondialdehyde levels, inflammation (TNF-α, NF-κB, and Ifng), and cell death markers (Caspase 3, GSDMD, and MLKL). Both treatments supported balanced mitochondrial homeostasis/dynamics, as indicated by regulating related genes (Cpt1b, Ndufb8, Mfn1, Mfn2, Opa1, and Dnm1l). However, only the HF200-treated rats restored aquaporin-5 and antioxidants (SOD2 and GPX4) expression to control levels. CONCLUSIONS Cyclosorus terminans mitigates metabolic disturbances and submandibular gland changes in HFD-feeding rats. The high dose was more effective, improving gland function by increasing aquaporin5 and antioxidants. These results suggest Cyclosorus terminans may be a promising therapeutic for metabolic disturbances and submandibular gland changes in obese-insulin resistant patients.
Collapse
Affiliation(s)
- Nopphakhun Lungruammit
- Department of Oral Biology and Oral Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Hiranya Pintana
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sujinda Songtrai
- Faculty of Medical Technology, Rangsit University, Pathumthan 12000, Thailand
| | - Sireewan Kaewsuwan
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand; Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 90110, Thailand
| | - Jitjiroj Ittichaichareon
- Department of Oral Biology and Oral Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Department of Oral Biology and Oral Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
6
|
Ezhilarasan D, Karthikeyan S, Najimi M, Vijayalakshmi P, Bhavani G, Jansi Rani M. Preclinical liver toxicity models: Advantages, limitations and recommendations. Toxicology 2025; 511:154020. [PMID: 39637935 DOI: 10.1016/j.tox.2024.154020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Experimental animal models are crucial for elucidating the pathophysiology of liver injuries and for assessing new hepatoprotective agents. Drugs and chemicals such as acetaminophen, isoniazid, valproic acid, ethanol, carbon tetrachloride (CCl4), dimethylnitrosamine (DMN), and thioacetamide (TAA) are metabolized by the CYP2E1 enzyme, producing hepatotoxic metabolites that lead to both acute and chronic liver injuries. In experimental settings, acetaminophen (centrilobular necrosis), carbamazepine (centrilobular necrosis and inflammation), sodium valproate (necrosis, hydropic degeneration and mild inflammation), methotrexate (sinusoidal congestion and inflammation), and TAA (centrilobular necrosis and inflammation) are commonly used to induce various types of acute liver injuries. Repeated and intermittent low-dose administration of CCl4, TAA, and DMN activates quiescent hepatic stellate cells, transdifferentiating them into myofibroblasts, which results in abnormal extracellular matrix production and fibrosis induction, more rapidly with DMN and CCL4 than TAA (DMN > CCl4 > TAA). Regarding toxicity and mortality, CCl4 is more toxic than DMN and TAA (CCl4 > DMN > TAA). Models used to induce metabolic dysfunction-associated liver disease (MAFLD) vary, but MAFLD's multifactorial nature driven by factors like obesity, fatty liver, dyslipidaemia, type II diabetes, hypertension, and cardiovascular disease makes it challenging to replicate human metabolic dysfunction-associated steatohepatitis accurately. From an experimental point of view, the degree and pattern of liver injury are influenced by various factors, including the type of hepatotoxic agent, exposure duration, route of exposure, dosage, frequency of administration, and the animal model utilized. Therefore, there is a pressing need for standardized protocols and regulatory guidelines to streamline the selection of animal models in preclinical studies.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Sivanesan Karthikeyan
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Paramasivan Vijayalakshmi
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Asan Memorial Dental College and Hospital, Chengalpattu, Tamil Nadu, India
| | - Ganapathy Bhavani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Muthukrishnan Jansi Rani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
7
|
Elkins M, Horrelt M, Woods B, Lawton S, Ohsumi TK, Fleischman A, Taudte V, Chou J. Overfeeding and overweight rapidly reprogram inflammatory signaling. Clin Immunol 2025; 271:110428. [PMID: 39788355 DOI: 10.1016/j.clim.2025.110428] [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: 10/30/2024] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Epidemiologic studies have shown a continuous increase in mortality risk associated with overweight, thus highlighting the health risks beginning before the onset of obesity. However, early changes in inflammatory signaling induced by an obesogenic diet remain largely unknown since studies of obesity typically utilize models induced by months of continuous exposure to a high-fat diet. Here, we investigated how short-term overfeeding remodels inflammatory signaling. We developed and characterized a mouse model of overweight induced by seven days of the Western diet enriched in saturated fats and sucrose, compared to the standard, low-fat laboratory diet or a long-term Western diet for 22 weeks. The short-term Western diet caused a median weight gain of 6 %, while the long-term Western diet increased weight by 92 %. Circulating levels of cholesterol, triglycerides, insulin, and leptin were increased by both diets, but only the long-term Western diet caused transaminitis and significant hepatic steatosis. Both models reduced the alpha and beta diversity of the microbiome. Tryptophan metabolism was perturbed by both models; the long-term Western diet also affected histidine and vitamin B6 metabolism. The short-term and long-term Western diets increased expression of TLR4 on peritoneal immune cells and TLR4-driven plasma levels of proinflammatory cytokines comparably, showing one week of the Western diet was sufficient for inducing inflammation typical of chronic obesity. These findings highlight the importance of diet not only in preclinical studies, but also in the clinical care of individuals with inflammatory disorders.
Collapse
Affiliation(s)
- Megan Elkins
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States of America.
| | - Merle Horrelt
- Research Group Translational Metabolomics, Core Facility for Metabolomics, Philipps University Marburg, Marburg, Germany
| | - Brian Woods
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States of America
| | - Samira Lawton
- Microbiome Analysis, Transnetyx, Memphis, TN, United States of America
| | | | - Amy Fleischman
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States of America
| | - Verena Taudte
- Research Group Translational Metabolomics, Core Facility for Metabolomics, Philipps University Marburg, Marburg, Germany
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States of America.
| |
Collapse
|
8
|
Nagalievska M, Vilkiv K, Mishchenko L, Sybirna N. Effects of yacon on carbohydrates and lipid metabolism, oxidative-nitrative stress markers changes in rats with experimental metabolic syndrome. Heliyon 2025; 11:e42147. [PMID: 39925358 PMCID: PMC11804531 DOI: 10.1016/j.heliyon.2025.e42147] [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: 07/13/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Background and aim Smallanthus sonchifolius (Poepp. and Hendl.) H. Robinson, commonly known as yacon, is a medicinal species recognized for its therapeutic properties. The hypoglycemic and hypolipidemic effects of yacon root tubers have been well-documented across various models of metabolic syndrome. However, research on the effects of biologically active compounds derived from yacon leaves remains limited. The aim of the study was to evaluate the hypoglycemic, hypolipidemic and antioxidant effects of an aqueous extract of yacon leaves under the fructose-induced experimental metabolic syndrome. Experimental procedure In the study were used male Wistar white rats, in which metabolic syndrome was induced by consuming a 10 % fructose solution. Animals were given an aqueous extract of yacon leaves at a dose of 500 mg per kilogram of body weight for seven and fourteen days following the establishment of metabolic syndrome. Hypoglycemic (glucose and glycosylate hemoglobin concentrations) and hypolipidemic (plasma cholesterol, triglycerides, low-density lipoproteins and high-density lipoproteins levels) effects and antioxidant activity (activity of superoxide dismutase, catalase, glutathione peroxidase, NO-synthase, the content of nitrite anion (NO₂‾) and nitrate anion (NO₃‾), content of carbonyl groups and thiobarbituric acid reactive substances) of extract were then evaluated. Results The 14 days use of aqueous extract of yacon for the treatment of fructose-induced metabolic syndrome leads to a decrease of animals' body weight (59.94 %), glucose concentration (10.33 %), glycosylated hemoglobin content (61.58 %), blood plasma triglycerides (50.35 %), cholesterol (24.46 %), low-density lipoproteins (21.56 %), as well as to increase in high-density lipoproteins concentration (29.29 %), paraoxonase activity (56.03 %). In animals with experimental MetS yacon cause oxidative-nitrative stress indicators normalization: increase in SOD (47.85 %) and GPO activity (16.55 %); decrease in TBARS content (23.77 %) and proteins oxidative modification products of neutral character (52.56 %); decrease in NOS activity (12.30 %), which was accompanied by a decrease in nitrate content (10.44 %). Conclusions The aqueous extract of yacon demonstrates significant hypoglycemic and hypolipidemic effects in a fructose-induced model of experimental metabolic syndrome, contributing to the normalization of both prooxidant and antioxidant states in rat erythrocytes.
Collapse
Affiliation(s)
- Mariia Nagalievska
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4 Hrushevskoho St., Lviv, 79005, Ukraine
| | - Khrystyna Vilkiv
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4 Hrushevskoho St., Lviv, 79005, Ukraine
| | - Lidiya Mishchenko
- Educational and Scientific Centre “Institute of Biology and Medicine” 64/13 Volodymyrska, 01601 Kyiv, Ukraine
| | - Nataliia Sybirna
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4 Hrushevskoho St., Lviv, 79005, Ukraine
- Collegium Medicum, Faculty of Biotechnology, University of Rzeszow, 8B Zelwerowicza St., Rzeszow 35-601, Poland
| |
Collapse
|
9
|
Wojcieszak J, Kuczyńska K, Leszczyńska A, Naraziński E, Cichalewska-Studzińska M. Access to high-fat diet results in increased sensitivity to the psychostimulant effects of MDPV in mice. Pharmacol Rep 2025:10.1007/s43440-025-00701-0. [PMID: 39869285 DOI: 10.1007/s43440-025-00701-0] [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: 09/26/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND The current study investigated the effects of high-fat diet on acute response to 3,4-methylenedioxypyrovalerone (MDPV) in mice. MDPV is a beta-cathinone derivative endowed with psychostimulant activity. Similarly to recreational substances, consumption of palatable food stimulates the mesolimbic dopaminergic system, resulting in neuroadaptive changes. METHODS Adolescent C57BL/6N mice were fed either control diet (CD), 10% of kcal from fat, or high-fat diet (HFD), 60% of kcal from fat. After eight weeks, one group of HFD-fed mice had their diet changed to CD for an additional two weeks. Fasting glucose levels and glucose tolerance were measured to detect impairment in glucose metabolism. Subsequently, the mice were treated with either MDPV (1 mg/kg) or saline, and their locomotor activity was measured. Using reverse transcriptase quantitative polymerase chain reaction (RT-qPCR), the expression of dopamine receptor D1 (Drd1), dopamine receptor D2 (Drd2), and FBJ osteosarcoma oncogene B (FosB) genes was measured in the striatum of mice. RESULTS Feeding with HFD caused obesity and glucose intolerance in mice. Restriction of fat reduced body mass and reversed impairment of glucose metabolism. HFD-fed mice responded to MDPV with higher potency than CD-fed counterparts, with an increased incidence of stereotypies. A change of diet partially reversed this effect. Downregulation of Drd2 was observed in the mice that switched from HFD to CD, whereas treatment with MDPV caused upregulation of FosB only in the CD-fed mice. CONCLUSIONS Current results suggest that obesity may increase sensitivity to psychostimulant effects of MDPV and elevate the risk of addiction as mice fed with HFD responded to acute treatment with MDPV with higher potency and showed tolerance of FosB induction in response to the drug.
Collapse
Affiliation(s)
- Jakub Wojcieszak
- Department of Pharmacodynamics, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland.
| | - Katarzyna Kuczyńska
- Department of Pharmacodynamics, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland
| | - Adrianna Leszczyńska
- Department of Pharmacodynamics, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland
| | - Eryk Naraziński
- Department of Pharmacodynamics, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland
| | | |
Collapse
|
10
|
Servin-Uribe RI, Castilla-Ramírez P, Ramírez IFP, Jiménez JP, Reynoso-Camacho R. In vivo Differential Effects of Extractable and Non-Extractable Phenolic Compounds from Grape Pomace on the Regulation of Obesity and Associated Metabolic Alterations. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2025; 80:37. [PMID: 39821690 DOI: 10.1007/s11130-024-01278-4] [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] [Accepted: 12/05/2024] [Indexed: 01/19/2025]
Abstract
Grape pomace (GP) is a by-product rich in phytochemicals, including extractable polyphenols (EPPs) and non-extractable polyphenols (NEPPs), which have distinct metabolic fates that may affect their biological activities. The benefits of GP have been reported in relation to obesity and its comorbidities, particularly when administered preventively focusing on EPPs. Therefore, the aim of this study was to investigate the effects of EPPs and NEPPs from GP as a treatment for obesity and its associated metabolic alterations. A previous comprehensive characterization of the selected GP revealed the most relevant individual compounds in the EPPs fraction (malvidin hexoside, (-)-epicatechin, quercetin, and procyanidin dimer B2 isomer II), as well as in the NEPPs fraction (hydroxybenzoic acid isomers I-II). The experiment was performed in obese rats with insulin resistance, treated for 8 weeks with EPPs or NEPPs grape pomace fractions (100 mg/kg). After the intervention, the HFFD + EPP group showed a significantly lower weight gain (9.6%) and body mass index (9.7%) compared to the HFFD group. While liver triglyceride levels were only significantly reduced in the HFFD + NEPP group (47%) compared to the HFFD group. Neither treatment resulted in a reduction of insulin resistance. Therefore, the supplementation with grape pomace phenolic fractions to an animal model of obesity exerted differential beneficial effects on body weight and liver lipid accumulation, overall contributing to an amelioration of some the metabolic alterations present in obesity, although not to aspects such as glycemic homeostasis.
Collapse
Affiliation(s)
- Rogelio I Servin-Uribe
- Departamento de Investigación y Posgrado en Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Qro., 76010, México
| | - Paloma Castilla-Ramírez
- Departamento de Investigación y Posgrado en Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Qro., 76010, México
| | - Iza F Pérez Ramírez
- Departamento de Investigación y Posgrado en Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Qro., 76010, México
| | - Jara Pérez Jiménez
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN-CSIC), José Antonio Novais 10, Madrid, 28040, Spain
- CIBER Diabetes and Associated Metabolic Diseases: Diabetes and Associated Metabolic Diseases Networking Biomedical Research Centre, CIBERDEM, Carlos III Health Institute (ISCIII), Madrid, 28029, España
| | - Rosalía Reynoso-Camacho
- Departamento de Investigación y Posgrado en Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Qro., 76010, México.
| |
Collapse
|
11
|
Tian Y, Gong J, He Z, Peng S, Huan Y, Cao H. Impact of protein intake from a caloric-restricted diet on liver lipid metabolism in overweight and obese rats of different sexes. Sci Rep 2025; 15:2340. [PMID: 39833384 PMCID: PMC11747403 DOI: 10.1038/s41598-025-86596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
In addition to being linked to an excess of lipid accumulation in the liver, being overweight or obese can also result in disorders of lipid metabolism. There is limited understanding regarding whether different levels of protein intake within an energy-restricted diet affect liver lipid metabolism in overweight and obese rats and whether these effects differ by gender, despite the fact that both high protein intake and calorie restriction can improve intrahepatic lipid. The purpose of this study is to explore the effects and mechanisms of different protein intakes within a calorie-restricted diet on liver lipid metabolism, and to investigate whether these effects exhibit gender differences. The Sprague-Dawley rats, which were half female and half male, were used to construct a rat model of overweight and obesity attributed to a high-fat diet. They were then split up into five groups: the normal control (NC) group, the model control (MC) group, the calorie-restricted low protein (LP) group, the calorie-restricted normal protein (NP) group, and the calorie-restricted high protein (HP) group. Body weight was measured weekly. Samples of plasma and liver were obtained after eight weeks and analyzed for glucose, triglycerides, cholesterol, and hormones in the plasma as well as the liver fat and factors involved in the liver's synthesis and degradation. For the male rats, compared to the HP group, the weight of liver fat in the LP and NP group was significantly higher (P < 0.05). However, for the female rats, there was no significant variation among the three calorie-restricted groups (P > 0.05). There was no significant variation in the concentration of total cholesterol (TC), very low density lipoprotein (VLDL), low density lipoprotein (LDL), and high density lipoprotein (HDL) among the three male calorie-restricted groups (P > 0.05), while the TC and VLDL concentrations in the female LP and NP group were significantly higher compared to those in the HP group (P < 0.05). Moreover, the trend of expression in the signaling pathways of adiponectin/phosphorylated AMP-activated protein kinase (p-AMPK) and adiponectin/peroxisome proliferators-activated receptor alpha (PPARα) in the liver was consistent with that of the liver fat content, and leptin acted in the same way as adiponectin. Compared with the three calorie-restricted groups, the expressions of nuclear sterol-regulatory element-binding protein-2 (nSREBP-2) and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase) involved in cholesterol synthesis and low-density lipoprotein receptor (LDLR) and cholesterol 7-alpha hydroxylase (CYP7A1) involved in cholesterol clearance in the MC group were significantly lower (P < 0.05). A 40% energy restriction can significantly reduce the body weight, body fat, liver fat, and the blood concentration of TG in both male and female overweight and obese rats, but it can significantly increase the blood concentration of TC in overweight and obese male rats. At the same time of 40% calorie restriction, increasing dietary protein intake to twice the normal protein intake has a stronger effect on promoting hepatic triglyceride oxidation and reducing liver fat content in the male overweight and obese rats by increasing the levels of adiponectin and leptin in the blood, and can also significantly reduce the plasma cholesterol concentration in the female overweight and obese rats through inhibiting cholesterol synthesis most likely by increasing glucagon level in the blood.
Collapse
Affiliation(s)
- Ying Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Yangzhou University, Yangzhou, China.
| | - Jiawei Gong
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Zhiyan He
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Suwen Peng
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Yuping Huan
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Hongpeng Cao
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| |
Collapse
|
12
|
Nkoubat Tchoundjwen S, Kamgang Tchawou AG, Mvongo C, Mfopa A, Ngakou Mukam J, Noubissi PA, Fankem GO, Kamgang R, Essame Oyono JL. A Cameroon Western Regions high-fat diet (MACAPOS 2) induces visceral obesity in rat. Heliyon 2025; 11:e41011. [PMID: 39758382 PMCID: PMC11699333 DOI: 10.1016/j.heliyon.2024.e41011] [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: 02/24/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
The prevalence of obesity increases yearly in the world. The traditional local diet of the Western Regions of Cameroon was suspected to be the main contributor to the high prevalence of obesity in these Regions. This study aimed to evaluate the effects of a Cameroon-comparable fat diet on visceral obesity in rats. Two groups of male Wistar rats were fed for four months with respectively a normal diet (ND) (3400 kcal/kg of food) and a high-fat diet (HFD) containing maize, cassava, palm oil, and sugar (MACAPOS 2): 35 % carbohydrate, 55 % fat and 10 % proteins (4730 kcal/kg of food). Lee index, body weight, food intake, blood and hepatic lipids, body fat, insulin resistance, glucose tolerance, glycemia, serum insulin, leptin, and adiponectin were evaluated. HFD significantly (P < 0.01) increased body weight and decreased food intake. After four months of diet, 88.8 % of HFD rats were obese (Lee index >30 g/cm), and HFD significantly increased visceral and subcutaneous fats compared to ND. HFD increased triglyceride, total cholesterol, Low-density lipoprotein-cholesterol levels, and the atherogenic index, while the high-density lipoprotein-cholesterol level was decreased. The hepatic triglyceride and total cholesterol levels significantly (P < 0.01) increased in HFD, compared to ND. In HFD, the fasting blood glucose, serum insulin, and leptin levels significantly (P < 0.01) increased, meanwhile adiponectin decreased. HFD-induced glucose intolerance and insulin resistance in rats. Based on our findings, we can conclude that HFD MACAPOS 2 can induce central obesity. Therefore, it can be used as a model of diet-induced obesity.
Collapse
Affiliation(s)
- Sandrine Nkoubat Tchoundjwen
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Armel Georges Kamgang Tchawou
- Laboratory of Human Metabolism and Non-Communicable Diseases, Research Centre on Health and Priority Pathologies, (IMPM), P.O. Box. 13033, Yaoundé, Cameroon
| | - Clémence Mvongo
- Department of Life Science, Higher Teacher Training College, University of Bertoua, Cameroon
| | - Adamou Mfopa
- Laboratory of Phytoprotection and Valorization of Genetics Resources, Biotechnology Centre Nkolbisson, P.O. Box 17673 Etetak, Yaoundé, Cameroon
- Laboratory of Human Metabolism and Non-Communicable Diseases, Research Centre on Health and Priority Pathologies, (IMPM), P.O. Box. 13033, Yaoundé, Cameroon
| | - Joseph Ngakou Mukam
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Paul Aimé Noubissi
- Department of Zoology and Animal Physiology, Faculty of Science, University of Buea, Cameroon
| | - Gaetan Olivier Fankem
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - René Kamgang
- Laboratory of Human Metabolism and Non-Communicable Diseases, Research Centre on Health and Priority Pathologies, (IMPM), P.O. Box. 13033, Yaoundé, Cameroon
| | - Jean Louis Essame Oyono
- Laboratory of Human Metabolism and Non-Communicable Diseases, Research Centre on Health and Priority Pathologies, (IMPM), P.O. Box. 13033, Yaoundé, Cameroon
| |
Collapse
|
13
|
Kim EJ, Lee SH, Kim TH, Lee J, Choi CH, Lee SJ. Insect chitosan derived from Hermetia illucens larvae suppresses adipogenic signaling and promotes the restoration of gut microbiome balance. Int J Biol Macromol 2025; 284:138168. [PMID: 39613084 DOI: 10.1016/j.ijbiomac.2024.138168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
Chitosan, the deacetylated form of chitin, is considered a valuable source of compounds in the feed and food industries. However, the impact of Hermetia illucens larvae chitosan (HCS) with specific physicochemical characteristics on obesity mediated by lipid accumulation and microbiome dysbiosis has not been fully elucidated. We purified HCS with a low molecular weight (84 kDa), low crystallinity, and a high deacetylation rate, characterizing it through several analytical techniques, including gel permeation chromatography, FT-IR, 1H NMR, FE-SEM, and XRD analysis. HCS effectively inhibited the differentiation of 3T3-L1 preadipocytes by suppressing the production of reactive oxygen species. The adipogenic signaling of preadipocytes, mediated by the phosphorylation of mTOR and PPARγ, which are essential for the expression of fatty acid synthase, was attenuated by HCS. In mouse models fed high-fat diets, the oral administration of HCS prevented changes in white adipose tissue and liver weight and reduced plasma levels of total cholesterol. Additionally, the analysis of the microbiota using 16S rRNA revealed that HCS improved dysbiosis by modulating the composition and abundance of specific bacterial genera, including F. rodentium, L. gasseri, L. reuteri, and L. murinus. These findings highlight the potential of HCS as a candidate for the treatment of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Eun-Ju Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Seok-Hui Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Tae Hoon Kim
- FoodyWorm, Co. Ltd., 76 Geumwangtekeuno 3-gil, Geumwang-eup, Eumseong-gun, Chungcheongbuk-do 27680, Republic of Korea
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Chang-Hyung Choi
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
14
|
Krishnan I, Ling MTM, Ng MH, Law JX, Yusof MRM, Thangarajah T, Mahmood Z, Uda Zahli NI, Rajamanickam S, Subramani B, Lokanathan Y. Efficacy of Fetal Wharton's Jelly Mesenchymal Stem Cells-Derived Small Extracellular Vesicles in Metabolic Syndrome. Biomolecules 2025; 15:44. [PMID: 39858439 PMCID: PMC11763124 DOI: 10.3390/biom15010044] [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: 10/29/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVE Metabolic syndrome (MetS) is characterized by abdominal obesity, increased blood pressure (BP), fasting blood glucose (FBG) and triglyceride levels, and reduced high-density lipoprotein (HDL) levels. This study aims to investigate the efficacy of the Wharton's jelly mesenchymal stem cells (WJMSCs)-derived small extracellular vesicles' (sEVs) preparations in managing MetS. METHOD Twenty-four rats were fed with a high-fat and high-fructose diet to induce MetS for 16 weeks and randomized into three groups (n = 8/group): a MetS Control group treated with normal saline, MetS Low Dose (LD) group treated with a LD of sEVs preparations (3 × 109 particle/rat), and MetS High Dose (HD) group treated with a HD of sEVs preparations (9 × 109 particles/rat). The Control Non-Disease (ND) group was given a standard rat diet and autoclaved tap water with normal saline as treatment. Treatments were given via intravenous injection every three weeks for twelve weeks. Rats were assessed every six weeks for physical measurements, FBG, lipid profiles, CRP, leptin, adiponectin, and BP. Necropsy evaluation was performed on the lungs, liver, spleen, and kidney. RESULTS Significant reductions in FBG, triglycerides, BP, and increased HDL levels were observed in the treated groups compared to the control group. However, significant abdominal circumference (AC) improvement was not observed in the treated groups. Non-significant associations were found between fasting CRP, leptin, and adiponectin levels with MetS rats after treatment. In addition, sEVs preparations improved inflammation and hemorrhage in the lung and mineralisation in the renal of the treated group. CONCLUSIONS Human fetal WJMSCs-derived sEVs preparations improve all the clusters of MetS in rats except AC and could be further explored as a treatment for MetS.
Collapse
Affiliation(s)
- Illayaraja Krishnan
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (M.T.M.L.); (M.H.N.); (J.X.L.)
| | - Magdalene Tan Mei Ling
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (M.T.M.L.); (M.H.N.); (J.X.L.)
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Universiti Malaya (UM), Kuala Lumpur 50603, Malaysia
| | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (M.T.M.L.); (M.H.N.); (J.X.L.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (M.T.M.L.); (M.H.N.); (J.X.L.)
| | - Mohd Rafizul Mohd Yusof
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur 56000, Malaysia;
| | - Thavachelvi Thangarajah
- Department of Obstetrics and Gynaecology, Hospital Angkatan Tentera (HAT) Tuanku Mizan, Wangsa Maju, Kuala Lumpur 53300, Malaysia;
| | - Zalina Mahmood
- Production and Blood Supply Management Division, National Blood Centre, Jalan Tun Razak, Kuala Lumpur 50400, Malaysia;
| | - Nurul Izzati Uda Zahli
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia (UPM), Serdang 43400, Malaysia;
| | - Shathiya Rajamanickam
- Medixcell Sdn. Bhd., Level 5, Equatorial Plaza, Lot 5-5 & 5-6, Jalan Sultan Ismail, Kuala Lumpur 50250, Malaysia; (S.R.); (B.S.)
| | - Baskar Subramani
- Medixcell Sdn. Bhd., Level 5, Equatorial Plaza, Lot 5-5 & 5-6, Jalan Sultan Ismail, Kuala Lumpur 50250, Malaysia; (S.R.); (B.S.)
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (M.T.M.L.); (M.H.N.); (J.X.L.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
15
|
Sharma T, Ranawat P, Garg A, Rastogi P, Kaushal N. Short-chain fatty acids as a novel intervention for high-fat diet-induced metabolic syndrome. Mol Cell Biochem 2024:10.1007/s11010-024-05185-9. [PMID: 39709317 DOI: 10.1007/s11010-024-05185-9] [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: 10/09/2024] [Accepted: 12/07/2024] [Indexed: 12/23/2024]
Abstract
Metabolic syndrome (MetS) is driven by a complex interplay of genetic, lifestyle, and dietary factors, leading to weight gain, insulin resistance, dyslipidemia, and chronic inflammation. Gut microbiota dysbiosis has been recently recognized as a key contributor to MetS, leading to advancements in gut microbiome-based interventions to improve health outcomes. Considering the unique challenges associated with the use of pre/probiotics, short-chain fatty acids (SCFA), also known as postbiotics, have emerged as promising therapeutic agents due to their role in modulating host metabolism and physiology. Considering this, the aim of the current study was to explore the therapeutic potential of SCFA (butyrate, propionate, and acetate) supplementation against a high-fat diet (HFD)-induced experimental model of MetS in male Wistar rats. Alterations in body weight, lipid profile, histopathology, and adipose tissue accumulation were assessed to establish SCFA-mediated amelioration of experimental MetS. Further, the enzymatic (GPx, Catalase, GR, and GST) and non-enzymatic (LPO, total ROS, and Redox ratio were evaluated. The results indicated that SCFA supplementation could effectively mitigate key features of MetS. A significant reduction in body weight gain and fasting blood glucose levels, along with markedly lowered triglycerides, total cholesterol, and LDL levels, with partial restoration of HDL levels was observed following SCFA supplementation. SCFA administration also attenuated MetS-associated hepatic damage as studied by histopathological investigation and analysis of liver function marker enzyme activities. Such ameliorative effects of SCFA against HFD-induced MetS were owed to potential redox modulation studied using enzymatic and non-enzymatic oxidative stress markers. In conclusion, the study's outcomes show that SCFA supplementation could potentially be used against managing MetS. It underscores the therapeutic potential of SCFA by placing them as a novel gut microbiome-based dietary approach to improve metabolic health and reduce the risk of MetS-associated complications. However, more detailed mechanistic explorations are warranted in the future, leading to their beneficial role in MetS contributing to holistic health outcomes.
Collapse
Affiliation(s)
- Tanvi Sharma
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Pavitra Ranawat
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Ayushi Garg
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Pulkit Rastogi
- Department of Hematology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Naveen Kaushal
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
16
|
Shukla A, Meena K, Gupta A, Sandhir R. 1H NMR-Based Metabolomic Signatures in Rodent Models of Sporadic Alzheimer's Disease and Metabolic Disorders. ACS Chem Neurosci 2024; 15:4478-4499. [PMID: 39629865 DOI: 10.1021/acschemneuro.4c00510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological disorder that impacts the elderly population all over the globe. Evidence suggests association between AD and metabolic disorders such as diabetes mellitus (DM) and obesity (OB). The present study is an attempt to evaluate metabolic alterations in the serum and brain through NMR spectroscopy with the aim to identify shared metabolic signatures. AD was induced in rats by stereotactic intracerebroventricular injection of oligomerized Aβ-42 peptide into the brain. DM and OB were induced by intraperitoneal injection of streptozotocin and feeding rats on a high-fat diet, respectively. The metabolic alterations obtained through 1H NMR spectroscopy were further subjected to multivariate analysis by principal component analysis and partial least-squares discrimination for identification of metabolic signatures. In the serum, the levels of lactate and betaine were increased in AD, DM, and OB rats. On the other hand, the metabolite profile of brain indicated increase in the levels of lactate, N-acetylaspartate, and creatinine in AD, DM, and OB rats. Additionally, the concentration of neurochemicals such as glutamate, GABA, N-acetylglutamate, and myo-inositol were also elevated. The alterations in neurotransmitters and cerebral energy metabolism were accompanied by deficits in cognition assessed by Morris water maze in AD, DM, and OB rats. The perturbed metabolic profiles were accompanied by the presence of pathogenic amyloid deposits visualized by Congo red stain in the brains of AD, DM, and OB rats. Overall, the study identifies common metabolic signatures in AD, DM, and OB that may be involved in etiopathogenesis and also suggests linkages between these three conditions.
Collapse
Affiliation(s)
- Ananya Shukla
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| | - Khushbhu Meena
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Ashish Gupta
- Centre of Bio-Medical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow, Uttar Pradesh 226014, India
| | - Rajat Sandhir
- Department of Biochemistry, Hargobind Khorana Block (BMS Block II), Panjab University, Sector-25, Chandigarh 160014, India
| |
Collapse
|
17
|
Thunuguntla VBSC, Gadanec LK, McGrath C, Griggs JL, Sinnayah P, Apostolopoulos V, Zulli A, Mathai ML. Caralluma fimbriata Extract Improves Vascular Dysfunction in Obese Mice Fed a High-Fat Diet. Nutrients 2024; 16:4296. [PMID: 39770917 PMCID: PMC11678847 DOI: 10.3390/nu16244296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Obesity is a risk factor for developing cardiovascular diseases (CVDs) by impairing normal vascular function. Natural products are gaining momentum in the clinical setting due to their high efficacy and low toxicity. Caralluma fimbriata extract (CFE) has been shown to control appetite and promote weight loss; however, its effect on vascular function remains poorly understood. This study aimed to determine the effect that CFE had on weight loss and vascular function in mice fed a high-fat diet (HFD) to induce obesity, comparing this effect to that of lorcaserin (LOR) (an anti-obesity pharmaceutical) treatment. METHODS C57BL/6J male mice (n = 80) were fed a 16-week HFD to induce obesity prior to being treated with CFE and LOR as standalone treatments or in conjunction. Body composition data, such as weight gain and fat mass content were measured, isometric tension analyses were performed on isolated abdominal aortic rings to determine relaxation responses to acetylcholine, and immunohistochemistry studies were utilized to determine the expression profiles on endothelial nitric oxide synthase (eNOS) and cell stress markers (nitrotyrosine (NT) and 78 kDa glucose-regulated protein (GRP78)) in the endothelial, medial and adventitial layers of aortic rings. RESULTS The results demonstrated that CFE and CFE + LOR treatments significantly reduced weight gain (17%; 24%) and fat mass deposition (14%; 16%). A HFD markedly reduced acetylcholine-mediated relaxation (p < 0.05, p < 0.0001) and eNOS expression (p < 0.0001, p < 0.01) and significantly increased NT (p < 0.05, p < 0.0001) and GRP78 (p < 0.05, p < 0.01, p < 0.001). Obese mice treated with CFE exhibited significantly improved ACh-induced relaxation responses, increased eNOS (p < 0.05, p < 0.01) and reduced NT (p < 0.01) and GRP78 (p < 0.05, p < 0.01) expression. CONCLUSIONS Thus, CFE alone or in combination with LOR could serve as an alternative strategy for preventing obesity-related cardiovascular diseases.
Collapse
Affiliation(s)
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.B.S.C.T.); (L.K.G.); (C.M.); (J.L.G.); (P.S.); (V.A.)
| | - Catherine McGrath
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.B.S.C.T.); (L.K.G.); (C.M.); (J.L.G.); (P.S.); (V.A.)
| | - Joanne Louise Griggs
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.B.S.C.T.); (L.K.G.); (C.M.); (J.L.G.); (P.S.); (V.A.)
| | - Puspha Sinnayah
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.B.S.C.T.); (L.K.G.); (C.M.); (J.L.G.); (P.S.); (V.A.)
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.B.S.C.T.); (L.K.G.); (C.M.); (J.L.G.); (P.S.); (V.A.)
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.B.S.C.T.); (L.K.G.); (C.M.); (J.L.G.); (P.S.); (V.A.)
| | - Michael L. Mathai
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (V.B.S.C.T.); (L.K.G.); (C.M.); (J.L.G.); (P.S.); (V.A.)
| |
Collapse
|
18
|
Talib N, Mohamad NE, Yeap SK, Ho CL, Masarudin MJ, Abd-Aziz S, Izham MNM, Kumar MR, Hussin Y, Alitheen NB. Anti-Diabetic Effect of Lactobacillus Paracasei Isolated from Malaysian Water Kefir Grains. Probiotics Antimicrob Proteins 2024; 16:2161-2180. [PMID: 37755545 DOI: 10.1007/s12602-023-10159-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2023] [Indexed: 09/28/2023]
Abstract
The prevalence of type 2 diabetes mellitus (T2DM) is alarming because it is always linked to the increase in chronic diseases, mortality, and socioeconomic burden. Water kefir has a wide range of functional and probiotic characteristics attributed to the microorganisms present in the kefir grains. The present study aims to evaluate the in vivo anti-diabetic potential of the isolated Lactobacillus paracasei from Malaysian water kefir grains (MWKG) which was reported to have excellent probiotic properties and high antioxidant activities as reported previously. High-fat diet/streptozotocin (HFD/STZ) induction was used to obtain a T2DM model followed by treatment with the isolated L. paracasei from MWKG. The levels of glucose, insulin, and in vivo liver antioxidants were quantified after 14 weeks. Gene expression analysis of the liver was also carried out using microarray analysis, and several genes were selected for validation using quantitative real-time PCR. Insulin tolerance test demonstrated that the L. paracasei isolated from the MWKG alleviated T2DM by improving the area under the curve of the insulin tolerance test whereby low-dose and high-dose concentrations treated groups showed 2424.50 ± 437.02 mmol/L·min and 2017.50 ± 347.09 mmol/L·min, respectively, compared to untreated diabetic mice which was 3884.50 ± 39.36 mmol/L·min. Additionally, treatment with the isolated L. paracasei from MWKG regulated the expression of several genes related to glucose homeostasis and lipid metabolism in diabetic mice. These results suggested that the isolated L. paracasei from MWKG could be a potential dietary supplement for T2DM.
Collapse
Affiliation(s)
- Noorshafadzilah Talib
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Nurul Elyani Mohamad
- Biotechnology Research Institute, Universiti Malaysia Sabah, 88400, Kota Kinabalu, Sabah, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900, Sepang, Malaysia
| | - Chai Ling Ho
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Mas Jaffri Masarudin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Suraini Abd-Aziz
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Mira Nadiah Mohd Izham
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Muganti Rajah Kumar
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Yazmin Hussin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia.
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
19
|
Alasmari AA, Alhussain MH, Al-Khalifah AS, Alshiban NM, Alharthi R, Alyami NM, Alodah HS, Alahmed MF, Aljahdali BA, BaHammam AS. Ramadan fasting model modulates biomarkers of longevity and metabolism in male obese and non-obese rats. Sci Rep 2024; 14:28731. [PMID: 39567585 PMCID: PMC11579461 DOI: 10.1038/s41598-024-79557-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: 03/27/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
The health advantages of Ramadan fasting, a time-restricted eating from dawn to dusk, have garnered attention. Nevertheless, prior observational studies have found inconsistent findings because of challenges regulating variables such as sleep patterns, dietary habits, and physical activity. This study sought to investigate the impact of the Ramadan fasting model (RFM) on longevity and metabolic biomarkers in obese and non-obese rats. For 12 weeks, 48 male Wistar albino rats were separated into two groups and fed either a standard or a high-fat diet (HFD). During the final four weeks, rats in each group were separated into four subgroups to investigate the effect of RFM with/without training (on Treadmill) or glucose administration on the biomarkers of interest. The HFD groups subjected to RFM had significantly lower Insulin-like growth factor 1 (IGF-1) and mechanistic target of rapamycin (mTOR) serum, whereas AMPK, anti-inflammatory, and antioxidative stress serum levels were significantly higher. All groups reported decreased serum levels of Interleukin-6 (IL-6) and Tumor Necrosis Factor-alpha (TNF-α) compared to the HFD control group. Furthermore, the Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR) results indicated a significant elevation in the TP53 gene expression in groups subjected to RFM. The data indicate that RFM can improve longevity and metabolic biomarkers and reduce pro-inflammation and oxidative stress. Also, RFM improves anti-inflammatory and antioxidant markers in HFD-induced obese rats.
Collapse
Affiliation(s)
- Abeer Abdallah Alasmari
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Maha H Alhussain
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Abdulrahman Saleh Al-Khalifah
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Noura Mohammed Alshiban
- Experimental Surgery and Animal Lab, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rawan Alharthi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nouf M Alyami
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hesham S Alodah
- Experimental Surgery and Animal Lab, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed F Alahmed
- Experimental Surgery and Animal Lab, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Bayan A Aljahdali
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed S BaHammam
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Steg A, Oczkowicz M, Świątkiewicz M. Effects of High-Dose Vitamin D3 Supplementation on Pig Performance, Vitamin D Content in Meat, and Muscle Transcriptome in Pigs. J Anim Physiol Anim Nutr (Berl) 2024. [PMID: 39567837 DOI: 10.1111/jpn.14066] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/19/2024] [Indexed: 11/22/2024]
Abstract
Vitamin D is known for its role in calcium homeostasis, bone health, and immune function. Recent research has explored its effects on muscle functionality and meat quality in pigs. This study examined high-dose vitamin D3 supplementation in pigs, focusing on growth, blood and tissue vitamin D3 levels, and muscle transcriptome changes. Thirty pigs were divided into three groups, given different amounts of oral supplementation: control, 5000 IU/kg and 10,000 IU/kg vitamin D3. Biochemical and haematological blood parameters, vitamin D content in blood and muscle, and kidney calcium content were evaluated. RNA-seq and qPCR analysed muscle transcriptome changes, while gene set enrichment analysis (GSEA) identified gene expression enrichments. Results showed that 5000 IU/kg vitamin D3 supplementation altered blood parameters like platelet anisocytosis and glucose levels but did not affect body weight, weight gain, or feed intake. Kidney calcium content increased with supplementation. The muscle (longissimus dorsi) vitamin D content increased, suggesting the potential for biofortified pork, although still not optimal as a dietary vitamin D source. Transcriptome analysis revealed minimal gene expression changes, with only the interferon-gamma receptor 2 (IFNGR2) gene differentially expressed at the highest dose. GSEA indicated enrichment in ATP metabolic processes and electron transport chain genes in the 5000 IU/kg group, and immune system, cholesterol, steroid, and fatty acid metabolism genes in the 10,000 IU/kg group. Despite literature suggesting a role for vitamin D in muscle gene expression and growth improvement, this study found its effects limited.
Collapse
Affiliation(s)
- Anna Steg
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska, Poland
| | - Maria Oczkowicz
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska, Poland
| | - Małgorzata Świątkiewicz
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, Krakowska, Poland
| |
Collapse
|
21
|
Yunin MA, Boychenko SS, Lebedev P, Deykin AV, Pokrovskii MV, Egorov AD. Gene Therapy Approach for Treatment of Obese Agouti Mice. Int J Mol Sci 2024; 25:12144. [PMID: 39596212 PMCID: PMC11594101 DOI: 10.3390/ijms252212144] [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: 09/27/2024] [Revised: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a significant metabolic disorder associated with excessive fat accumulation and insulin resistance. In this study, we explored a gene therapy approach to treat obesity in agouti mice using adeno-associated viruses (AAVs) carrying PRDM16, FoxP4, or Follistatin (FST) genes, which are known to promote the browning of white adipose tissue. Mice treated with AAVs encoding PRDM16, FoxP4, or FST genes showed a reduction in body weight (10-14%) within the first three weeks after administration, compared to the control groups. A lipidomic analysis of the adipose tissue revealed a dramatic reduction in triacylglycerol (TAG) species with low carbon numbers (40-54 acyl carbons) in treated mice.
Collapse
Affiliation(s)
- Maxim A. Yunin
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sirius, Russia; (M.A.Y.); (S.S.B.)
| | - Stanislav S. Boychenko
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sirius, Russia; (M.A.Y.); (S.S.B.)
| | - Petr Lebedev
- Center for Preclinical and Clinical Research, Belgorod State National Research University, 85 Pobedy St., 308015 Belgorod, Russia; (P.L.); (A.V.D.); (M.V.P.)
| | - Alexey V. Deykin
- Center for Preclinical and Clinical Research, Belgorod State National Research University, 85 Pobedy St., 308015 Belgorod, Russia; (P.L.); (A.V.D.); (M.V.P.)
| | - Mikhail V. Pokrovskii
- Center for Preclinical and Clinical Research, Belgorod State National Research University, 85 Pobedy St., 308015 Belgorod, Russia; (P.L.); (A.V.D.); (M.V.P.)
| | - Alexander D. Egorov
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave. 1, 354340 Sirius, Russia; (M.A.Y.); (S.S.B.)
| |
Collapse
|
22
|
Chen B, Yuan C, Guo T, Liu J, Yang B, Lu Z. The molecular regulated mechanism of METTL3 and FTO in lipid metabolism of Hu sheep. Genomics 2024; 116:110945. [PMID: 39341298 DOI: 10.1016/j.ygeno.2024.110945] [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: 06/20/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Balanced lipid metabolism can improve the growth performance and meat quality of livestock. The m6A methylation-related genes METTL3 and FTO play important roles in animal lipid metabolism; however, the mechanism through which they regulate lipid metabolism in sheep is unclear. RESULTS We established lipid deposition models of hepatocytes and preadipocytes in Hu sheep. In the hepatocyte lipid deposition model, the genes expression levels of FABP4, Accα, ATGL and METTL3, METTL14, and FTO-were significantly up-regulated after lipid deposition (P < 0.05). Transcriptomic and metabolomic analyses showed that lipid deposition had a significant effect on MAPK, steroid biosynthesis, and glycerophospholipid metabolism pathway in hepatocytes. The m6A methylation level decreased but the difference was not significant after METTL3 interference, and the expression levels of FABP4 and ATGL increased significantly (P < 0.05); the m6A methylation level significantly increased following METTL3 overexpression, and LPL and ATGL expression levels significantly decreased (P < 0.05), indicating that overexpression of METTL3 inhibited the expression of lipid deposition-related genes in a m6A-dependent manner. The m6A methylation level was significantly increased, ATGL expression was significantly decreased (P < 0.05), and LPL, FABP4, and Accα expression was not significantly changed following FTO interference (P > 0.05); the m6A methylation level was significantly decreased after FTO overexpression, and LPL, FABP4, and ATGL expression was significantly increased (P < 0.05), indicating that FTO overexpression increased the expression of lipid deposition-related genes in a m6A-dependent manner. Transcriptomic and metabolomic analyses showed that m6A methylation modification mainly regulated lipid metabolism through triglyceride metabolism, adipocytokine signaling, MAPK signaling, and fat digestion and absorption in hepatocytes. In the lipid deposition model of preadipocytes, the regulation of gene expression is the same as that in hepatocytes. CONCLUSIONS METTL3 significantly inhibited the expression of lipid deposition-related genes, whereas FTO overexpression promoted lipid deposition. Our study provides a theoretical basis and reference for accurately regulating animal lipid deposition by mastering METTL3 and FTO genes to promote high-quality animal husbandry.
Collapse
Affiliation(s)
- Bowen Chen
- Key Laboratory of Animal Genetics and Breeding on the Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Chao Yuan
- Key Laboratory of Animal Genetics and Breeding on the Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Tingting Guo
- Key Laboratory of Animal Genetics and Breeding on the Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jianbin Liu
- Key Laboratory of Animal Genetics and Breeding on the Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Bohui Yang
- Key Laboratory of Animal Genetics and Breeding on the Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
| | - Zengkui Lu
- Key Laboratory of Animal Genetics and Breeding on the Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; Sheep Breeding Engineering Technology Research Center of Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
| |
Collapse
|
23
|
Al-Onaizi M, Braysh K, Alkafeef SS, Altarrah D, Dannoon S, Alasousi D, Adel H, Al-Ajmi M, Kandari A, Najem R, Nizam R, Williams MR, John S, Thanaraj TA, Ahmad R, Al-Hussaini H, Al-Mulla F, Alzaid F. Glucose intolerance induces anxiety-like behaviors independent of obesity and insulin resistance in a novel model of nutritional metabolic stress. Nutr Neurosci 2024; 27:1143-1161. [PMID: 38319634 DOI: 10.1080/1028415x.2024.2310419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
OBJECTIVES Type 2 diabetes (T2D) is a metabolic disease of major public health concern. It impacts peripheral tissues and the central nervous system, leading to systemic dysmetabolism and neurocognitive impairments, including memory deficits, anxiety, and depression. The metabolic determinants of these neurocognitive impairments remain unidentified. Here, we sought to address this question by developing a proprietary (P-) high-fat diet (HFD), in which glucose intolerance precedes weight gain and insulin resistance. METHODS The P-HFD model was nutritionally characterized, and tested in vivo in mice that underwent behavioral and metabolic testing. The diet was benchmarked against reference models. . RESULTS P-HFD has 42% kcal from fat, high monounsaturated/polyunsaturated fatty acid ratio, and 10% (w/v) sucrose in drinking water. When administered, from the early stages of glucose intolerance alone, animals exhibit anxiety-like behavior, without depression nor recognition memory deficits. Long-term P-HFD feeding leads to weight gain, brain glucose hypometabolism as well as impaired recognition memory. Using an established genetic model of T2D (db/db) and of diet-induced obesity (60% kcal from fat) we show that additional insulin resistance and obesity are associated with depressive-like behaviors and recognition memory deficits. DISCUSSION Our findings demonstrate that glucose intolerance alone can elicit anxiety-like behavior. Through this study, we also provide a novel nutritional model (P-HFD) to characterize the discrete effects of glucose intolerance on cognition, behavior, and the physiology of metabolic disease.
Collapse
Affiliation(s)
- Mohammed Al-Onaizi
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Kawthar Braysh
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
| | - Selma S Alkafeef
- Faculty of Medicine, Department of Biochemistry, Kuwait University, Kuwait City, Kuwait
| | - Dana Altarrah
- Faculty of Public Health, Department of Social and Behavioral Science, Kuwait University, Kuwait City, Kuwait
| | - Shorouk Dannoon
- Faculty of Medicine, Department of Nuclear Medicine, Kuwait University, Kuwait City, Kuwait
| | - Dalal Alasousi
- Faculty of Science, Department of Biochemistry, Kuwait University, Kuwait City, Kuwait
| | - Hawraa Adel
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
| | - Mariam Al-Ajmi
- Faculty of Science, Department of Biochemistry, Kuwait University, Kuwait City, Kuwait
| | - Anwar Kandari
- Dasman Diabetes Institute, Kuwait City, Kuwait
- Ministry of Health, Kuwait City, Kuwait
| | - Rawan Najem
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | | | - Sumi John
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | | | - Heba Al-Hussaini
- Faculty of Medicine, Department of Anatomy, Kuwait University, Kuwait City, Kuwait
| | | | - Fawaz Alzaid
- Dasman Diabetes Institute, Kuwait City, Kuwait
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| |
Collapse
|
24
|
Chesters RA, Zhu J, Coull BM, Baidoe-Ansah D, Baumer L, Palm L, Klinghammer N, Chen S, Hahm A, Yagoub S, Cantacorps L, Bernardi D, Ritter K, Lippert RN. Fasting-induced activity changes in MC3R neurons of the paraventricular nucleus of the thalamus. Life Sci Alliance 2024; 7:e202402754. [PMID: 39107065 PMCID: PMC11303869 DOI: 10.26508/lsa.202402754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024] Open
Abstract
The brain controls energy homeostasis by regulating food intake through signaling within the melanocortin system. Whilst we understand the role of the hypothalamus within this system, how extra-hypothalamic brain regions are involved in controlling energy balance remains unclear. Here we show that the melanocortin 3 receptor (MC3R) is expressed in the paraventricular nucleus of the thalamus (PVT). We tested whether fasting would change the activity of MC3R neurons in this region by assessing the levels of c-Fos and pCREB as neuronal activity markers. We determined that overnight fasting causes a significant reduction in pCREB levels within PVT-MC3R neurons. We then questioned whether perturbation of MC3R signaling, during fasting, would result in altered refeeding. Using chemogenetic approaches, we show that modulation of MC3R activity, during the fasting period, does not impact body weight regain or total food intake in the refeeding period. However, we did observe significant differences in the pattern of feeding-related behavior. These findings suggest that the PVT is a region where MC3R neurons respond to energy deprivation and modulate refeeding behavior.
Collapse
Affiliation(s)
- Robert A Chesters
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Jiajie Zhu
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - Bethany M Coull
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - David Baidoe-Ansah
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - Lea Baumer
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Lydia Palm
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Niklas Klinghammer
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Seve Chen
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Anneke Hahm
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Selma Yagoub
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Lídia Cantacorps
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Daniel Bernardi
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Katrin Ritter
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Rachel N Lippert
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
25
|
Santos KCC, Domingos LF, Nunes FM, Simmer LM, Cordeiro ER, Filetti FM, Bocalini DS, Corrêa CR, Lima-Leopoldo AP, Leopoldo AS. Capsinoids Increase Antioxidative Enzyme Activity and Prevent Obesity-Induced Cardiac Injury without Positively Modulating Body Fat Accumulation and Cardiac Oxidative Biomarkers. Nutrients 2024; 16:3183. [PMID: 39339783 PMCID: PMC11434772 DOI: 10.3390/nu16183183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Capsinoids are potential antioxidant agents capable of reducing oxidative damage and the resulting complications triggered by obesity. Thus, this study aimed to investigate the effects of capsinoids on adiposity and biomarkers of cardiac oxidative stress in obese rats induced by a high-fat diet. METHODS Male Wistar rats were exposed to a high-fat diet for 27 consecutive weeks. After the characterization of obesity (week 19), some of the obese animals began to receive capsinoids (10 mg/kg/day) by orogastric gavage. Adiposity and comorbidities were assessed. In the heart, remodeling, injury, and biomarkers of oxidative stress were determined. RESULTS The treatment did not reduce obesity-induced adiposity but was efficient in reducing cholesterol levels. Capsinoid treatment did not cause a difference in heart and LV mass, despite having reduced troponin I concentrations. Furthermore, capsinoids did not reduce the increase in the advanced oxidation of protein products and carbonylated proteins caused by obesity in cardiac tissue. In addition, obese rats treated with capsinoids presented high levels of malondialdehyde and greater antioxidant enzyme activity compared to untreated obese rats. CONCLUSIONS In conclusion, treatment with capsinoids increases antioxidative enzyme activity and prevents obesity-induced cardiac injury without positively modulating body fat accumulation and cardiac oxidative biomarkers.
Collapse
Affiliation(s)
- Késsia Cristina Carvalho Santos
- Postgraduate Program in Nutrition and Health, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - Lucas Furtado Domingos
- Postgraduate Program in Nutrition and Health, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - Fabiane Merigueti Nunes
- Postgraduate Program in Nutrition and Health, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - Luisa Martins Simmer
- Postgraduate Program in Nutrition and Health, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - Evellyn Rodrigues Cordeiro
- Postgraduate Program in Physiological Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - Filipe Martinuzo Filetti
- Postgraduate Program in Physiological Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - Danilo Sales Bocalini
- Postgraduate Program in Physical Education, Physical Education and Sports Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - Camila Renata Corrêa
- Medical School, São Paulo State University (UNESP), Botucatu 18618-686, SP, Brazil
| | - Ana Paula Lima-Leopoldo
- Postgraduate Program in Nutrition and Health, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
- Postgraduate Program in Physical Education, Physical Education and Sports Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| | - André Soares Leopoldo
- Postgraduate Program in Nutrition and Health, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
- Postgraduate Program in Physiological Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória 29075-910, ES, Brazil
| |
Collapse
|
26
|
Guy AR, Klores M, Prestia K, Raymond M, Rasmussen S. Effect of Novel High-fat Diet Feeding Methods on Food Wastage, Weight Gain, Hair Coat Grease Accumulation, and Scratching Behavior in C57BL/6NCrl Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2024; 64:98-105. [PMID: 39242184 PMCID: PMC11808372 DOI: 10.30802/aalas-jaalas-24-059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/30/2024] [Accepted: 08/16/2024] [Indexed: 09/09/2024]
Abstract
Soft-pelleted, high-fat diets (HFD) are greasy and crumble easily leading to food wastage and hair coat grease accumulation when mice are fed using commercially available feeders. The ideal HFD feeder design should reduce food wastage, facilitate mouse weight gain, and minimize variables such as hair coat grease accumulation that have the potential to alter scratching behaviors. Our study compared the feeding efficiency of 2 commercially available feeders (feeders A and E) to 4 novel feeder designs (feeders B, C, D, and F). Novel feeders had alterations in feeding aperture size, feeding surface area, feeder configuration, and level of food presentation. Male C57BL/6NCrl mice (n = 120; 4/cage) were randomly assigned to cages containing one of the 6 feeder types and were fed HFD for 12 wk. Feeders and cage bottoms were weighed before use and then weekly at the time of cage change. Mice were weighed before starting the HFD and then biweekly. Scratching behavior was video recorded at 0, 4, 8, and 12 wk. Hair coat grease accumulation was visually scored biweekly. Feeder A use was associated with the highest feed cost due to HFD wastage ($36.98 ± 1.54/cage/wk). Mice fed using Feeder A had the highest average weight gain (23.75 ± 0.8 g, P < 0.005). However, mice also had significantly higher hair coat grease accumulation scores (P < 0.05) and significantly increased scratching frequency at 4 wk (P < 0.05) when compared with mice fed using other feeder types. Novel feeder designs utilized 10 to 21 times less HFD dispensed when compared to feeder A. Mice fed using novel feeders also displayed improved welfare, as evidenced by low hair coat grease accumulation scores, and no significant differences in scratching frequency when compared with baseline behavior.
Collapse
Affiliation(s)
- Alyson R Guy
- NYU-Regeneron Postdoctoral Training Program in Laboratory Animal Medicine, NYU Langone Health, New York, New York
| | - Molly Klores
- NYU-Regeneron Postdoctoral Training Program in Laboratory Animal Medicine, NYU Langone Health, New York, New York
| | - Kevin Prestia
- NYU-Regeneron Postdoctoral Training Program in Laboratory Animal Medicine, NYU Langone Health, New York, New York
| | - Mark Raymond
- NYU-Regeneron Postdoctoral Training Program in Laboratory Animal Medicine, NYU Langone Health, New York, New York
| | - Skye Rasmussen
- NYU-Regeneron Postdoctoral Training Program in Laboratory Animal Medicine, NYU Langone Health, New York, New York
| |
Collapse
|
27
|
Wan K, Jin Y, Fan R, Xu Q, Li X, Yan H, Wang R. Exploring molecular mechanisms of exercise on metabolic syndrome: a bibliometric and visualization study using CiteSpace. Front Endocrinol (Lausanne) 2024; 15:1408466. [PMID: 39290329 PMCID: PMC11405195 DOI: 10.3389/fendo.2024.1408466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Objective To investigate the molecular mechanisms through which exercise influences metabolic syndrome (MS) and identify key research trends and collaborative networks using bibliometric and visualization techniques. Methods We conducted a systematic literature search using the Web of Science Core Collection for articles published from 2014 to 2023. Using CiteSpace, we performed a bibliometric analysis of 562 eligible papers, generating visual knowledge maps to identify prevailing patterns, popular subjects, and emerging trends in the literature. Results The study reveals that exercise mitigates MS by reversing high-fat diet-induced abdominal obesity, reducing lipid accumulation and inflammation, enhancing insulin sensitivity, and improving cardiovascular function. Key molecular pathways include PPAR-γ/CPT-1/MCAD signaling, AMPK activation, and nitric oxide production. The USA leads in research output, with significant contributions from American institutions. Collaboration among researchers is limited, highlighting the need for more extensive and high-quality research initiatives. Conclusions Regular, moderate-to-high-intensity exercise is crucial for managing MS. Exercise activates beneficial molecular pathways, improving metabolic health and cardiovascular function. Future research should focus on expanding collaborations and exploring novel molecular targets to enhance the therapeutic potential of exercise in metabolic syndrome management.
Collapse
Affiliation(s)
- Kang Wan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Physical Education College, Henan Sport University, Zhengzhou, China
| | - Yue Jin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Ruobing Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Qizi Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiaoshi Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hongmei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Endocrinology and Metabolism, Wusong Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
28
|
Goodus MT, Alfredo AN, Carson KE, Dey P, Pukos N, Schwab JM, Popovich PG, Gao J, Mo X, Bruno RS, McTigue DM. Spinal cord injury-induced metabolic impairment and steatohepatitis develops in non-obese rats and is exacerbated by premorbid obesity. Exp Neurol 2024; 379:114847. [PMID: 38852834 DOI: 10.1016/j.expneurol.2024.114847] [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: 02/03/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Impaired sensorimotor functions are prominent complications of spinal cord injury (SCI). A clinically important but less obvious consequence is development of metabolic syndrome (MetS), including increased adiposity, hyperglycemia/insulin resistance, and hyperlipidemia. MetS predisposes SCI individuals to earlier and more severe diabetes and cardiovascular disease compared to the general population, which trigger life-threatening complications (e.g., stroke, myocardial infarcts). Although each comorbidity is known to be a risk factor for diabetes and other health problems in obese individuals, their relative contribution or perceived importance in propagating systemic pathology after SCI has received less attention. This could be explained by an incomplete understanding of MetS promoted by SCI compared with that from the canonical trigger diet-induced obesity (DIO). Thus, here we compared metabolic-related outcomes after SCI in lean rats to those of uninjured rats with DIO. Surprisingly, SCI-induced MetS features were equal to or greater than those in obese uninjured rats, including insulin resistance, endotoxemia, hyperlipidemia, liver inflammation and steatosis. Considering the endemic nature of obesity, we also evaluated the effect of premorbid obesity in rats receiving SCI; the combination of DIO + SCI exacerbated MetS and liver pathology compared to either alone, suggesting that obese individuals that sustain a SCI are especially vulnerable to metabolic dysfunction. Notably, premorbid obesity also exacerbated intraspinal lesion pathology and worsened locomotor recovery after SCI. Overall, these results highlight that normal metabolic function requires intact spinal circuitry and that SCI is not just a sensory-motor disorder, but also has significant metabolic consequences.
Collapse
Affiliation(s)
- Matthew T Goodus
- The Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anthony N Alfredo
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Kaitlin E Carson
- The Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India
| | - Nicole Pukos
- The Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Jan M Schwab
- The Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Phillip G Popovich
- The Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jie Gao
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaokui Mo
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Richard S Bruno
- Human Nutrition Program, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Dana M McTigue
- The Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
29
|
Kleeschulte S, Fischinger V, Öhlke L, Bode J, Kamler M, Dobrev D, Grandoch M, Fender AC. The thrombin receptor PAR4 supports visceral adipose tissue inflammation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7187-7200. [PMID: 38652276 PMCID: PMC11422268 DOI: 10.1007/s00210-024-03097-5] [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: 03/20/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Thrombin inhibition suppresses adiposity, WAT inflammation and metabolic dysfunction in mice. Protease-activated receptor (PAR)1 does not account for thrombin-driven obesity, so we explored the culprit role of PAR4 in this context. Male WT and PAR-4-/- mice received a high fat diet (HFD) for 8 weeks, WT controls received standard chow. Body fat was quantified by NMR. Epididymal WAT was assessed by histology, immunohistochemistry, qPCR and lipase activity assay. 3T3-L1 preadipocytes were differentiated ± thrombin, acutely stimulated ± PAR4 activating peptide (AP) and assessed by immunoblot, qPCR and U937 monocyte adhesion. Epicardial adipose tissue (EAT) from obese and lean patients was assessed by immunoblot. PAR4 was upregulated in mouse WAT under HFD. PAR4-/- mice developed less visceral adiposity and glucose intolerance under HFD, featuring smaller adipocytes, fewer macrophages and lower expression of adipogenic (leptin, PPARγ) and pro-inflammatory genes (CCL2, IL-1β) in WAT. HFD-modified activity and expression of lipases or perilipin were unaffected by PAR4 deletion. 3T3-L1 adipocytes differentiated with thrombin retained Ki67 expression, further upregulated IL-1β and CCL2 and were more adhesive for monocytes. In mature adipocytes, PAR4-AP increased phosphorylated ERK1/2 and AKT, upregulated Ki67, CCl2, IL-β and hyaluronan synthase 1 but not TNF-α mRNA, and augmented hyaluronidase-sensitive monocyte adhesion. Obese human EAT expressed more PAR4, CD68 and CD54 than lean EAT. PAR4 upregulated in obesity supports adipocyte hypertrophy, WAT expansion and thrombo-inflammation. The emerging PAR4 antagonists provide a therapeutic perspective in this context beyond their canonical antiplatelet action.
Collapse
Affiliation(s)
- Sonja Kleeschulte
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Vivien Fischinger
- Institute for Pharmacology and Clinical Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Lisa Öhlke
- Institute for Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
| | - Johannes Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, University Hospital Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute for Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Maria Grandoch
- Institute for Translational Pharmacology and CARID Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Anke C Fender
- Institute for Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany.
| |
Collapse
|
30
|
Ferreira NB, Dias CT, Chaaban AFA, Beserra-Filho JIA, Ribeiro AM, Lambertucci RH, Mendes-da-Silva C. Improving dietary patterns in obese mice: Effects on body weight, adiposity, anhedonia-like behavior, pro-BDNF expression and 5-HT system. Brain Res 2024; 1838:148996. [PMID: 38744387 DOI: 10.1016/j.brainres.2024.148996] [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: 01/26/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION The excessive fat accumulation in obesity, resulting from an unbalanced diet, can lead to metabolic and neurological disorders and increase the risk of developing anxiety and depression. AIM Assess the impact of dietary intervention (DI) on the serotonergic system, brain-derived neurotrophic factor (BDNF) expression and behaviors of obese mice. METHODS Male C57BL/6 mice, 5 weeks old, received a high-fat diet (HFD) for 10 weeks for the induction of obesity. After this period, for 8 weeks, half of these animals received a control diet (CD), group obese (OB) + control diet (OB + CD, n = 10), and another half continued being fed HFD, group obese + HFD (OB + HFD, n = 10). At the end of the eighth week of intervention, behavioral tests were performed (sucrose preference test, open field, novel object recognition, elevated plus maze and tail suspension). Body weight and food intake were assessed weekly. Visceral adiposity, the hippocampal and hypothalamic protein expression of BDNF, 5-HT1A (5-HT1A serotonin receptor) and TPH2 (key enzyme in serotonin synthesis), were evaluated after euthanasia. RESULTS The dietary intervention involved changing from a HFD to a CD over an 8-week period, effectively reduced body weight gain, adiposity, and anhedonia-like behavior. In the OB + HFD group, we saw a lower sucrose preference and shorter traveled distance in the open field, along with increased pro-BDNF expression in the hypothalamus compared to the OB + CD mice. However, the levels of TPH2 and 5-HT1A remained unchanged. CONCLUSION The HFD model induced both obesity and anhedonia, but the dietary intervention successfully improved these conditions.
Collapse
Affiliation(s)
- Nicoly Bédia Ferreira
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Clarissa Tavares Dias
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Ana Flávia Alves Chaaban
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - José Ivo Araújo Beserra-Filho
- Laboratory of Neuroscience and Bioprospecting of Natural Products, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Alessandra Mussi Ribeiro
- Laboratory of Neuroscience and Bioprospecting of Natural Products, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Rafael Herling Lambertucci
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil
| | - Cristiano Mendes-da-Silva
- Laboratory of Neuroscience and Nutrition, Department of Biosciences, Federal University of Sao Paulo/UNIFESP, Santos, SP, Brazil.
| |
Collapse
|
31
|
Saengmearnuparp T, Pintana H, Apaijai N, Chunchai T, Thonusin C, Kongkaew A, Lojanapiwat B, Chattipakorn N, Chattipakorn SC. Long-term Treatment with a 5-Alpha-Reductase Inhibitor Alleviates Depression-like Behavior in Obese Male Rats. Behav Brain Res 2024; 472:115155. [PMID: 39032869 DOI: 10.1016/j.bbr.2024.115155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Several studies have reported side effects of finasteride (FIN), such as anxiety/depression in young men. Obesity is also positively associated with anxiety/depression symptoms; however, the impacts of long-term FIN treatment and FIN withdrawal in young obese individuals are still elusive. The present study aimed to investigate the effect of long-term treatment and its withdrawal on anxiety/depression and brain pathologies in lean and obese adult male rats. Forty-eight male Wistar rats were equally divided into two groups and fed either a normal or high-fat diet. At age 13 weeks, rats in each dietary group were divided into three subgroups: 1) the control group receiving drinking water, 2) the long-term treatment group receiving FIN orally at 5 mg/kg/day for 6 weeks, and 3) the withdrawal group receiving FIN orally at 5 mg/kg/day for 2 weeks followed by a 4-week withdrawal period. Anxiety/depression-like behaviors, biochemical analysis, brain inflammation, oxidative stress, neuroactive steroids, brain metabolites, and microglial complexity were tested. The result showed that lean rats treated with long-term FIN and its withdrawal exhibited metabolic disturbances, depressive-like behavior, and both groups showed increased neurotoxic metabolites and reduced microglial complexity. Obesity itself led to metabolic disturbances and brain pathologies, including increased inflammation, oxidative stress, and quinolinic acid, as well as reduced microglial complexity, resulting in increased anxiety- and depression-like behaviors. Interestingly, the long-term FIN treatment group in obese rats showed attenuation of depressive-like behaviors, brain inflammation, and oxidative stress, along with increased brain antioxidants, suggesting the possible benefits of FIN in obese conditions.
Collapse
Affiliation(s)
- Thiraphat Saengmearnuparp
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Urology division, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Hiranya Pintana
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Titikorn Chunchai
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanisa Thonusin
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Bannakij Lojanapiwat
- Urology division, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
32
|
Barr B, Gollahon L. The Modification of Dietary Protein with Ammonium Hydroxide Enhancement Improves Longevity and Metabolic Outcomes in a Sex-Dependent Manner. Nutrients 2024; 16:2787. [PMID: 39203925 PMCID: PMC11357104 DOI: 10.3390/nu16162787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
(1) Background: Dietary protein is a key component of all dietary patterns. It has been demonstrated that there are subtle differences in health implications associated with the source of dietary protein consumed. This study examined dietary protein sources (DPSs) in a long-term study of diet-induced obesity ± ammonium hydroxide enhancement (AHE) and its role in improving long-term health outcomes. (2) Methods: Over 18 months, 272 C3H/HeJ mice (136 male and 136 female) were monitored on high-fat diets with varying DPSs ± AHE. Mice were monitored for weekly change in total mass, as well as 6-month assessments of lean and fat mass. At each assessment, a cohort (~8 mice per diet per sex) was censored for a cross-sectional examination of organ function. (3) Results: Longevity was improved in females fed AHE diets, regardless of DPSs. Females' measures of fat and lean mass were markedly elevated with casein protein diets compared to beef protein diets regardless of AHE. Females fed a beef protein diet + AHE demonstrated reduced fat mass and increased lean mass with aging. In males, AHE beef protein diet-fed mice showed marked improvement to longevity and increased lean mass at 6 months. (4) Conclusions: This study demonstrates that dietary protein modification by AHE attenuates the negative impacts of HF diets in both males and females in a sex-dependent manner. Furthermore, the results from this study emphasize the importance of identifying the differences in the utilization of dietary proteins in both a sex- and age-related manner and demonstrate the potential of DPS modification by AHE as a dietary intervention.
Collapse
Affiliation(s)
- Benjamin Barr
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| |
Collapse
|
33
|
Zheng Y, Chen J, Macwan V, Dixon CL, Li X, Liu S, Yu Y, Xu P, Sun Q, Hu Q, Liu W, Raught B, Fairn GD, Neculai D. S-acylation of ATGL is required for lipid droplet homoeostasis in hepatocytes. Nat Metab 2024; 6:1549-1565. [PMID: 39143266 DOI: 10.1038/s42255-024-01085-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/21/2024] [Indexed: 08/16/2024]
Abstract
Lipid droplets (LDs) are organelles specialized in the storage of neutral lipids, cholesterol esters and triglycerides, thereby protecting cells from the toxicity of excess lipids while allowing for the mobilization of lipids in times of nutrient deprivation. Defects in LD function are associated with many diseases. S-acylation mediated by zDHHC acyltransferases modifies thousands of proteins, yet the physiological impact of this post-translational modification on individual proteins is poorly understood. Here, we show that zDHHC11 regulates LD catabolism by modifying adipose triacylglyceride lipase (ATGL), the rate-limiting enzyme of lipolysis, both in hepatocyte cultures and in mice. zDHHC11 S-acylates ATGL at cysteine 15. Preventing the S-acylation of ATGL renders it catalytically inactive despite proper localization. Overexpression of zDHHC11 reduces LD size, whereas its elimination enlarges LDs. Mutating ATGL cysteine 15 phenocopies zDHHC11 loss, causing LD accumulation, defective lipolysis and lipophagy. Our results reveal S-acylation as a mode of regulation of ATGL function and LD homoeostasis. Modulating this pathway may offer therapeutic potential for treating diseases linked to defective lipolysis, such as fatty liver disease.
Collapse
Affiliation(s)
- Yuping Zheng
- Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jishun Chen
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Vinitha Macwan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Charneal L Dixon
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Xinran Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Shengjie Liu
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yuyun Yu
- Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Pinglong Xu
- Life Science Institute, Zhejiang University, Hangzhou, China
| | - Qiming Sun
- Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qi Hu
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
| | - Wei Liu
- Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
| | - Gregory D Fairn
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University Health Network, Toronto, Ontario, Canada.
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Dante Neculai
- Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
| |
Collapse
|
34
|
Sridhar A, Khan D, Babu G, Irwin N, Gault VA, Flatt PR, Moffett CR. Chronic exposure to incretin metabolites GLP-1(9-36) and GIP(3-42) affect islet morphology and beta cell health in high fat fed mice. Peptides 2024; 178:171254. [PMID: 38815655 DOI: 10.1016/j.peptides.2024.171254] [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: 02/19/2024] [Revised: 04/24/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), are rapidly degraded by dipeptidyl peptidase-4 (DPP-4) to their major circulating metabolites GLP-1(9-36) and GIP(3-42). This study investigates the possible effects of these metabolites, and the equivalent exendin molecule Ex(9-39), on pancreatic islet morphology and constituent alpha and beta cells in high-fat diet (HFD) fed mice. Male Swiss TO-mice (6-8 weeks-old) were maintained on a HFD or normal diet (ND) for 4 months and then received twice-daily subcutaneous injections of GLP-1(9-36), GIP(3-42), Ex(9-39) (25 nmol/kg bw) or saline vehicle (0.9% (w/v) NaCl) over a 60-day period. Metabolic parameters were monitored and excised pancreatic tissues were used for immunohistochemical analysis. Body weight and assessed metabolic indices were not changed by peptide administration. GLP-1(9-36) significantly (p<0.001) increased islet density per mm2 tissue, that was decreased (p<0.05) by HFD. Islet, beta and alpha cell areas were increased (p<0.01) following HFD and subsequently reduced (p<0.01-p<0.001) by GIP(3-42) and Ex(9-39) treatment. While GLP-1(9-36) did not affect islet and beta cell areas in HFD mice, it significantly (p<0.01) decreased alpha cell area. Compared to ND and HFD mice, GIP(3-42) treatment significantly (p<0.05) increased beta cell proliferation. Whilst HFD increased (p<0.001) beta cell apoptosis, this was reduced (p<0.01-p<0.001) by both GLP-1(9-36) and GIP(3-42). These data indicate that the major circulating forms of GLP-1 and GIP, namely GLP-1(9-36) and GIP(3-42) previously considered largely inactive, may directly impact pancreatic morphology, with an important protective effect on beta cell health under conditions of beta cell stress.
Collapse
Affiliation(s)
- Ananyaa Sridhar
- Biomedical Sciences Research Institute, Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| | - Dawood Khan
- Biomedical Sciences Research Institute, Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Gayathri Babu
- Biomedical Sciences Research Institute, Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- Biomedical Sciences Research Institute, Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Victor A Gault
- Biomedical Sciences Research Institute, Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Charlotte R Moffett
- Biomedical Sciences Research Institute, Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
35
|
Al-Harbi LN. Morin Prevents Non-Alcoholic Hepatic Steatosis in Obese Rats by Targeting the Peroxisome Proliferator-Activated Receptor Alpha (PPARα). Life (Basel) 2024; 14:945. [PMID: 39202687 PMCID: PMC11355712 DOI: 10.3390/life14080945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/13/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Obesity has become a widespread issue globally. Morin, a flavonoid with traditional use in managing hyperglycemia and hyperlipidemia, has demonstrated antioxidant and anti-inflammatory properties in experimental studies. This research aims to explore the anti-obesity potential of morin in rats subjected to a high-fat diet (HFD) and investigate whether its effects are mediated through PPARα regulation. METHODS Young adult male Wistar albino rats were divided into four groups (n = 8/group): normal, morin (50 mg/kg/BWT, oral), HFD, and HFD + morin (50 mg/kg/BWT, oral). Treatments were administered daily for 17 consecutive weeks. RESULTS Morin mitigated the elevation in glucose levels and decreased fasting glucose and insulin levels, along with the HOMA-IR index, in HFD-fed rats. Furthermore, morin reduced calorie intake, final body weights, and the masses of subcutaneous, epididymal, peritoneal, and mesenteric fat in these rats. It also attenuated the rise in systolic blood pressure in HFD-fed rats and decreased serum levels of triglycerides, cholesterol, free fatty acids, LDL-c, and leptin, while increasing levels of HDL-c and adiponectin in both normal and HFD-fed rats. Moreover, morin restored normal liver structure and reduced fat vacuole accumulation in HFD-fed rats. Notably, it upregulated mRNA levels of PPARα in the livers and white adipose tissue of both normal and HFD-fed rats. CONCLUSIONS These findings suggest the potential use of morin to enhance fatty acid oxidation in white adipose tissue and mitigate obesity, warranting further clinical investigation into its therapeutic applications.
Collapse
Affiliation(s)
- Laila Naif Al-Harbi
- Department of Food Science and Nutrition, College of Food Science and Agriculture, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
36
|
Al-Sabah S, Al-Khairi I, Jamal M, Qaddoumi M, Alajmi F, Kumar J, Abukhalaf N, Cherian P, Madhu D, Arefanian H, Dsouza C, Alam-Eldin N, AlSabbagh A, Al Madhoun A, Al-Sabah S, Al-Mulla F, Abu-Farha M, Abubaker J. Effect of Dual Glucagon-Like Peptide 1/Glucose-Dependent Insulinotropic Polypeptide Receptor Agonist (Tirzepatide) versus Bariatric Surgery on Weight Loss and Nonalcoholic Fatty Liver Disease. Med Princ Pract 2024; 33:478-490. [PMID: 39047721 PMCID: PMC11460980 DOI: 10.1159/000540534] [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: 12/31/2023] [Accepted: 07/23/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVES Bariatric surgery is a well-established treatment for obesity and type 2 diabetes. Tirzepatide, a dual GIP/GLP-1 receptor agonist, has emerged as a promising therapy for type 2 diabetes. This study aimed to compare the effects of bariatric surgery, semaglutide (a GLP-1 receptor agonist), and tirzepatide in Sprague-Dawley rats fed a high-fat diet. METHODS Rats were divided into surgery, semaglutide, and tirzepatide treatment groups, along with a control group (sham). Weight, oral glucose tolerance, and levels of metabolic markers were assessed, along with adipose and liver tissue analysis. RESULTS Surgery led to a 15.5% weight reduction, while rats treated with semaglutide exhibited a 10.7% reduction. Tirzepatide treatment at various concentrations (10, 50, and 100 nmol/kg) resulted in weight reductions of 5.0%, 14.9%, and 17.7%, respectively, compared to the sham group. Metabolic analyte levels decreased in intervention groups compared to the sham group, indicating improved metabolic health and glucose tolerance. Adipose tissue weight and hepatic liver fat droplets decreased in the intervention groups. CONCLUSION Bariatric surgery and tirzepatide treatment significantly improved metabolic parameters in obese rats. Tirzepatide, particularly at higher concentrations, showed pronounced improvements compared to surgery and semaglutide. These findings suggest that high doses of tirzepatide could be explored as an alternative to bariatric surgery for the treatment of obesity.
Collapse
Affiliation(s)
- Salman Al-Sabah
- Department of Surgery, College of Medicine, Kuwait University, Kuwait City, Kuwait
- Department of Surgery, Jaber Al-Ahmed Hospital, Kuwait City, Kuwait
| | - Irina Al-Khairi
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Mohammad Jamal
- Department of Surgery, College of Medicine, Kuwait University, Kuwait City, Kuwait
- Department of Surgery, Jaber Al-Ahmed Hospital, Kuwait City, Kuwait
| | - Mohammad Qaddoumi
- Department of Pharmacology and Therapeutics, College of Pharmacy, Kuwait University, Kuwait City, Kuwait
| | - Fahad Alajmi
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Jijin Kumar
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Nermeen Abukhalaf
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Preethi Cherian
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Dhanya Madhu
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Hossein Arefanian
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Carol Dsouza
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Nada Alam-Eldin
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Abdullah AlSabbagh
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Suleiman Al-Sabah
- Department of Pharmacology and Toxicology, College of Medicine, Health Sciences Centre, Kuwait University, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| |
Collapse
|
37
|
Rosen N, Mukherjee R, Pancholi P, Sharma M, Solomon H, Timaul M, Thant C, McGriskin R, Hayatt O, Markov V, D'Allara J, Bekker S, Candelier J, Carrasco S, de Stanchina E, Vanaja K. Diet induced insulin resistance is due to induction of PTEN expression. RESEARCH SQUARE 2024:rs.3.rs-4021885. [PMID: 38978604 PMCID: PMC11230483 DOI: 10.21203/rs.3.rs-4021885/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Type 2 Diabetes (T2D) is a condition that is often associated with obesity and defined by reduced sensitivity of PI3K signaling to insulin (insulin resistance), hyperinsulinemia and hyperglycemia. Molecular causes and early signaling events underlying insulin resistance are not well understood. Insulin activation of PI3K signaling causes mTOR dependent induction of PTEN translation, a negative regulator of PI3K signaling. We speculated that insulin resistance is due to insulin dependent induction of PTEN protein that prevent further increases in PI3K signaling. Here we show that in a diet induced model of obesity and insulin resistance, PTEN levels are increased in fat, muscle and liver tissues. Onset of hyperinsulinemia and PTEN induction in tissue is followed by hyperglycemia, hepatic steatosis and severe glucose intolerance. Treatment with a PTEN phosphatase inhibitor prevents and reverses these phenotypes, whereas an mTORC1 kinase inhibitor reverses all but the hepatic steatosis. These data suggest that induction of PTEN by increasing levels of insulin elevates feedback inhibition of the pathway to a point where downstream PI3K signaling is reduced and hyperglycemia ensues. PTEN induction is thus necessary for insulin resistance and the type 2 diabetes phenotype and a potential therapeutic target.
Collapse
|
38
|
Skalski HJ, Arendt AR, Harkins SK, MacLachlan M, Corbett CJM, Goy RW, Kapoor A, Hostetter G, Chandler RL. Key Considerations for Studying the Effects of High-Fat Diet on the Nulligravid Mouse Endometrium. J Endocr Soc 2024; 8:bvae104. [PMID: 38854907 PMCID: PMC11156617 DOI: 10.1210/jendso/bvae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Indexed: 06/11/2024] Open
Abstract
The obesity epidemic continues to increase, with half of US women predicted to be obese by 2030. Women with obesity are at increased risk for not only cardiovascular and liver disease, but also reproductive disorders. Although mouse models are useful in studying the effects of obesity, there is inconsistency in obesity-induction methods, diet composition, and mouse strains, and studies using female mice are limited. In this study, we sought to compare the effects of a 45% high-fat diet (HFD) versus a 60% HFD on the uterine estrous cycle of nulligravid C57BL/6J mice. For 22 weeks, we placed a total of 20 mice on either a 60% HFD, 45% HFD, or each HFD-matched control diet (CD). Both HFDs produced significant weight gain, with 60% HFD and 45% HFD gaining significant weight after 2 weeks and 15 weeks, respectively. Additionally, both HFDs led to glucose intolerance, fatty liver, and adipocyte hypertrophy. Mice fed 60% HFD displayed hyperphagia in the first 12 weeks of HFD treatment. Moreover, 60% HFD-treated mice had a longer estrous cycle length and an increased percentage of estrus stage samplings compared to CD-treated mice. Estrous cycle stage-controlled 60% HFD-treated mice displayed an increased estrogen-to-progesterone ratio and decreased ovarian corpora lutea compared to CD-treated mice, which may underlie the observed estrous cycle differences. There was no significant difference between diets regarding endometrial morphology or the percent of endometrial CD45+ immune cells. Our results indicate that consideration is needed when selecting a HFD-induced obesity mouse model for research involving female reproductive health.
Collapse
Affiliation(s)
- Hilary J Skalski
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Amelia R Arendt
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Shannon K Harkins
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Madison MacLachlan
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Cody J M Corbett
- Wisconsin National Primate Research Center, Assay Services, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Robinson W Goy
- Wisconsin National Primate Research Center, Assay Services, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Amita Kapoor
- Wisconsin National Primate Research Center, Assay Services, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ronald L Chandler
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
39
|
Li B, Chen J, Ou X, Liu X, Xu Z, Xiang X, Yang Y, Wang Q. In-depth multiomic characterization of the effects of obesity in high-fat diet-fed mice. FEBS Open Bio 2024; 14:771-792. [PMID: 38479983 PMCID: PMC11073502 DOI: 10.1002/2211-5463.13788] [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: 09/28/2023] [Revised: 12/08/2023] [Accepted: 03/01/2024] [Indexed: 05/07/2024] Open
Abstract
High-fat diet (HFD)-fed mice have been widely used in the clinical investigation of obesity. However, the long-term effect of HFD on gut microbiota and metabolites, plasma and liver metabolomics, colonic and liver transcriptomics remain largely unknown. In this study, 6-week-old C57BL/6J male mice fed with HFD for 14 weeks showed increased obesity-related indexes including alanine aminotransferase, aspartate aminotransferase, total cholesterol, total triglyceride, free fatty acids, lipopolysaccharides, IL-6, and TNFα. Furthermore, microbial diversity and richness were also significantly decreased. In the colon, genes involved in tryptophan metabolism, PPAR signaling pathway, cholesterol metabolism, and lipid localization and transport, were upregulated. While in the liver, MAPK signaling and unsaturated fatty acid biosynthesis were upregulated. Metabolomic analyses revealed decreased levels of glycerophospholipids and fatty acyl, but increased amino acids, coenzymes and vitamins, and organic acids in the colon, suggesting high absorption of oxidized lipids, while acyl-carnitine, lysophosphatidylcholine, lysophosphatidylethanolamine, and oxidized lipids were reduced in the liver, suggesting a more active lipid metabolism. Finally, correlation analyses revealed a positive correlation between gut microbiota and metabolites and the expression of genes associated with lipid localization, absorption, and transport in the colon, and nutrients and energy metabolism in the liver. Taken together, our results provide a comprehensive characterization of long-term HFD-induced obesity in mice.
Collapse
Affiliation(s)
- Boping Li
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Medicine, Longdong University, Qingyang, China
| | - Juanjuan Chen
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaobin Ou
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Life Sciences and Technology, Longdong University, Qingyang, China
| | - Xiuli Liu
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Life Sciences and Technology, Longdong University, Qingyang, China
| | - Zaoxu Xu
- Gansu Key Laboratory of Protection and Utilization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang, China
- College of Life Sciences and Technology, Longdong University, Qingyang, China
| | - Xuesong Xiang
- Element Nutrition of National Health Commission, National Institute of Nutrition and Health, China CDC, Beijing, China
| | - Yan Yang
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, China
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
40
|
Rigaudière JP, Jouve C, Capel F, Patrac V, Miguel B, Tournadre A, Demaison L. An experimental model of western diet in female Wistar rats leads to cardiac hypoxia related to a stimulated contractility. J Physiol Biochem 2024; 80:287-302. [PMID: 38175500 DOI: 10.1007/s13105-023-01003-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
Previous studies in Western diet (WD)-fed male rats have highlighted a link between the stimulation of cardiac contractility, mitochondrial adaptations and a pro-inflammatory fatty acid profile of phospholipids in the heart. Our objectives were to determine (1) if WD-fed female Wistar rats and obese humans display a similar pro-inflammatory profile in their cardiac phospholipids and (2) if this lipid profile is associated with deleterious effects on the heart of the female rodents. Female Wistar rats were fed WD for 5 weeks or a laboratory chow as a control. Ionic homeostasis, redox status, inflammation markers, and fatty acid composition of phospholipids were analysed in the heart. WD increased the abdominal fat mass without modifying the body weight of female rats. As previously found in males, a WD induced a shift in membrane fatty acid composition toward a pro-inflammatory profile in the female rats, but not in obese humans. It was associated with an increased COX2 expression suggesting an increased pro-inflammatory eicosanoid production. Signs of increased intracellular calcium strongly supported a stimulation of cardiac contractility without any induction of apoptosis. The heart of WD-fed rats exhibited a hypoxic state as a higher HIF1-α expression was reported. The expressions of antioxidant enzymes were increased, but the redox reserves against reactive oxygen species were lowered. In conclusion, as previously observed in males, we suppose that cardiac abnormalities are magnified with severe obesity in female rats, leading to hypoxia and intense oxidative stress which could ultimately induce cell death and heart failure.
Collapse
Affiliation(s)
- Jean-Paul Rigaudière
- UMR 1019 Unité de Nutrition Humaine, CRNH Auvergne Université Clermont Auvergne, INRAE, 28 Place Henri Dunant, TSA 50400, 63000 Cedex 1, Clermont-Ferrand, France
| | - Chrystèle Jouve
- UMR 1019 Unité de Nutrition Humaine, CRNH Auvergne Université Clermont Auvergne, INRAE, 28 Place Henri Dunant, TSA 50400, 63000 Cedex 1, Clermont-Ferrand, France
| | - Frédéric Capel
- UMR 1019 Unité de Nutrition Humaine, CRNH Auvergne Université Clermont Auvergne, INRAE, 28 Place Henri Dunant, TSA 50400, 63000 Cedex 1, Clermont-Ferrand, France
| | - Véronique Patrac
- UMR 1019 Unité de Nutrition Humaine, CRNH Auvergne Université Clermont Auvergne, INRAE, 28 Place Henri Dunant, TSA 50400, 63000 Cedex 1, Clermont-Ferrand, France
| | - Bruno Miguel
- Heart Surgery Department, Gabriel Montpied Hospital, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Anne Tournadre
- Service de Rhumatologie, Centre Hospitalier Universitaire Gabriel Montpied, 63000, Clermont-Ferrand, France
| | - Luc Demaison
- UMR 1019 Unité de Nutrition Humaine, CRNH Auvergne Université Clermont Auvergne, INRAE, 28 Place Henri Dunant, TSA 50400, 63000 Cedex 1, Clermont-Ferrand, France.
| |
Collapse
|
41
|
de Medeiros WF, Gomes AFT, Aguiar AJFC, de Queiroz JLC, Bezerra IWL, da Silva-Maia JK, Piuvezam G, Morais AHDA. Anti-Obesity Therapeutic Targets Studied In Silico and In Vivo: A Systematic Review. Int J Mol Sci 2024; 25:4699. [PMID: 38731918 PMCID: PMC11083175 DOI: 10.3390/ijms25094699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
In the age of information technology and the additional computational search tools and software available, this systematic review aimed to identify potential therapeutic targets for obesity, evaluated in silico and subsequently validated in vivo. The systematic review was initially guided by the research question "What therapeutic targets have been used in in silico analysis for the treatment of obesity?" and structured based on the acronym PECo (P, problem; E, exposure; Co, context). The systematic review protocol was formulated and registered in PROSPERO (CRD42022353808) in accordance with the Preferred Reporting Items Checklist for Systematic Review and Meta-Analysis Protocols (PRISMA-P), and the PRISMA was followed for the systematic review. The studies were selected according to the eligibility criteria, aligned with PECo, in the following databases: PubMed, ScienceDirect, Scopus, Web of Science, BVS, and EMBASE. The search strategy yielded 1142 articles, from which, based on the evaluation criteria, 12 were included in the systematic review. Only seven these articles allowed the identification of both in silico and in vivo reassessed therapeutic targets. Among these targets, five were exclusively experimental, one was exclusively theoretical, and one of the targets presented an experimental portion and a portion obtained by modeling. The predominant methodology used was molecular docking and the most studied target was Human Pancreatic Lipase (HPL) (n = 4). The lack of methodological details resulted in more than 50% of the papers being categorized with an "unclear risk of bias" across eight out of the eleven evaluated criteria. From the current systematic review, it seems evident that integrating in silico methodologies into studies of potential drug targets for the exploration of new therapeutic agents provides an important tool, given the ongoing challenges in controlling obesity.
Collapse
Affiliation(s)
- Wendjilla F. de Medeiros
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil; (W.F.d.M.); (A.F.T.G.); (I.W.L.B.); (J.K.d.S.-M.)
| | - Ana Francisca T. Gomes
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil; (W.F.d.M.); (A.F.T.G.); (I.W.L.B.); (J.K.d.S.-M.)
| | - Ana Júlia F. C. Aguiar
- Biochemistry and Molecular Biology Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (A.J.F.C.A.); (J.L.C.d.Q.)
| | - Jaluza Luana C. de Queiroz
- Biochemistry and Molecular Biology Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (A.J.F.C.A.); (J.L.C.d.Q.)
| | - Ingrid Wilza L. Bezerra
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil; (W.F.d.M.); (A.F.T.G.); (I.W.L.B.); (J.K.d.S.-M.)
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
| | - Juliana Kelly da Silva-Maia
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil; (W.F.d.M.); (A.F.T.G.); (I.W.L.B.); (J.K.d.S.-M.)
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
| | - Grasiela Piuvezam
- Public Health Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-400, Brazil;
- Public Health Department, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
| | - Ana Heloneida de A. Morais
- Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil; (W.F.d.M.); (A.F.T.G.); (I.W.L.B.); (J.K.d.S.-M.)
- Biochemistry and Molecular Biology Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil; (A.J.F.C.A.); (J.L.C.d.Q.)
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
| |
Collapse
|
42
|
Moral R, Kapravelou G, Cubedo M, Solanas M, Escrich E. Body weight gain and control: beneficial effect of extra virgin olive oil versus corn oil in an experimental model of mammary cancer. J Nutr Biochem 2024; 125:109549. [PMID: 38104866 DOI: 10.1016/j.jnutbio.2023.109549] [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: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Obesity is a known risk factor for breast cancer, the most common malignancy among women worldwide. We have previously described different effects of high-fat diets on mammary experimental carcinogenesis. In this work, we analyzed the animal growth data obtained in six experimental assays, in healthy and carcinogen-induced rats undergoing different dietary interventions. The animals were fed with three experimental diets administered at different periods of development: a control low-fat diet, and two isocaloric high-fat diets (rich in corn oil or in extravirgin olive oil -EVOO-). Weekly weight throughout the development of 818 animals have been compiled and reanalyzed using adjusted mathematical models. Molecular mechanisms have been investigated: ethanolamides in small intestine, neuropeptides controlling satiety in hypothalamus, and proteins controlling lipid metabolism in adipose and mammary tissues. The results indicated that the effect of diets depended on type of lipid, timing of intervention and health status. The high corn oil diet, but not the high EVOO diet, increased body weight and mass, especially if administered from weaning, in healthy animals and in those that received a moderate dose of carcinogen. The potential protective effect of EVOO on weight maintenance may be related to anorexigenic neuropeptides such as oxytocin and lipolysis/deposition balance in adipose tissue (increasing phospho-PKA, HSL, MGL and decreasing FAS). In animals with cancer, body weight gain was related to the severity of the disease. Taken together, our results suggest that EVOO has a beneficial effect on body weight maintenance in both health and cancer.
Collapse
Affiliation(s)
- Raquel Moral
- Department of Cell Biology, Physiology and Immunology, Physiology Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| | - Garyfallia Kapravelou
- Department of Cell Biology, Physiology and Immunology, Physiology Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Marta Cubedo
- Department of Statistics, Universitat de Barcelona, Barcelona, Spain
| | - Montserrat Solanas
- Department of Cell Biology, Physiology and Immunology, Physiology Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Eduard Escrich
- Department of Cell Biology, Physiology and Immunology, Physiology Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| |
Collapse
|
43
|
Beaudry AG, Law ML, Gilley-Connor KR, Buley H, Dungan CM, Nascimento CMC, Vichaya EG, Wiggs MP. Diet-induced obesity does not exacerbate cachexia in male mice bearing Lewis-lung carcinoma tumors. Am J Physiol Regul Integr Comp Physiol 2024; 326:R254-R265. [PMID: 38252513 DOI: 10.1152/ajpregu.00208.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
Cachexia is a muscle-wasting syndrome commonly observed in patients with cancer, which can significantly worsen clinical outcomes. Because of a global rise in obesity, the coexistence of cachexia in obese individuals poses unique challenges, with the impact of excessive adiposity on cachexia severity and underlying pathophysiology not well defined. Understanding the interplay between cachexia and obesity is crucial for improving diagnosis and treatment strategies for these patients; therefore, the present study examined differences in cachexia between lean and obese mice bearing Lewis lung carcinoma (LLC) tumors. Nine-week-old, male C57Bl6J mice were placed on either a chow or a high-fat diet (HFD) for 9 wk. After the diet intervention, mice were inoculated with LLC or vehicle. Markers of cachexia, such as body and muscle loss, were noted in both chow and HFD groups with tumors. Tumor weight of HFD animals was greater than that of chow. LLC tumors reduced gastrocnemius, plantaris, and soleus mass, regardless of diet. The tibialis anterior and plantaris mass and cross-sectional area of type IIb/x fibers in the gastrocnemius were not different between HFD-chow, HFD-tumor, and chow-tumor. Using RNA sequencing (RNA-seq) of the plantaris muscle from chow-tumor and HFD-tumor groups, we identified ∼400 differentially expressed genes. Bioinformatic analysis identified changes in lipid metabolism, mitochondria, bioenergetics, and proteasome degradation. Atrophy was not greater despite larger tumor burden in animals fed an HFD, and RNA-seq data suggests that partial protection is mediated through differences in mitochondrial function and protein degradation, which may serve as future mechanistic targets.NEW & NOTEWORTHY This study provides timely information on the interaction between obesity and cancer cachexia. Lean and obese animals show signs of cachexia with reduced body weight, adipose tissue, and gastrocnemius muscle mass. There was not significant wasting in the tibialis anterior, plantaris, or fast twitch fibers in the gastrocnemius muscle of obese animals with tumors. RNA-seq analysis reveals that obese tumor bearing animals had differential expression of mitochondria- and degradation-related genes, which may direct future studies in mechanistic research.
Collapse
Affiliation(s)
- Anna G Beaudry
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, Texas, United States
| | - Michelle L Law
- Department of Human Sciences and Design, Baylor University, Waco, Texas, United States
| | - Kayla R Gilley-Connor
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States
| | - Hailey Buley
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States
| | - Cory M Dungan
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, Texas, United States
| | | | - Elisabeth G Vichaya
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, United States
| | - Michael P Wiggs
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, Texas, United States
| |
Collapse
|
44
|
Pradel-Mora JJ, Marín G, Castillo-Rangel C, Hernández-Contreras KA, Vichi-Ramírez MM, Zarate-Calderon C, Herran Motta FS. Oxidative Stress in Postbariatric Patients: A Systematic Literature Review Exploring the Long-term Effects of Bariatric Surgery. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e5646. [PMID: 38515558 PMCID: PMC10956951 DOI: 10.1097/gox.0000000000005646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 01/16/2024] [Indexed: 03/23/2024]
Abstract
Background The present study investigates the impact of oxidative stress after bariatric surgery in patients with obesity. This field of study has gained great interest in recent years due to the role that oxidative stress plays in metabolic diseases. Obesity, by itself, can generate an increase in reactive oxygen and nitrogen species, intensifying cellular damage and promoting the progression of adverse metabolic conditions. In this context, bariatric surgery emerges as a candidate capable of modifying oxidative stress biomarkers, facilitating the patient's metabolic recovery. Methods A systematic review was carried out, identifying 30 studies found in databases such as PubMed, Scopus, Web of Science, and Google Scholar. It looked at the link between oxidative stress and recovery after bariatric surgery in patients. The selection of studies was based on the measurement of oxidative stress biomarkers before and after surgical intervention. Results The results reveal a significant decrease in oxidative stress biomarkers after bariatric surgery. However, a notable variability in antioxidant activity is observed between different patients, as well as a significant influence of comorbidities. Conclusions Bariatric surgery is postulated as an effective intervention in reducing oxidative stress in patients with obesity, enhancing antioxidant activity and improving patient recovery. This finding highlights the importance of considering oxidative stress management as an integral part of postoperative care, suggesting the need to implement complementary treatment strategies to optimize health outcomes.
Collapse
Affiliation(s)
- Jessica Juliana Pradel-Mora
- From the Plastic and Reconstructive Surgery, “UMAE Hospital de Especialidades Dr. Bernardo Sepúlveda Gutiérrez, Centro Médico Nacional Siglo XXI, Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Gerardo Marín
- Neural Dynamics and Modulation Lab, Cleveland Clinic, Cleveland, Ohio
| | - Carlos Castillo-Rangel
- Department of Neurosurgery, “Hospital Regional 1º de Octubre,” Institute of Social Security and Services for State Workers (ISSSTE), Mexico City, Mexico
| | | | | | | | - Fanny Stella Herran Motta
- Plastic and Reconstructive Surgery, “Centro Médico Nacional 20 de noviembre,” Institute of Social Security and Services for State Workers (ISSSTE), Mexico City, Mexico
| |
Collapse
|
45
|
Eisinger K, Girke P, Buechler C, Krautbauer S. Adipose tissue depot specific expression and regulation of fibrosis-related genes and proteins in experimental obesity. Mamm Genome 2024; 35:13-30. [PMID: 37884762 PMCID: PMC10884164 DOI: 10.1007/s00335-023-10022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Transforming growth factor beta (Tgfb) is a well-studied pro-fibrotic cytokine, which upregulates cellular communication network factor 2 (Ccn2), collagen, and actin alpha 2, smooth muscle (Acta2) expression. Obesity induces adipose tissue fibrosis, which contributes to metabolic diseases. This work aimed to analyze the expression of Tgfb, Ccn2, collagen1a1 (Col1a1), Acta2 and BMP and activin membrane-bound inhibitor (Bambi), which is a negative regulator of Tgfb signaling, in different adipose tissue depots of mice fed a standard chow, mice fed a high fat diet (HFD) and ob/ob mice. Principally, these genes were low expressed in brown adipose tissues and this difference was less evident for the ob/ob mice. Ccn2 and Bambi protein as well as mRNA expression, and collagen1a1 mRNA were not induced in the adipose tissues upon HFD feeding whereas Tgfb and Acta2 mRNA increased in the white fat depots. Immunoblot analysis showed that Acta2 protein was higher in subcutaneous and perirenal fat of these mice. In the ob/ob mice, Ccn2 mRNA and Ccn2 protein were upregulated in the fat depots. Here, Tgfb, Acta2 and Col1a1 mRNA levels and serum Tgfb protein were increased. Acta2 protein was, however, not higher in subcutaneous and perirenal fat of these mice. Col6a1 mRNA was shown before to be higher in obese fat tissues. Current analysis proved the Col6a1 protein was induced in subcutaneous fat of HFD fed mice. Notably, Col6a1 was reduced in perirenal fat of ob/ob mice in comparison to the respective controls. 3T3-L1 cells express Ccn2 and Bambi protein, whose levels were not changed by fatty acids, leptin, lipopolysaccharide, tumor necrosis factor and interleukin-6. All of these factors led to higher Tgfb in 3T3-L1 adipocyte media but did not increase its mRNA levels. Free fatty acids induced necrosis whereas apoptosis did not occur in any of the in vitro incubations excluding cell death as a main reason for higher Tgfb in cell media. In summary, Tgfb mRNA is consistently induced in white fat tissues in obesity but this is not paralleled by a clear increase of its target genes. Moreover, discrepancies between mRNA and protein expression of Acta2 were observed. Adipocytes seemingly do not contribute to higher Tgfb mRNA levels in obesity. These cells release more Tgfb protein when challenged with obesity-related metabolites connecting metabolic dysfunction and fibrosis.
Collapse
Affiliation(s)
- Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Philipp Girke
- Department of Genetics, University of Regensburg, 93040, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany.
| | - Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| |
Collapse
|
46
|
Connolly K, Batacan R, Jackson D, Fenning AS. Effects of epicatechin on cardiovascular function in middle-aged diet-induced obese rat models of metabolic syndrome. Br J Nutr 2024; 131:593-605. [PMID: 37732427 PMCID: PMC10803822 DOI: 10.1017/s000711452300209x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/19/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
The current study aimed to investigate the cardiovascular effects of epicatechin, a flavonoid found in green tea and cocoa, in attenuating complications associated with metabolic syndrome in diet-induced obese rats. Male Wistar-Kyoto (WKY) rats aged 16 weeks were fed either standard rat chow or given a high-fat-high-carbohydrate (HFHC) diet for 20 weeks. Epicatechin treatment (5 mg/kg/d) was administered to a subset of WKY rats commencing at week 8 of the 20 week HFHC feeding period. Body weights, food, water and energy intakes, blood pressure, heart rate and glucose tolerance were measured throughout the treatment period. Oxidative stress and inflammatory markers, lipid levels, cardiac collagen deposition, cardiac electrical function, aortic and mesenteric vessel reactivity were examined after the treatment. Twenty weeks of HFHC feeding in WKY rats resulted in the development of metabolic syndrome indicated by the presence of abdominal obesity, dyslipidaemia, glucose intolerance and increased blood pressure. Epicatechin treatment was found to enhance the oxidative stress status in HFHC groups through an increase in serum nitric oxide levels and a decrease in 8-isoprostane concentrations. Furthermore, WKY-HFHC rats displayed a decrease in IL-6 levels. The lipid profiles in HFHC groups showed improvement, with a decrease in LDL-cholesterol and TAG and an increase in HDL-cholesterol levels observed in WKY-HFHC rats. However, epicatechin was not effective in preventing weight gain, glucose intolerance or hypertension in HFHC fed rats. Overall, the results of this study suggest that epicatechin has the potential to improve the underlying mechanisms associated with metabolic syndrome in obese rats.
Collapse
Affiliation(s)
- Kylie Connolly
- School of Health, Medical and Applied Sciences, Central Queensland University, Bruce Highway, Rockhampton, QLD4701, Australia
| | - Romeo Batacan
- School of Health, Medical and Applied Sciences, Central Queensland University, Bruce Highway, Rockhampton, QLD4701, Australia
| | - Douglas Jackson
- Australian Catholic University, 40 Edward St, North Sydney, NSW2060, Australia
| | - Andrew Stuart Fenning
- School of Health, Medical and Applied Sciences, Central Queensland University, Bruce Highway, Rockhampton, QLD4701, Australia
| |
Collapse
|
47
|
Eng M, Suthaaharan K, Newton L, Sheikh F, Fox-Robichaud A. Sepsis and obesity: a scoping review of diet-induced obesity murine models. Intensive Care Med Exp 2024; 12:15. [PMID: 38388878 PMCID: PMC10884395 DOI: 10.1186/s40635-024-00603-0] [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: 10/16/2023] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Sepsis, the life-threatening host response to infection, is a major cause of mortality. Obesity increases vulnerability to sepsis; however, some degree of obesity may be protective, called the "obesity paradox". This scoping review systematically maps the literature on outcomes associated with diet-induced obesity and sepsis-induced organ injury, focusing on non-transgenic murine models. METHODS A literature search of primary articles was conducted from database inception to June 2023. Eligible articles compared diet-induced obesity to non-obese mice in sepsis models involving live pathogens. Two reviewers screened articles and extracted data on obesogenic and sepsis models utilized, and organ injury outcomes, including physiological dysfunction, histological alterations, and biochemical changes. RESULTS Seventeen studies met eligibility criteria; 82% used male C57BL/6 mice, and 88% used cecal ligation and puncture to induce sepsis. Most studies used 60% high-fat diets compared to 10-16% fat in controls. Seven (64%) studies reported increased mortality in obese septic mice, one (9%) observed a decrease, and three (37%) found no significant difference. The liver, lungs, and kidneys were the most studied organs. Alanine transaminase results were inconclusive. Myeloperoxidase levels were increased in the livers of two studies and inconclusive in the lungs of obese septic mice. Creatinine and neutrophil gelatinase-associated lipocalin were elevated in obese septic mice. CONCLUSIONS There is variability in the methodology and measured outcomes in murine models of diet-induced obesity and sepsis and a lack of studies in female mice. The absence of standardized models has produced conflicting findings on the impact of obesity on sepsis outcomes.
Collapse
Affiliation(s)
- Mikaela Eng
- Thrombosis and Atherosclerosis Research Institute (TaARI), Hamilton, Canada
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Keshikaa Suthaaharan
- Thrombosis and Atherosclerosis Research Institute (TaARI), Hamilton, Canada
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Logan Newton
- Thrombosis and Atherosclerosis Research Institute (TaARI), Hamilton, Canada
| | - Fatima Sheikh
- Department of Health Research Methods, Evidence and Impact, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Alison Fox-Robichaud
- Thrombosis and Atherosclerosis Research Institute (TaARI), Hamilton, Canada.
- Division of Critical Care, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada.
| |
Collapse
|
48
|
Chávez-Ortega MP, Almanza-Pérez JC, Sánchez-Muñoz F, Hong E, Velázquez-Reyes E, Romero-Nava R, Villafaña-Rauda S, Pérez-Ontiveros A, Blancas-Flores G, Huang F. Effect of Supplementation with Omega-3 Polyunsaturated Fatty Acids on Metabolic Modulators in Skeletal Muscle of Rats with an Obesogenic High-Fat Diet. Pharmaceuticals (Basel) 2024; 17:222. [PMID: 38399437 PMCID: PMC10892617 DOI: 10.3390/ph17020222] [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: 10/26/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 02/25/2024] Open
Abstract
Previous studies provided evidence of the benefits of omega-3 polyunsaturated fatty acids (ω-3 PUFA) on the cardiovascular system and inflammation. However, its possible effect on skeletal muscle is unknown. This study aimed to evaluate whether ω-3 PUFA reverses the dysregulation of metabolic modulators in the skeletal muscle of rats on a high-fat obesogenic diet. For this purpose, an animal model was developed using male Wistar rats with a high-fat diet (HFD) and subsequently supplemented with ω-3 PUFA. Insulin resistance was assessed, and gene and protein expression of metabolism modulators in skeletal muscle was also calculated using PCR-RT and Western blot. Our results confirmed that in HFD rats, zoometric parameters and insulin resistance were increased compared to SD rats. Furthermore, we demonstrate reduced gene and protein expression of peroxisome proliferator-activated receptors (PPARs) and insulin signaling molecules. After ω-3 PUFA supplementation, we observed that glucose (24.34%), triglycerides (35.78%), and HOMA-IR (40.10%) were reduced, and QUICKI (12.16%) increased compared to HFD rats. Furthermore, in skeletal muscle, we detected increased gene and protein expression of PPAR-α, PPAR-γ, insulin receptor (INSR), insulin receptor substrate 1 (ISR-1), phosphatidylinositol-3-kinase (PI3K), and glucose transporter 4 (GLUT-4). These findings suggest that ω-3 PUFAs decrease insulin resistance of obese skeletal muscle.
Collapse
Affiliation(s)
- Mara Patricia Chávez-Ortega
- Posgrado en Biología Experimental, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico;
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| | - Julio Cesar Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico; (J.C.A.-P.); (E.V.-R.)
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico;
| | - Enrique Hong
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico;
| | - Elihu Velázquez-Reyes
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico; (J.C.A.-P.); (E.V.-R.)
| | - Rodrigo Romero-Nava
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.R.-N.); (S.V.-R.)
| | - Santiago Villafaña-Rauda
- Laboratorio de Señalización Intracelular, Sección de Estudios de Posgrado, Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (R.R.-N.); (S.V.-R.)
| | - Alfredo Pérez-Ontiveros
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| | - Gerardo Blancas-Flores
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Iztapalapa, Ciudad de México 02200, Mexico; (J.C.A.-P.); (E.V.-R.)
| | - Fengyang Huang
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| |
Collapse
|
49
|
Mannozzi J, Massoud L, Stavres J, Al-Hassan MH, O’Leary DS. Altered Autonomic Function in Metabolic Syndrome: Interactive Effects of Multiple Components. J Clin Med 2024; 13:895. [PMID: 38337589 PMCID: PMC10856260 DOI: 10.3390/jcm13030895] [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: 01/03/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Metabolic syndrome (MetS) describes a set of disorders that collectively influence cardiovascular health, and includes hypertension, obesity, insulin resistance, diabetes, and dyslipidemia. All these components (hypertension, obesity, dyslipidemia, and prediabetes/diabetes) have been shown to modify autonomic function. The major autonomic dysfunction that has been documented with each of these components is in the control of sympathetic outflow to the heart and periphery at rest and during exercise through modulation of the arterial baroreflex and the muscle metaboreflex. Many studies have described MetS components in singularity or in combination with the other major components of metabolic syndrome. However, many studies lack the capability to study all the factors of metabolic syndrome in one model or have not focused on studying the effects of how each component as it arises influences overall autonomic function. The goal of this review is to describe the current understanding of major aspects of metabolic syndrome that most likely contribute to the consequent/associated autonomic alterations during exercise and discuss their effects, as well as bring light to alternative mechanisms of study.
Collapse
Affiliation(s)
- Joseph Mannozzi
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48001, USA
| | - Louis Massoud
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48001, USA
| | - Jon Stavres
- School of Kinesiology and Nutrition, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | | | - Donal S. O’Leary
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48001, USA
| |
Collapse
|
50
|
Gil TY, Park J, Park YJ, Kim HJ, Cominguez DC, An HJ. Drynaria rhizome water extract alleviates high‑fat diet‑induced obesity in mice. Mol Med Rep 2024; 29:30. [PMID: 38131179 PMCID: PMC10784730 DOI: 10.3892/mmr.2023.13153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Drynaria rhizome is a herbal medicine used for strengthening bones and treating bone diseases in East Asia. Although obesity is considered to benefit bone formation, it has been revealed that visceral fat accumulation can promote osteoporosis. Given the complex relationship between bone metabolism and obesity, bone‑strengthening medicines should be evaluated while considering the effects of obesity. The present study investigated the effects of Drynaria rhizome extract (DRE) on high‑fat diet (HFD)‑induced obese mice. DRE was supplemented with the HFD. Body weight, food intake, the expression levels of lipogenesis transcription factors, including sterol regulatory element binding protein (SREBP)‑1, peroxisome proliferator‑activated receptor (PPAR)‑γ and adenosine monophosphate‑activated protein kinase (AMPK)‑α, and AMPK activation were evaluated. Mice fed DRE and a HFD exhibited reduced body weight without differences in food intake compared with those in the HFD group. Furthermore, DRE; upregulated AMPK‑α of epididymal one; down‑regulated SREBP‑1 and PPAR‑γ, as determined using western blotting and quantitative polymerase chain reaction, respectively. Decreased lipid accumulation were observed in both fat pad and liver of HFD‑fed mice, which were suppressed by DRE treatment. These results demonstrated the potential of DRE as a dietary natural product for strengthening bones and managing obesity.
Collapse
Affiliation(s)
- Tae-Young Gil
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Junkyu Park
- Department of Science in Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yea-Jin Park
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon 26339, Republic of Korea
| | - Hyo-Jung Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Divina C. Cominguez
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon 26339, Republic of Korea
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|