1
|
Aguilar V, Le Master E, Paul A, Ahn SJ, Lazarko D, Febbraio M, Mehta D, Lee J, Levitan I. Endothelial Stiffening Induced by CD36-Mediated Lipid Uptake Leads to Endothelial Barrier Disruption and Contributes to Atherosclerotic Lesions. Arterioscler Thromb Vasc Biol 2025. [PMID: 40207364 DOI: 10.1161/atvbaha.124.322244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Endothelial stiffening induced by Western diet was proposed to be an important factor in vascular dysfunction. In this study, we determine the role of endothelial CD36 (cluster of differentiation 36) in stiffening, disruption of aortic endothelial barrier, and atherosclerosis in mouse models of obesity and hypercholesterolemia. METHODS To address this goal, we generated an endothelial-specific inducible knockdown mouse model of CD36, Cdh5.CreERT2CD36fl/fl, on C57/BL6J wild-type and LDLR-/- genetic backgrounds. Endothelial stiffness is assessed by atomic force microscopy; endothelial barrier integrity is assessed by imaging VE-cadherin junctions and by penetration of Evans blue dye into the aortic wall. Atherosclerotic plaques are quantified using oil red O staining. RESULTS Endothelial-specific downregulation of CD36 abrogates stiffening of aortic endothelium induced by Western diet in Cdh5.CreERT2CD36fl/fl and in Cdh5.CreERT2CD36fl/flLDLR-/- mice. Prevention of Western diet-induced endothelial stiffening by downregulation of CD36 is associated with a protective effect against endothelial barrier disruption in both mouse models and with a significant decrease in the areas of atherosclerotic lesions in Cdh5.CreERT2CD36fl/flLDLR-/- mice. Mechanistically, stiffening of human aortic endothelial cells in vitro is induced by saturated fatty acids, particularly palmitic acid (PA), which results in activation of RhoA. Both PA-induced endothelial stiffening and RhoA activation are abrogated by CD36 siRNA. Furthermore, PA-induced endothelial stiffening of excised aortas ex vivo is lost in aortas isolated from mice, where endothelial CD36 is downregulated. We also demonstrate that PA-induced activation of RhoA and endothelial stiffening require expressing an RhoA-inhibitory protein, Rho-GDI1 (Rho guanosine dissociation inhibitor 1). Finally, we discover that PA disrupts the colocalization of RhoA with Rho-GDI1. CONCLUSIONS We conclude that stiffening of the aortic endothelium by CD36-mediated uptake of fatty acids contributes significantly to Western diet-induced vascular dysfunction and atherosclerosis. We further propose that fatty acids may activate RhoA by inducing its dissociation from Rho-GDI1.
Collapse
Affiliation(s)
- Victor Aguilar
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
- Department of Biomedical Engineering, University of Illinois at Chicago. (V.A., J.L., I.L.)
| | - Elizabeth Le Master
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Amit Paul
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Sang Joon Ahn
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Dana Lazarko
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
| | - Maria Febbraio
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada (M.F.)
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago. (D.M.)
| | - James Lee
- Department of Biomedical Engineering, University of Illinois at Chicago. (V.A., J.L., I.L.)
| | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago. (V.A., E.L.M., A.P., S.J.A., D.L., I.L.)
- Department of Biomedical Engineering, University of Illinois at Chicago. (V.A., J.L., I.L.)
| |
Collapse
|
2
|
Sun Y, Shan X, Li M, Niu Y, Sun Z, Ma X, Wang T, Zhang J, Niu D. Autoimmune mechanisms and inflammation in obesity-associated type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease. Funct Integr Genomics 2025; 25:84. [PMID: 40205260 DOI: 10.1007/s10142-025-01587-0] [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: 03/11/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Obesity, characterized by the excessive accumulation of white adipose tissue, is a significant global health burden and a major risk factor for a range of diseases, including malignancies and metabolic disorders. Individuals with high visceral fat content are particularly susceptible to severe complications such as type 2 diabetes, cardiovascular diseases, and liver disorders. However, the pathogenesis of obesity-related metabolic diseases extends beyond simple adiposity. Chronic obesity triggers a prolonged inflammatory response, which leads to tissue fibrosis and sustained organ damage, contributing to multi-organ dysfunction. This review explores the autoimmune mechanisms and inflammatory pathways underlying obesity-induced type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease, with an emphasis on their interrelated pathophysiology and the potential for therapeutic interventions.
Collapse
Grants
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
Collapse
Affiliation(s)
- Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yifan Niu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, 211300, Jiangsu, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
3
|
Bao Y, Osowiecka M, Ott C, Tziraki V, Meusburger L, Blaßnig C, Krivda D, Pjevac P, Séneca J, Strauss M, Steffen C, Heck V, Aygün S, Duszka K, Doppelmayer K, Grune T, Pignitter M. Dietary oxidized lipids in redox biology: Oxidized olive oil disrupts lipid metabolism and induces intestinal and hepatic inflammation in C57BL/6J mice. Redox Biol 2025; 81:103575. [PMID: 40043451 PMCID: PMC11927754 DOI: 10.1016/j.redox.2025.103575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Olive oil, rich in oleic acid, is often regarded as a healthier alternative to animal fats high in saturated fatty acids and plant oils rich in oxidizable polyunsaturated fatty acids. However, the redox biological implications and health effects of oxidized olive oil (ox-OO) remain underexplored. Our study investigated its impact on lipid metabolism, intestinal and hepatic inflammation, and gut microbiota. Female C57BL/6J mice were fed either a standard normal (NFD), high-fat diet (HFD), an NFD-ox-OO or HFD-ox-OO, in which ox-OO (180 °C heating, 10 min) was the sole lipid source. Inflammation was assessed using macrophage marker F4/80 immunohistochemical (IHC) staining. Gene expression of inflammatory and lipid metabolism markers (IL-10, NF-kBp65, IL-1β, TNFα, TLR4, COX2, PPARα, PPARγ, CPT1a, SCAD, MCAD, LCAD) was analyzed by qRT-PCR. Soluble epoxide hydrolase (sEH) protein expression was measured using IHC. Oxylipin and carnitine profiles were determined by LC-MS/MS. Gut microbiota was analyzed by 16S rRNA sequencing. Ox-OO disrupted redox homeostasis, leading to lipid metabolic dysfunction in the intestines and liver. In the duodenum and proximal jejunum, ox-OO decreased the levels of anti-inflammatory oxylipins and increased pro-inflammatory mediators, leading to inflammation. In the ileum and colon, ox-OO caused lipid metabolic dysregulation and inflammation. Colon inflammation was linked to inhibited mitochondrial β-oxidation and decreased short-chain fatty acid-producing microbiomes. Notably, redox imbalances were further implicated by the identification of 9,10-epoxy-stearic acid, a novel inflammatory lipid mediator oxidized from dietary oleic acid, which upregulated sEH. Ox-OO affects lipid metabolism and may contribute to inflammation in the gut and liver, raising questions about the assumption that olive oil is always beneficial and suggesting possible risks linked to oxidized oleic acid.
Collapse
Affiliation(s)
- Yifan Bao
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Doctoral School of Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Magdalena Osowiecka
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Doctoral School of Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christiane Ott
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Vasiliki Tziraki
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Lukas Meusburger
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Claudia Blaßnig
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Daniela Krivda
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
| | - Joana Séneca
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
| | - Matthias Strauss
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Doctoral School of Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christina Steffen
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Verena Heck
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Soner Aygün
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Kalina Duszka
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Kevin Doppelmayer
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Tilman Grune
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Marc Pignitter
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.
| |
Collapse
|
4
|
Mirzababaei A, Shiraseb F, Mohamadi A, Mehri Hajmir M, Ebrahimi S, Zarrinvafa Z, Kazemian E, Mehrvar A, Mirzaei K. The association between nutrient patterns and hospital stay duration and symptoms in COVID-19 in Iranian patients: cross-sectional study. Front Nutr 2025; 12:1542449. [PMID: 40098737 PMCID: PMC11911187 DOI: 10.3389/fnut.2025.1542449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Background An excessively reactive immune system results in the cytokine storm COVID-19. A healthy diet is essential to maintain the balance between the immune system and inflammatory and oxidative stress. Associations between single foods and nutrients and COVID-19 have been examined. However, no prior study has examined associations between nutrient patterns and COVID-19. This study assessed the link between nutrient patterns and the COVID-19 severity and length of hospital stay in Iranian adults. Methods This cross-sectional study included 107 Iranian adults aged 20-60 years, who were admitted to Amir Alam Hospital in Tehran, Iran, due to COVID-19. Data on their symptoms were collected through a demographic questionnaire and verified against their hospital records. Three non-consecutive 24-h dietary recalls were used to collect participants' food and beverage intake. Principal component analysis (PCA) was used to derive nutrient patterns. Result A total of 95 Covid patients with a mean age of 46.2 years were included. Four major dietary patterns were identified using the Scree Plot chart, including high carbohydrate and high minerals pattern; high protein and high vitamins pattern; high fat pattern; and poor nutrient pattern. Adherence to the poor nutrient patterns was associated with a higher number of hospitalization days and lower appetite (p < 0.05). The poor dietary patterns were associated with an increased likelihood of headache, fever, and respiratory distress syndrome (RDS). Also, headaches were more common with adherence to the high-fat pattern (p < 0.05). Conclusion The findings of this study show that a poor nutrient pattern is related to longer hospital stays and reduced appetite. It also connected to an increased likelihood of symptoms including headaches, fever, and respiratory distress syndrome. A strong association was found between respiratory distress syndrome, headaches, and a high-fat diet was found. Further studies with prospective designs are needed to better understand and validate these findings.
Collapse
Affiliation(s)
- Atieh Mirzababaei
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Azam Mohamadi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahya Mehri Hajmir
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Sara Ebrahimi
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Zeinab Zarrinvafa
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elham Kazemian
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Amir Mehrvar
- Department of Orthopedics, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Campos-Bayardo TI, Román-Rojas D, García-Sánchez A, Cardona-Muñoz EG, Sánchez-Lozano DI, Totsuka-Sutto S, Gómez-Hermosillo LF, Casillas-Moreno J, Andrade-Sierra J, Pazarín-Villaseñor L, Campos-Pérez W, Martínez-López E, Miranda-Díaz AG. The Role of TLRs in Obesity and Its Related Metabolic Disorders. Int J Mol Sci 2025; 26:2229. [PMID: 40076851 PMCID: PMC11900219 DOI: 10.3390/ijms26052229] [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: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Obesity affects the adaptability of adipose tissue (AT), impairing its ability to regulate energy and metabolism. Obesity is associated with many metabolic disorders, including dyslipidemia, hypertension, sleep disorders, non-alcoholic liver disease, and some types of cancer. Toll-like receptors (TLRs) are important in obesity and related metabolic disorders. TLRs are pattern-recognizing receptors (PRRs) involved in the innate immune system and recognize pathogen-associated molecular patterns (PAMPs) and endogenous ligands. TLRs, especially TLR2 and TLR4, are activated by fatty acids, endotoxins, and other ligands. TLR2 and TLR4 activation triggers inflammatory responses. Chronic inflammation driven by TLR activation is a hallmark of obesity and metabolic diseases. The inflammatory response triggered by TLR activation alters insulin signaling, contributing to insulin resistance, a key feature of metabolic syndrome and type 2 diabetes. Modulation of TLR activity through lifestyle changes (diet and exercise), obesity surgery, and pharmacological agents is under study as a possible therapeutic approach to controlling obesity and its complications.
Collapse
Affiliation(s)
- Tannia Isabel Campos-Bayardo
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Daniel Román-Rojas
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Andrés García-Sánchez
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Ernesto Germán Cardona-Muñoz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Daniela Itzel Sánchez-Lozano
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Sylvia Totsuka-Sutto
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Luis Francisco Gómez-Hermosillo
- Department of Laparoscopic Surgery, Hospital Civil de Guadalajara, “Juan I Menchaca”, Guadalajara 44360, Jalisco, Mexico; (L.F.G.-H.); (J.C.-M.)
| | - Jorge Casillas-Moreno
- Department of Laparoscopic Surgery, Hospital Civil de Guadalajara, “Juan I Menchaca”, Guadalajara 44360, Jalisco, Mexico; (L.F.G.-H.); (J.C.-M.)
| | - Jorge Andrade-Sierra
- Department of Nephrology, National Medical Center of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico; (J.A.-S.); (L.P.-V.)
| | - Leonardo Pazarín-Villaseñor
- Department of Nephrology, National Medical Center of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico; (J.A.-S.); (L.P.-V.)
| | - Wendy Campos-Pérez
- Department of Molecular Biology and Genomics, Institute of Nutrigenetics and Translational Nutrigenomics, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (W.C.-P.); (E.M.-L.)
| | - Erika Martínez-López
- Department of Molecular Biology and Genomics, Institute of Nutrigenetics and Translational Nutrigenomics, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (W.C.-P.); (E.M.-L.)
| | - Alejandra Guillermina Miranda-Díaz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| |
Collapse
|
6
|
Escamilla-Ruiz M, Zarzoza-Medina MG, Ríos-Ramírez M, Hernández-Adame PL, Ruiz-García J. Spontaneous Formation of Micelles and Vesicles in Langmuir Monolayers of Heneicosanoic Acid. ACS OMEGA 2025; 10:4224-4232. [PMID: 39959046 PMCID: PMC11822483 DOI: 10.1021/acsomega.4c03100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/27/2024] [Accepted: 11/06/2024] [Indexed: 02/18/2025]
Abstract
In Langmuir monolayers of heneicosanoic acid (C21H42O2), at low temperature, in the L'2 and CS crystalline phases, a blinking phenomenon occurs at the same positions of the monolayer, which is called localized oscillations (LO), but its origin has not been clarified. In this study, the LO phenomenon was correlated with the ejection of material out of the monolayer which was analyzed to understand this phenomenon. The techniques used for this purpose were pressure-area isotherms on a Langmuir balance and simultaneous observation of the monolayer by Brewster angle microscopy (BAM). Subsequently, using the Langmuir-Blodgett technique, the monolayers were transferred using freshly cleaved mica substrates for analysis by atomic force microscopy (AFM). Our results showed that the origin of the LO is related to a spontaneous formation of micelles and vesicles, since in AFM images these structures were observed in a size range from 4 to 16 nm. In addition, the AFM images showed that the difference between the heights of the L'2 and CS crystalline phases ranges from 13 to 15 Å.
Collapse
Affiliation(s)
- Martha
I. Escamilla-Ruiz
- Laboratorio de Física
Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí 78290, México
| | - Moises G. Zarzoza-Medina
- Laboratorio de Física
Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí 78290, México
| | - Maricarmen Ríos-Ramírez
- Laboratorio de Física
Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí 78290, México
| | - Pablo L. Hernández-Adame
- Laboratorio de Física
Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí 78290, México
| | - Jaime Ruiz-García
- Laboratorio de Física
Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí 78290, México
| |
Collapse
|
7
|
Bitterlich LM, Tunstead C, Hogan AE, Ankrum JA, English K. Mesenchymal stromal cells can block palmitate training of macrophages via cyclooxygenase-2 and interleukin-1 receptor antagonist. Cytotherapy 2025; 27:169-180. [PMID: 39580716 DOI: 10.1016/j.jcyt.2024.10.011] [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: 06/26/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
Innate training of macrophages can be beneficial for the clearance of pathogens. However, for certain chronic conditions, innate training can have detrimental effects due to an excessive production of pro-inflammatory cytokines. Obesity is a condition that is associated with a range of increased pro-inflammatory training stimuli including the free fatty acid palmitate. Mesenchymal stromal cells (MSCs) are powerful immunomodulators and known to suppress inflammatory macrophages via a range of soluble factors. We show that palmitate training of murine bone-marrow-derived macrophages and human monocyte-derived macrophages (MDMs) results in an increased production of TNFα and IL-6 upon stimulation with lipopolysaccharide and is associated with epigenetic remodeling. Palmitate training led to metabolic changes, however, MSCs did not alter the metabolic profile of human MDMs. Using a transwell system, we demonstrated that human bone marrow MSCs block palmitate training in both murine and human macrophages suggesting the involvement of secreted factors. MSC disruption of the training process occurs through more than one pathway. Suppression of palmitate-enhanced TNFα production is associated with cyclooxygenase-2 activity in MSCs, while secretion of interleukin-1 receptor antagonist by MSCs is required to suppress palmitate-enhanced IL-6 production in MDMs.
Collapse
Affiliation(s)
- Laura M Bitterlich
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland
| | - Courteney Tunstead
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland
| | - Andrew E Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland
| | - James A Ankrum
- University of Iowa Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA; Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Karen English
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Maynooth University, Maynooth, Ireland.
| |
Collapse
|
8
|
Tang T, Chen B, Hu J, Fan H, Zhang Z, Zhai T, Li C, Wang D, Xue W, Pei L, Chen F, Mi B, Zhao Y. The Association Between Egg and Egg-Derived Cholesterol Consumption, and Their Change Trajectories, with Obesity Among Chinese Adults: Results from the China Health and Nutrition Survey. Nutrients 2025; 17:333. [PMID: 39861463 PMCID: PMC11767974 DOI: 10.3390/nu17020333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: As a widely consumed, nutritious, and affordable food, eggs and their derivatives' impacts on obesity remain inconclusive. In this study, we aimed to determine the association between egg and egg-derived cholesterol consumption, and their change trajectories, with obesity among Chinese adults. Methods: Longitudinal data collected by the China Health and Nutrition Survey from 1997 to 2015 were analyzed. The latent growth mixture model was used to identify eggs and egg-derived cholesterol consumption trajectories. Cox proportional hazard models with shared frailty were used to analyze the association between egg and egg-derived cholesterol consumption, and their change trajectories, with obesity. Results: Data from 10,971 and 9483 participants aged ≥18 years old were used for the analyses of general obesity and central obesity, respectively. Compared to participants with an average egg intake of 0.1-50.0 g/d during the follow-up period, adults who never consumed eggs or those with an average egg intake of 50.1-100.0 g/d and >100.0 g/d had a higher risk of general obesity, with hazard ratios (HRs) and 95% confidence intervals (CIs) of 1.31 (1.08, 1.58), 1.30 (1.07, 1.60), and 1.98 (1.17, 3.35), respectively, and had a higher risk of central obesity, with HRs (95% CIs) of 1.17 (1.04, 1.31), 1.31 (1.14, 1.50), and 1.64 (1.15, 2.36), respectively. Participants with a "Baseline Low-Significant Rising Pattern" or a "Baseline High-Rising then Falling Pattern" of egg consumption trajectories during the follow-up period had a higher risk of general obesity, with HRs (95% CIs) of 1.56 (1.25, 1.93) and 1.38 (1.13, 1.69), respectively, and central obesity, with HRs (95% CIs) of 1.47 (1.29, 1.68) and 1.52 (1.34, 1.72), respectively. Compared to the second quartile (Q2) group of the average egg-derived cholesterol intake during the follow-up period, Q1, Q3, and Q4 groups had a higher risk of general obesity, with HRs (95% CIs) of 1.28 (1.06,1.54), 1.21 (1.02, 1.44), and 1.43 (1.19, 1.71), respectively, and a higher risk of central obesity, with HRs (95% CIs) of 1.20 (1.08, 1.33), 1.11 (1.01, 1.23), and 1.32 (1.19, 1.46), respectively. Participants with a "Baseline Low-Significant Rising Pattern" or with a "Baseline High-Rising then Falling Pattern" of egg-derived cholesterol consumption during the follow-up period had a higher risk of general obesity, with HRs (95% CIs) of 1.54 (1.25, 1.92) and 1.37 (1.15, 1.64), respectively, and a higher risk of central obesity, with HRs (95% CIs) of 1.46 (1.28, 1.68) and 1.47 (1.32, 1.64), respectively. Conclusions: Both the insufficient and excessive intake of eggs and egg-derived cholesterol tended to be associated with a higher risk of general and central obesity. Suddenly increasing or consistently high levels of egg and egg-derived cholesterol intake seemed to be associated with a higher risk of obesity. To prevent obesity, people should consume a moderate amount of eggs and egg-derived cholesterol.
Collapse
Affiliation(s)
- Tianhui Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Binghua Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Jiahao Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Hangzhao Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Zilan Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Tianyang Zhai
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Chunxiao Li
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Wanli Xue
- Department of Nutrition and Food Safety, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Leilei Pei
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Fangyao Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Baibing Mi
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| | - Yaling Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Center, No. 76, Yanta West Road, Xi’an 710061, China
| |
Collapse
|
9
|
Yang J, Wu Y, Jiang Z, Jiao L, Wang Y. Fecal fatty acid profile reveals the therapeutic effect of red ginseng acidic polysaccharide on type 2 diabetes mellitus in rats. J Food Sci 2025; 90:e70015. [PMID: 39832227 DOI: 10.1111/1750-3841.70015] [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: 10/12/2024] [Revised: 12/13/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
This study aimed to investigate the potential hypoglycemic mechanism of red ginseng acidic polysaccharides (RGAP) from the perspective of fatty acid (FA) regulation. A high-glucose/high-fat diet in conjunction with streptozotocin administration was employed to establish type 2 diabetes mellitus (T2DM) rat models, and their fecal FAs were detected using the liquid chromatography-mass spectrometry (LC-MS) method. RGAP treatment alleviated the polyphagia, polydipsia, weight loss, and hyperglycemia observed in T2DM rats. FA profile was disturbed by T2DM modeling, and 11 marker FAs were selected from statistical analysis, whose intensities were reversely changed by RGAP administration. Among these marker FAs, short-chain FAs were negatively correlated with the fasting blood glucose (FBG) level, while positive correlations were observed between long-chain FA and the FBG level. Combined with the metabolite-enzyme-gene network analysis, we inferred that the mechanistic mechanism RGAP on T2DM may be associated with the regulation of FA metabolism and inflammation-related signaling pathways. This study confirmed the regulatory effect of RGAP on fecal FA, which can provide a scientific basis and new ideas for developing red ginseng as a functional food for supplementary treatment of T2DM.
Collapse
Affiliation(s)
- Jingxuan Yang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Yi Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Ziye Jiang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Lili Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
10
|
Rohun J, Dudzik D, Raczak-Gutknecht J, Wabich E, Młodziński K, Markuszewski MJ, Daniłowicz-Szymanowicz L. Metabolomics in Atrial Fibrillation: Unlocking Novel Biomarkers and Pathways for Diagnosis, Prognosis, and Personalized Treatment. J Clin Med 2024; 14:34. [PMID: 39797116 PMCID: PMC11722095 DOI: 10.3390/jcm14010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Atrial fibrillation (AF) is the most frequent arrhythmia in the adult population associated with a high rate of severe consequences leading to significant morbidity and mortality worldwide. Therefore, its prompt recognition is of high clinical importance. AF detection often remains challenging due to unspecific symptoms and a lack of reliable biomarkers for its prediction. Herein, novel bioanalytical methodologies, such as metabolomics, offer new opportunities for a better understanding of the underlying pathological mechanisms of cardiovascular diseases, including AF. The metabolome, considered a complete set of small molecules present in the organism, directly reflects the current phenotype of the studied system and is highly sensitive to any changes, including arrhythmia's onset. A growing body of evidence suggests that metabolite profiling has prognostic value in AF prediction, highlighting its potential role not only in early diagnosis but also in guiding therapeutic interventions. By identifying specific metabolites as a disease biomarker or recognising particular metabolomic pathways involved in the AF pathomechanisms, metabolomics could be of great clinical value for further clinical decision-making, risk stratification, and an individual personalised approach. The presented narrative review aims to summarise the current state of knowledge on metabolomics in AF with a special emphasis on its implications for clinical practice and personalised medicine.
Collapse
Affiliation(s)
- Justyna Rohun
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Danuta Dudzik
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Elżbieta Wabich
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Krzysztof Młodziński
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Michał J. Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Ludmiła Daniłowicz-Szymanowicz
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| |
Collapse
|
11
|
Méndez López LF, González Llerena JL, Vázquez Rodríguez JA, Medellín Guerrero AB, González Martínez BE, Solís Pérez E, López-Cabanillas Lomelí M. Dietary Modulation of the Immune System. Nutrients 2024; 16:4363. [PMID: 39770983 PMCID: PMC11676904 DOI: 10.3390/nu16244363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Recent insights into the influence of nutrition on immune system components have driven the development of dietary strategies targeting the prevention and management of major metabolic-inflammatory diseases. This review summarizes the bidirectional relationship between nutrition and immunocompetence, beginning with an overview of immune system components and their functions. It examines the effects of nutritional status, dietary patterns, and food bioactives on systemic inflammation, immune cell populations, and lymphoid tissues, as well as their associations with infectious and chronic disease pathogenesis. The mechanisms by which key nutrients influence immune constituents are delineated, focusing on vitamins A, D, E, C, and B, as well as minerals including zinc, iron, and selenium. Also highlighted are the immunomodulatory effects of polyunsaturated fatty acids as well as bioactive phenolic compounds and probiotics, given their expanding relevance. Each section addresses the implications of nutritional and nutraceutical interventions involving these nutrients within the broader context of major infectious, metabolic, and inflammatory diseases. This review further underscores that, while targeted nutrient supplementation can effectively restore immune function to optimal levels, caution is necessary in certain cases, as it may increase morbidity in specific diseases. In other instances, dietary counseling should be integrated to ensure that therapeutic goals are achieved safely and effectively.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Manuel López-Cabanillas Lomelí
- Universidad Autónoma de Nuevo León, Facultad de Salud Pública y Nutrición, Centro de Investigación en Nutrición y Salud Pública, Monterrey 64460, México; (L.F.M.L.)
| |
Collapse
|
12
|
Chen R, An W, Liu X, Yan J, Huang Y, Zhang J. Risk factors of allergic rhinitis and its prevention strategies. FRONTIERS IN ALLERGY 2024; 5:1509552. [PMID: 39665077 PMCID: PMC11632107 DOI: 10.3389/falgy.2024.1509552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
Allergic rhinitis (AR) is a global disease with high prevalence. It reduces the patient's quality of life seriously. The health care and management of AR was also a heavy social burden. Specific immunotherapy (SIT) is the only curative treatment for AR that may alter the natural course of this disease. However, acceptance and compliance of SIT in AR patients are still not high and many patients are not effectively controlled. Disease prevention based on known risk factors is much more cost-effective compared to post-diagnosis treatment. There have been some reports on the risk factors of AR up to now, but the information is fragmented. This review systemically clarified the risk factors of AR including hereditary factors and family history, maternal situation & mode of delivery and feeding, personal characteristics, nutrition and food intake, personal behavior and habits, acquired environmental and chemical exposure, diseases and health status. The preventive strategies were also proposed briefly. This review was hopeful to improve people's awareness of the risk factors of AR and put forward AR prevention.
Collapse
Affiliation(s)
- Ruzhi Chen
- Guangdong ProvincialKey Laboratory of Allergy & Clinical Immunology, Department of Allergy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei An
- Department of Nuclear Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xueting Liu
- Guangdong ProvincialKey Laboratory of Allergy & Clinical Immunology, Department of Allergy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Yan
- Guangdong ProvincialKey Laboratory of Allergy & Clinical Immunology, Department of Allergy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuyi Huang
- Guangdong ProvincialKey Laboratory of Allergy & Clinical Immunology, Department of Allergy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junyan Zhang
- Guangdong ProvincialKey Laboratory of Allergy & Clinical Immunology, Department of Allergy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
13
|
Wang K, Nie Y, Maguire C, Syphurs C, Sheen H, Karoly M, Lapp L, Gygi JP, Jayavelu ND, Patel RK, Hoch A, Corry D, Kheradmand F, McComsey GA, Fernandez-Sesma A, Simon V, Metcalf JP, Higuita NIA, Messer WB, Davis MM, Nadeau KC, Kraft M, Bime C, Schaenman J, Erle D, Calfee CS, Atkinson MA, Brackenridge SC, Hafler DA, Shaw A, Rahman A, Hough CL, Geng LN, Ozonoff A, Haddad EK, Reed EF, van Bakel H, Kim-Schultz S, Krammer F, Wilson M, Eckalbar W, Bosinger S, Langelier CR, Sekaly RP, Montgomery RR, Maecker HT, Krumholz H, Melamed E, Steen H, Pulendran B, Augustine AD, Cairns CB, Rouphael N, Becker PM, Fourati S, Shannon CP, Smolen KK, Peters B, Kleinstein SH, Levy O, Altman MC, Iwasaki A, Diray-Arce J, Ehrlich LIR, Guan L. Unraveling SARS-CoV-2 Host-Response Heterogeneity through Longitudinal Molecular Subtyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624784. [PMID: 39651165 PMCID: PMC11623532 DOI: 10.1101/2024.11.22.624784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Hospitalized COVID-19 patients exhibit diverse immune responses during acute infection, which are associated with a wide range of clinical outcomes. However, understanding these immune heterogeneities and their links to various clinical complications, especially long COVID, remains a challenge. In this study, we performed unsupervised subtyping of longitudinal multi-omics immunophenotyping in over 1,000 hospitalized patients, identifying two critical subtypes linked to mortality or mechanical ventilation with prolonged hospital stay and three severe subtypes associated with timely acute recovery. We confirmed that unresolved systemic inflammation and T-cell dysfunctions were hallmarks of increased severity and further distinguished patients with similar acute respiratory severity by their distinct immune profiles, which correlated with differences in demographic and clinical complications. Notably, one critical subtype (SubF) was uniquely characterized by early excessive inflammation, insufficient anticoagulation, and fatty acid dysregulation, alongside higher incidences of hematologic, cardiac, and renal complications, and an elevated risk of long COVID. Among the severe subtypes, significant differences in viral clearance and early antiviral responses were observed, with one subtype (SubC) showing strong early T-cell cytotoxicity but a poor humoral response, slower viral clearance, and greater risks of chronic organ dysfunction and long COVID. These findings provide crucial insights into the complex and context-dependent nature of COVID-19 immune responses, highlighting the importance of personalized therapeutic strategies to improve both acute and long-term outcomes.
Collapse
|
14
|
Chen L, Tan KML, Xu J, Mishra P, Mir SA, Gong M, Narasimhan K, Ng B, Lai JS, Tint MT, Cai S, Sadananthan SA, Michael N, Yaligar J, Velan SS, Leow MKS, Tan KH, Chan J, Meaney MJ, Chan SY, Chong YS, Eriksson JG. Exploring multi-omics and clinical characteristics linked to accelerated biological aging in Asian women of reproductive age: insights from the S-PRESTO study. Genome Med 2024; 16:128. [PMID: 39516835 PMCID: PMC11549770 DOI: 10.1186/s13073-024-01403-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Phenotypic age (PhenoAge), a widely used marker of biological aging, has been shown to be a robust predictor of all-cause mortality and morbidity in different populations. Existing studies on biological aging have primarily focused on individual domains, resulting in a lack of a comprehensive understanding of the multi-systemic dysregulation that occurs in aging. METHODS PhenoAge was evaluated based on a linear combination of chronological age (CA) and 9 clinical biomarkers in 952 multi-ethnic Asian women of reproductive age. Phenotypic age acceleration (PhenoAgeAccel), an aging biomarker, represents PhenoAge after adjusting for CA. This study conducts an in-depth association analysis of PhenoAgeAccel with clinical, nutritional, lipidomic, gut microbiome, and genetic factors. RESULTS Higher adiposity, glycaemia, plasma saturated fatty acids, kynurenine pathway metabolites, GlycA, riboflavin, nicotinamide, and insulin-like growth factor binding proteins were positively associated with PhenoAgeAccel. Conversely, a healthier diet and higher levels of pyridoxal phosphate, all-trans retinol, betaine, tryptophan, glutamine, histidine, apolipoprotein B, and insulin-like growth factors were inversely associated with PhenoAgeAccel. Lipidomic analysis found 132 lipid species linked to PhenoAgeAccel, with PC(O-36:0) showing the strongest positive association and CE(24:5) demonstrating the strongest inverse association. A genome-wide association study identified rs9864994 as the top genetic variant (P = 5.69E-07) from the ZDHHC19 gene. Gut microbiome analysis revealed that Erysipelotrichaceae UCG-003 and Bacteroides vulgatus were inversely associated with PhenoAgeAccel. Integrative network analysis of aging-related factors underscored the intricate links among clinical, nutritional and lipidomic variables, such as positive associations between kynurenine pathway metabolites, amino acids, adiposity, and insulin resistance. Furthermore, potential mediation effects of blood biomarkers related to inflammation, immune response, and nutritional and energy metabolism were observed in the associations of diet, adiposity, genetic variants, and gut microbial species with PhenoAgeAccel. CONCLUSIONS Our findings provide a comprehensive analysis of aging-related factors across multiple platforms, delineating their complex interconnections. This study is the first to report novel signatures in lipidomics, gut microbiome and blood biomarkers specifically associated with PhenoAgeAccel. These insights are invaluable in understanding the molecular and metabolic mechanisms underlying biological aging and shed light on potential interventions to mitigate accelerated biological aging by targeting modifiable factors.
Collapse
Affiliation(s)
- Li Chen
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Karen Mei-Ling Tan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore.
| | - Jia Xu
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Priti Mishra
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sartaj Ahmad Mir
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Min Gong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kothandaraman Narasimhan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bryan Ng
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jun Shi Lai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mya Thway Tint
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shirong Cai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suresh Anand Sadananthan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Navin Michael
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jadegoud Yaligar
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sambasivam Sendhil Velan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Medicine and Human Potential Translational Research Programme. Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Melvin Khee Shing Leow
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Jerry Chan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Michael J Meaney
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Sackler Program for Epigenetics & Psychobiology, McGill University, Montréal, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Shiao-Yng Chan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhalsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
Dassoff ES, Hamad S, Campagna E, Thilakarathna SH, Michalski MC, Wright AJ. Influence of Emulsion Lipid Droplet Crystallinity on Postprandial Endotoxin Transporters and Atherogenic And Inflammatory Profiles in Healthy Men - A Randomized Double-Blind Crossover Acute Meal Study. Mol Nutr Food Res 2024; 68:e2400365. [PMID: 39388527 DOI: 10.1002/mnfr.202400365] [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: 05/21/2024] [Revised: 08/15/2024] [Indexed: 10/12/2024]
Abstract
SCOPE Consumption of high-fat meals is associated with increased endotoxemia, inflammation, and atherogenic profiles, with repeated postprandial responses suggested as contributors to chronically elevated risk factors. However, effects of lipid solid versus liquid state specifically have not been investigated. METHODS AND RESULTS This exploratory randomized crossover study tests the impact of lipid crystallinity on plasma levels of endotoxin transporters (lipopolysaccharide [LPS] binding protein [LBP] and soluble cluster of differentiation 14 [sCD14]) and select proinflammatory and atherogenic markers (tumor necrosis factor-alpha [TNF-α], C-reactive protein [CRP], interleukin-1-beta [IL-1β], interferon-gamma [IFN-γ], interleukin-6 [IL-6], soluble intercellular adhesion molecule [sICAM], soluble vascular cell adhesion molecule [sVCAM], monocyte chemoattractant protein-1 [MCP-1/CCL2], plasminogen activator inhibitor-1 [PAI-1], and fibrinogen). Fasted healthy men (n = 14, 28 ± 5.5 years, 24.1 ± 2.6 kg m-2) consumed two 50 g palm stearin oil-in-water emulsions tempered to contain either liquid or crystalline lipid droplets at 37 °C on separate occasions with blood sampling at 0, 2-, 4-, and 6-h post-meal. Timepoint data, area under the curve, and peak concentration values are compared. Overall, no treatment effects are seen (p > 0.05). There are significant effects of time, with values decreasing from baseline, for TNF-α, MCP-1/CCL2, PAI-1, and fibrinogen (p < 0.05). CONCLUSION Responder analysis pointed to differential treatment effects associated with some participant baseline characteristics but, overall, palm-stearin emulsion droplet crystallinity does not acutely affect plasma endotoxin transporters nor select inflammatory and atherogenic markers.
Collapse
Affiliation(s)
- Erik S Dassoff
- Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Samar Hamad
- Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Elaina Campagna
- Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Surangi H Thilakarathna
- Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Marie-Caroline Michalski
- INRAE, CarMeN Laboratory, Inserm, Univ-Lyon, Université Claude Bernard Lyon, Centre de Recherche en Nutrition Humain Rhône-Alpes, Pierre Bénite, France
| | - Amanda J Wright
- Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
16
|
Zhuo B, Ran S, Qian AM, Zhang J, Tabet M, Howard SW, Zhang Z, Tian F, Lin H. Air Pollution Metabolomic Signatures and Chronic Respiratory Diseases Risk: A Longitudinal Study. Chest 2024; 166:975-986. [PMID: 39059576 DOI: 10.1016/j.chest.2024.06.3809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Although evidence has documented the associations of ambient air pollution with chronic respiratory diseases (CRDs) and lung function, the underlying metabolic mechanisms remain largely unclear. RESEARCH QUESTION How does the metabolomic signature for air pollution relate to CRD risk, respiratory symptoms, and lung function? STUDY DESIGN AND METHODS We retrieved 171,132 participants free of COPD and asthma at baseline from the UK Biobank, who had data on air pollution and metabolomics. Exposures to air pollutants (particulate matter with diameter ≤ 2.5 μm [PM2.5], particulate matter with a diameter ≤ 10 μm, nitrogen oxide [NOX], and NO2) were assessed for 4 years before baseline considering residential address histories. We used 10-fold cross-validation elastic net regression to identify air pollution-associated metabolites. Multivariable Cox models were used to assess the associations between metabolomic signatures and CRD risk. Mediation and pathway analysis were conducted to explore the metabolic mechanism underlying the associations. RESULTS During a median follow-up of 12.51 years, 8,951 and 5,980 incident COPD and asthma cases were recorded. In multivariable Cox regressions, air pollution was positively associated with CRD risk (eg, hazard ratio per interquartile range increment in PM2.5, 1.09; 95% CI, 1.06-1.13). We identified 103, 86, 85, and 90 metabolites in response to PM2.5, particulate matter with a diameter ≤ 10 μm, NOX, and NO2 exposure, respectively. The metabolomic signatures showed significant associations with CRD risk (hazard ratio per SD increment in PM2.5 metabolomic signature, 1.11; 95% CI, 1.09-1.14). Mediation analysis showed that peripheral inflammatory and erythrocyte-related markers mediated the effects of metabolomic signatures on CRD risk. We identified 14 and 12 perturbed metabolic pathways (energy metabolism and amino acid metabolism pathways, etc) for PM2.5 and NOX metabolomic signatures. INTERPRETATION Our study identifies metabolomic signatures for air pollution exposure. The metabolomic signatures showed significant associations with CRD risk, and inflammatory- and erythrocyte-related markers partly mediated the metabolomic signatures-CRD links.
Collapse
Affiliation(s)
- Bingting Zhuo
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shanshan Ran
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Aaron M Qian
- Department of Psychology, College of Arts and Sciences, Saint Louis University, Saint Louis, MO
| | - Junguo Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Maya Tabet
- College of Global Population Health, University of Health Sciences and Pharmacy, Saint Louis, MO
| | - Steven W Howard
- Department of Health Services Administration, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL
| | - Zilong Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fei Tian
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
17
|
Stanek E, Czamara K, Kaczor A. Increased obesogenic action of palmitic acid during early stage of adipogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159525. [PMID: 38876269 DOI: 10.1016/j.bbalip.2024.159525] [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: 01/27/2024] [Revised: 04/18/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
The functional differences between preadipocytes and fully differentiated mature adipocytes derived from stromal vascular fraction stem cells, as well as primary adipocytes have been analysed by evaluating their response to the obesogenic factor (a saturated fatty acid) and TNF-triggered inflammation. The analysis of single adipocytes shows that the saturated fatty acid (palmitic acid) accumulation is accompanied by inflammation and considerably dependent on the stage of the adipogenesis. In particular, preadipocytes show the exceptional potential for palmitic acid uptake resulting in their hypertrophy and the elevated cellular expression of the inflammation marker (ICAM-1). Our research provides new information on the impact of obesogenic factors on preadipocytes that is important in the light of childhood obesity prevention.
Collapse
Affiliation(s)
- Ewa Stanek
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, 11 Lojasiewicza Str., 30-348 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Krzysztof Czamara
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), 14 Bobrzynskiego Str., 30-348 Krakow, Poland.
| | - Agnieszka Kaczor
- Jagiellonian University, Faculty of Chemistry, 2 Gronostajowa Str., 30-387 Krakow, Poland.
| |
Collapse
|
18
|
Rajamanickam V, Desouza CV, Castillo RT, Saraswathi V. Blocking Thromboxane-Prostanoid Receptor Signaling Attenuates Lipopolysaccharide- and Stearic Acid-Induced Inflammatory Response in Human PBMCs. Cells 2024; 13:1320. [PMID: 39195211 PMCID: PMC11352481 DOI: 10.3390/cells13161320] [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: 07/05/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Inflammation is implicated in the etiology of obesity-related diseases. Thromboxane-prostanoid receptor (TPR) is known to play a role in mediating an inflammatory response in a variety of cells. Gut-derived lipopolysaccharide (LPS), a TLR4 agonist, is elevated in obesity. Moreover, free fatty acids (FFAs) are important mediators of obesity-related inflammation. However, the role and mechanisms by which TPR regulates the inflammatory response in human immune cells remain unclear. We sought to determine the link between TPR and obesity and the role/mechanisms by which TPR alters LPS- or stearic acid (SA)-induced inflammatory responses in PBMCs. Cells were pre-treated with agents blocking TPR signaling, followed by treatment with LPS or stearic acid (SA). Our findings showed that TPR mRNA levels are higher in PBMCs from individuals with obesity. Blockade of TPR as well as ROCK, which acts downstream of TPR, attenuated LPS- and/or SA-induced pro-inflammatory responses. On the other hand, TPR activation using its agonist enhanced the pro-inflammatory effects of LPS and/or SA. Of note, the TPR agonist by itself elicits an inflammatory response, which was attenuated by blocking TPR or ROCK. Our data suggest that TPR plays a key role in promoting an inflammatory response in human PBMCs, and this effect is mediated via TLR4 and/or ROCK signaling.
Collapse
Affiliation(s)
- Vinothkumar Rajamanickam
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.R.); (C.V.D.); (R.T.C.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Cyrus V. Desouza
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.R.); (C.V.D.); (R.T.C.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Romilia T. Castillo
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.R.); (C.V.D.); (R.T.C.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Viswanathan Saraswathi
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.R.); (C.V.D.); (R.T.C.)
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
19
|
Vieira LG, de Noronha SISR, Chírico MTT, de Souza AB, de Matos NA, Chianca-Jr DA, Bezerra FS, de Menezes RC. The impact of high-fat diet consumption and inulin fiber supplementation on anxiety-related behaviors and liver oxidative status in female Wistar rats. Behav Brain Res 2024; 470:115048. [PMID: 38761857 DOI: 10.1016/j.bbr.2024.115048] [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: 02/13/2024] [Revised: 05/04/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Obesity is a worldwide public health problem associated with cognitive and mental health problems in both humans and rats. Studies assessing the effect of fiber supplementation on behavioral deficits and oxidative stress caused by high-fat diet (HFD) consumption in female rats are still scarce. We hypothesized that HFD consumption would lead to anxiety-related behavior and hepatic oxidative stress and that inulin would protect against these changes. We analyzed the impact of HFD-induced obesity combined with fiber supplementation (inulin) on anxiety-related defensive behavior and hepatic oxidative stress. RESULTS Female rats were fed a high-fat diet (HFD; 45%) for nine weeks to induce obesity. The administration of inulin was found to decrease the adiposity index in both the control and obese groups. The consumption of a HFD combined with inulin supplementation resulted in a reduction in both CAT activity and carbonylated protein levels, leading to a shift in the hepatic redox balance. Interestingly, the behavioral data were conflicting. Specifically, animals that consumed a high-fat diet and received inulin showed signs of impaired learning and memory caused by obesity. The HFD did not impact anxiety-related behaviors in the female rats. However, inulin appears to have an anxiolytic effect, in the ETM, when associated with the HFD. On the other hand, inulin appears to have affected the locomotor activity in the HFD in both open field and light-dark box. CONCLUSION Our results show that consumption of a HFD induced obesity in female rats, similar to males. However, HFD consumption did not cause a consistent increase in anxiety-related behaviors in female Wistar rats. Treatment with inulin at the dosage used did not exert consistent changes on the behavior of the animals, but attenuated the abdominal WAT expansion and the hepatic redox imbalance elicited by high-fat diet-induced obesity.
Collapse
Affiliation(s)
- Lucas Gabriel Vieira
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| | | | - Máira Tereza Talma Chírico
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| | - Ana Beatriz de Souza
- Department of Biological Sciences, Laboratory of Experimental Pathophysiology, Federal University of Ouro Preto, MG 35400-000, Brazil.
| | - Natália Alves de Matos
- Department of Biological Sciences, Laboratory of Experimental Pathophysiology, Federal University of Ouro Preto, MG 35400-000, Brazil.
| | - Deoclécio Alves Chianca-Jr
- Department of Biological Sciences, Laboratory of Experimental Pathophysiology, Federal University of Ouro Preto, MG 35400-000, Brazil.
| | - Frank Silva Bezerra
- Department of Biological Sciences, Laboratory of Experimental Pathophysiology, Federal University of Ouro Preto, MG 35400-000, Brazil.
| | - Rodrigo Cunha de Menezes
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| |
Collapse
|
20
|
Khan MM, Khan ZA, Khan MA. Metabolic complications of psychotropic medications in psychiatric disorders: Emerging role of de novo lipogenesis and therapeutic consideration. World J Psychiatry 2024; 14:767-783. [PMID: 38984346 PMCID: PMC11230099 DOI: 10.5498/wjp.v14.i6.767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 05/05/2024] [Accepted: 05/23/2024] [Indexed: 06/19/2024] Open
Abstract
Although significant advances have been made in understanding the patho-physiology of psychiatric disorders (PDs), therapeutic advances have not been very convincing. While psychotropic medications can reduce classical symptoms in patients with PDs, their long-term use has been reported to induce or exaggerate various pre-existing metabolic abnormalities including diabetes, obesity and non-alcoholic fatty liver disease (NAFLD). The mechanism(s) underlying these metabolic abnormalities is not clear; however, lipid/fatty acid accumulation due to enhanced de novo lipogenesis (DNL) has been shown to reduce membrane fluidity, increase oxidative stress and inflammation leading to the development of the aforementioned metabolic abnormalities. Intriguingly, emerging evidence suggest that DNL dysregulation and fatty acid accumulation could be the major mechanisms associated with the development of obesity, diabetes and NAFLD after long-term treatment with psychotropic medications in patients with PDs. In support of this, several adjunctive drugs comprising of anti-oxidants and anti-inflammatory agents, that are used in treating PDs in combination with psychotropic medications, have been shown to reduce insulin resistance and development of NAFLD. In conclusion, the above evidence suggests that DNL could be a potential pathological factor associated with various metabolic abnormalities, and a new avenue for translational research and therapeutic drug designing in PDs.
Collapse
Affiliation(s)
- Mohammad M Khan
- Laboratory of Translational Neurology and Molecular Psychiatry, Department of Biotechnology, Era’s Lucknow Medical College and Hospital, and Faculty of Science, Era University, Lucknow 226003, India
| | - Zaw Ali Khan
- Era’s Lucknow Medical College and Hospital, Era University, Lucknow 226003, India
| | - Mohsin Ali Khan
- Era’s Lucknow Medical College and Hospital, Era University, Lucknow 226003, India
| |
Collapse
|
21
|
Kopczyńska J, Kowalczyk M. The potential of short-chain fatty acid epigenetic regulation in chronic low-grade inflammation and obesity. Front Immunol 2024; 15:1380476. [PMID: 38605957 PMCID: PMC11008232 DOI: 10.3389/fimmu.2024.1380476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Obesity and chronic low-grade inflammation, often occurring together, significantly contribute to severe metabolic and inflammatory conditions like type 2 diabetes (T2D), cardiovascular disease (CVD), and cancer. A key player is elevated levels of gut dysbiosis-associated lipopolysaccharide (LPS), which disrupts metabolic and immune signaling leading to metabolic endotoxemia, while short-chain fatty acids (SCFAs) beneficially regulate these processes during homeostasis. SCFAs not only safeguard the gut barrier but also exert metabolic and immunomodulatory effects via G protein-coupled receptor binding and epigenetic regulation. SCFAs are emerging as potential agents to counteract dysbiosis-induced epigenetic changes, specifically targeting metabolic and inflammatory genes through DNA methylation, histone acetylation, microRNAs (miRNAs), and long non-coding RNAs (lncRNAs). To assess whether SCFAs can effectively interrupt the detrimental cascade of obesity and inflammation, this review aims to provide a comprehensive overview of the current evidence for their clinical application. The review emphasizes factors influencing SCFA production, the intricate connections between metabolism, the immune system, and the gut microbiome, and the epigenetic mechanisms regulated by SCFAs that impact metabolism and the immune system.
Collapse
Affiliation(s)
- Julia Kopczyńska
- Laboratory of Lactic Acid Bacteria Biotechnology, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
22
|
Siewe N, Friedman A. A mathematical model of obesity-induced type 2 diabetes and efficacy of anti-diabetic weight reducing drug. J Theor Biol 2024; 581:111756. [PMID: 38307451 DOI: 10.1016/j.jtbi.2024.111756] [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: 08/17/2023] [Revised: 11/04/2023] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Abstract
The dominant paradigm for modeling the obesity-induced T2DM (type 2 diabetes mellitus) today focuses on glucose and insulin regulatory systems, diabetes pathways, and diagnostic test evaluations. The problem with this approach is that it is not possible to explicitly account for the glucose transport mechanism from the blood to the liver, where the glucose is stored, and from the liver to the blood. This makes it inaccurate, if not incorrect, to properly model the concentration of glucose in the blood in comparison to actual glycated hemoglobin (A1C) test results. In this paper, we develop a mathematical model of glucose dynamics by a system of ODEs. The model includes the mechanism of glucose transport from the blood to the liver, and from the liver to the blood, and explains how obesity is likely to lead to T2DM. We use the model to evaluate the efficacy of an anti-T2DM drug that also reduces weight.
Collapse
Affiliation(s)
- Nourridine Siewe
- School of Mathematics and Statistics, College of Science, Rochester Institute of Technology, Rochester, NY, USA.
| | - Avner Friedman
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
23
|
Godos J, Lanza G, Ferri R, Caraci F, Cano SS, Elio I, Micek A, Castellano S, Grosso G. Relation between dietary inflammatory potential and sleep features: Systematic review of observational studies. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2024; 17:1-14. [DOI: 10.3233/mnm-240008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
BACKGROUND: Diet and sleep represent key behavioral risk factors for major non-communicable diseases. Dietary factors may modulate systemic inflammation and potentially affect the human brain functionality, finally affecting the sleep-wake cycle. In this context, the Dietary Inflammatory Index (DII) has been studied as a tool to investigate the dietary inflammatory potential and its relationship with a variety of health conditions. OBJECTIVE: The aim of this study was to perform a systematic revision of observational studies on the inflammatory potential of the diet assessed through the DII and sleep features. METHODS: A systematic search of observational studies on DII and sleep features was conducted on Pubmed and EMBASE electronic databases. The Meta-analyses Of Observational Studies in Epidemiology (MOOSE) guidelines were used to plan the methodology. Studies identified through the search were selected according to inclusion/exclusion criteria. RESULTS: The systematic search and review led to the selection of 13 studies. Most studies including the general population reported an association between higher inflammatory potential of the diet and worse sleep quality and suboptimal sleep duration. Some studies also reported an association with daytime sleepiness and sleep apnea. However, some studies including younger individuals (i.e., college students or young employees) reported null findings. Four studies conducted in individuals with a variety of health conditions also showed some relation between DII and sleep features, although results were not fully consistent. CONCLUSIONS: Data published so far suggests an association between DII and sleep features. However, lack of randomized clinical trials and better designed observational studies, imply that further research on this matter is warranted to understand whether a causal relation could explain current findings.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
- Clinical Neurophysiology Research Unit, Oasi Research Institute-IRCCS, Troina, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute-IRCCS, Troina, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, Universityof Catania, Catania, Italy
- Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina, Italy
| | - Sandra Sumalla Cano
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, México
- Universidad de La Romana, LaRomana, República Dominicana
| | - Iñaki Elio
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, México
- Fundación Universitaria Internacional de Colombia, Bogotá, Colombia
| | - Agnieszka Micek
- Statistical Laboratory, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| |
Collapse
|
24
|
Sarkar A, Pawar SV, Chopra K, Jain M. Gamut of glycolytic enzymes in vascular smooth muscle cell proliferation: Implications for vascular proliferative diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167021. [PMID: 38216067 DOI: 10.1016/j.bbadis.2024.167021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the media of the blood vessels and are responsible for maintaining vascular tone. Emerging evidence confirms that VSMCs possess high plasticity. During vascular injury, VSMCs switch from a "contractile" phenotype to an extremely proliferative "synthetic" phenotype. The balance between both strongly affects the progression of vascular remodeling in many cardiovascular pathologies such as restenosis, atherosclerosis and aortic aneurism. Proliferating cells demand high energy requirements and to meet this necessity, alteration in cellular bioenergetics seems to be essential. Glycolysis, fatty acid metabolism, and amino acid metabolism act as a fuel for VSMC proliferation. Metabolic reprogramming of VSMCs is dynamically variable that involves multiple mechanisms and encompasses the coordination of various signaling molecules, proteins, and enzymes. Here, we systemically reviewed the metabolic changes together with the possible treatments that are still under investigation underlying VSMC plasticity which provides a promising direction for the treatment of diseases associated with VSMC proliferation. A better understanding of the interaction between metabolism with associated signaling may uncover additional targets for better therapeutic strategies in vascular disorders.
Collapse
Affiliation(s)
- Ankan Sarkar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manish Jain
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
25
|
Sirakawin C, Lin D, Zhou Z, Wang X, Kelleher R, Huang S, Long W, Pires‐daSilva A, Liu Y, Wang J, Vinnikov IA. SKN-1/NRF2 upregulation by vitamin A is conserved from nematodes to mammals and is critical for lifespan extension in Caenorhabditis elegans. Aging Cell 2024; 23:e14064. [PMID: 38100161 PMCID: PMC10928581 DOI: 10.1111/acel.14064] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 03/13/2024] Open
Abstract
Vitamin A (VA) is a micronutrient essential for the physiology of many organisms, but its role in longevity and age-related diseases remains unclear. In this work, we used Caenorhabditis elegans to study the impact of various bioactive compounds on lifespan. We demonstrate that VA extends lifespan and reduces lipofuscin and fat accumulation while increasing resistance to heat and oxidative stress. This resistance can be attributed to high levels of detoxifying enzymes called glutathione S-transferases, induced by the transcription factor skinhead-1 (SKN-1). Notably, VA upregulated the transcript levels of skn-1 or its mammalian ortholog NRF2 in both C. elegans, human cells, and liver tissues of mice. Moreover, the loss-of-function genetic models demonstrated a critical involvement of the SKN-1 pathway in longevity extension by VA. Our study thus provides novel insights into the molecular mechanism of anti-aging and anti-oxidative effects of VA, suggesting that this micronutrient could be used for the prevention and/or treatment of age-related disorders.
Collapse
Affiliation(s)
- Chaweewan Sirakawin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Dongfa Lin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory for Molecular Enzymology and Engineering, School of Life SciencesJilin UniversityChangchunChina
| | - Ziyue Zhou
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoxin Wang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | | | - Shangyuan Huang
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Weimiao Long
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | | | - Yu Liu
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Jingjing Wang
- Shanghai Key Laboratory of Pancreatic Diseases, Institute of Translational Medicine, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ilya A. Vinnikov
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
26
|
Molonia MS, Speciale A, Muscarà C, Salamone FL, Saija A, Cimino F. Low concentrations of α-lipoic acid reduce palmitic acid-induced alterations in murine hypertrophic adipocytes. Nat Prod Res 2024; 38:916-925. [PMID: 37129014 DOI: 10.1080/14786419.2023.2207137] [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: 01/27/2023] [Accepted: 04/14/2023] [Indexed: 05/03/2023]
Abstract
Obesity is a metabolic disorder with excessive body fat accumulation, increasing incidence of chronic metabolic diseases. Hypertrophic obesity is associated with local oxidative stress and inflammation. Herein, we evaluated the in vitro activity of micromolar concentrations of α-lipoic acid (ALA) on palmitic acid (PA)-exposed murine hypertrophic 3T3-L1 adipocytes, focussing on the main molecular pathways involved in adipogenesis, inflammation, and insulin resistance. ALA, starting from 1 µM, decreased adipocytes hypertrophy, reducing PA-triggered intracellular lipid accumulation, PPAR-γ levels, and FABP4 gene expression, and counteracted PA-induced intracellular ROS levels and NF-κB activation. ALA reverted PA-induced insulin resistance, restoring PI3K/Akt axis and inducing GLUT-1 and glucose uptake, showing insulin sensitizing properties since it increased their basal levels. In conclusion, this study supports the potential effects of low micromolar ALA against hypertrophy, inflammation, and insulin resistance in adipose tissue, suggesting its important role as pharmacological supplement in the prevention of conditions linked to obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Maria Sofia Molonia
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
- "Prof. Antonio Imbesi" Foundation, University of Messina,Messina, Italy
| | - Antonio Speciale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Claudia Muscarà
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Federica Lina Salamone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Antonella Saija
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Francesco Cimino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
27
|
Zhang C, Wang WS, Yao G, Zhu Y, Lin Y, Lu J, Sun K, Sun Y. Attenuation of palmitic acid-induced lysyl oxidase overexpression in the ovary contributes to the improvement of ovulation in obesity by metformin. Hum Reprod Open 2024; 2024:hoae002. [PMID: 38333108 PMCID: PMC10850847 DOI: 10.1093/hropen/hoae002] [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: 05/19/2023] [Revised: 12/20/2023] [Indexed: 02/10/2024] Open
Abstract
STUDY QUESTION Does palmitic acid (PA), the most common saturated free fatty acid (FFA) in individuals with obesity, contribute to anovulation through upregulation of the collagen-crosslinking enzyme lysyl oxidase (LOX) in the ovary? SUMMARY ANSWER Increased PA in individuals with obesity can cause LOX upregulation via the activation of hypoxia-inducible factor-1α (HIF-1α), resulting in abnormal collagen deposition in the ovary and anovulation, which can be ameliorated by metformin therapy. WHAT IS KNOWN ALREADY The underlying cause of anovulation in individuals with obesity is poorly defined, and accumulating evidence indicates that hormonal disturbance, insulin resistance, and inflammation may all play a role in the development of ovulation disorders in individuals with obesity. However, it remains to be determined whether PA plays a role in the regulation of LOX expression, thus disrupting ovarian extracellular matrix (ECM) remodelling in the ovary and resulting in impaired ovulation in individuals with obesity. STUDY DESIGN SIZE DURATION PA concentration and LOX protein abundance and activity in follicular fluid and ovarian tissue were compared between control (n = 21) subjects, patients with obesity with ovulation (n = 22), and patients with obesity with anovulation (n = 16). The effect of PA on LOX protein expression, and the underlying mechanism, was examined in primary human granulosa cells in vitro. The improvements in obesity conditions induced by LOX inhibition combined with metformin were investigated in a high-fat diet-induced obese rat model. PARTICIPANTS/MATERIALS SETTING METHODS The abundance of PA concentration and LOX activity was measured via a LOX activity assay and ELISA, respectively. The effect of PA on LOX protein expression was examined in the presence or absence of inhibitors of signalling molecules and siRNA-mediated knockdown of the putative transcription factor. Chromatin immunoprecipitation assays were subsequently conducted to further identify the responsible transcription factor. The role of metformin in the treatment of anovulation by LOX inhibition was investigated in a high-fat diet (HFD)-induced obese rat model. The numbers of retrieved total oocytes and metaphase II oocytes were recorded upon ovarian stimulation. Masson's trichrome staining was used to measure the total collagen content, and immunohistochemical staining and western blotting were used to measure LOX, HIF-1α, and collagen I and IV in the ovary. MAIN RESULTS AND THE ROLE OF CHANCE Significantly increased FFA, LOX, and collagen abundance were observed in the ovaries of obese women with anovulation, compared to healthy controls or obese women with ovulation. In a HFD-induced obese rat model, metformin corrected the distortion of ovarian morphology by decreasing LOX and collagen protein abundance in the ovary and improving oestrous cyclicity and ovulation. PA increased LOX expression via the activation of HIF-1α in human granulosa cells, which was attenuated by metformin. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION Several other saturated and polyunsaturated FFAs, such as stearic acid and arachidonic acid, are also increased in the blood of individuals with obesity, and increased levels of other FFAs may also contribute to the development of anovulation in individuals with obesity, which needs to be further verified in the future. WIDER IMPLICATIONS OF THE FINDINGS Elevated PA in individuals with obesity can cause LOX dysregulation via activation of HIF-1α, resulting in abnormal collagen deposition in the ovary and anovulation. This dysregulation can be ameliorated by metformin therapy through its local effect on ECM remodelling in the ovary, which is independent of its systemic effect on insulin sensitivity and chronic inflammation. STUDY FUNDING/COMPETING INTERESTS This work was supported by the National Natural Science Foundation of China (grant numbers 82101730, 82130046, and 31900598) and Innovative Research Team of High-level local Universities in Shanghai (SHSMU-ZLCX20210201). All the authors declare no conflicts of interest in relation to this work.
Collapse
Affiliation(s)
- Chuyue Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Guangxin Yao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yanan Zhu
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei, China
| | - Yikai Lin
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jiangwen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
28
|
Mao Y, Weng J, Xie Q, Wu L, Xuan Y, Zhang J, Han J. Association between dietary inflammatory index and Stroke in the US population: evidence from NHANES 1999-2018. BMC Public Health 2024; 24:50. [PMID: 38166986 PMCID: PMC10763382 DOI: 10.1186/s12889-023-17556-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND There is an increasing awareness that diet-related inflammation may have an impact on the stroke. Herein, our goal was to decipher the association of dietary inflammatory index (DII) with stroke in the US general population. METHODS We collected the cross-sectional data of 44,019 participants of the National Health and Nutrition Examination Survey (NHANES) 1999-2018. The association of DII with stroke was estimated using weighted multivariate logistic regression, with its nonlinearity being examined by restricted cubic spline (RCS) regression. The least absolute shrinkage and selection operator (LASSO) regression was applied for identifying key stroke-related dietary factors, which was then included in the establishment of a risk prediction nomogram model, with the receiver operating characteristic (ROC) curve being built to evaluate its discriminatory power for stroke. RESULTS After confounder adjustment, the adjusted odds ratios (ORs) with 95% confidence intervals (CIs) for stroke across higher DII quartiles were 1.19 (0.94-1.54), 1.46 (1.16-1.84), and 1.87 (1.53-2.29) compared to the lowest quartile, respectively. The RCS curve showed a nonlinear and positive association between DII and stroke. The nomogram model based on key dietary factors identified by LASSO regression displayed a considerable predicative value for stroke, with an area under the curve (AUC) of 79.8% (78.2-80.1%). CONCLUSIONS Our study determined a nonlinear and positive association between DII and stroke in the US general population. Given the intrinsic limitations of cross-sectional study design, it is necessary to conduct more research to ensure the causality of such association.
Collapse
Affiliation(s)
- Yukang Mao
- Department of Cardiology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, 215008, Suzhou, China
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| | - Jiayi Weng
- Department of Cardiology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, 215008, Suzhou, China
| | - Qiyang Xie
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| | - Lida Wu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
| | - Yanling Xuan
- Nanjing University of Chinese Medicine, 210006, Nanjing, China
| | - Jun Zhang
- Department of Cardiology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, 215008, Suzhou, China.
| | - Jun Han
- Department of Infectious Diseases, Affiliated Wuxi Fifth Hospital of Jiangnan University, The Fifth People's Hospital of Wuxi, 214065, Wuxi, China.
| |
Collapse
|
29
|
Li H, Wang XK, Tang M, Lei L, Li JR, Sun H, Jiang J, Dong B, Li HY, Jiang JD, Peng ZG. Bacteroides thetaiotaomicron ameliorates mouse hepatic steatosis through regulating gut microbial composition, gut-liver folate and unsaturated fatty acids metabolism. Gut Microbes 2024; 16:2304159. [PMID: 38277137 PMCID: PMC10824146 DOI: 10.1080/19490976.2024.2304159] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024] Open
Abstract
Gut microbiota plays an essential role in the progression of nonalcoholic fatty liver disease (NAFLD), making the gut-liver axis a potential therapeutic strategy. Bacteroides genus, the enriched gut symbionts, has shown promise in treating fatty liver. However, further investigation is needed to identify specific beneficial Bacteroides strains for metabolic disorders in NAFLD and elucidate their underlying mechanisms. In this study, we observed a positive correlation between the abundance of Bacteroides thetaiotaomicron (B. theta) and the alleviation of metabolic syndrome in the early and end stages of NAFLD. Administration of B. theta to HFD-fed mice for 12 weeks reduced body weight and fat accumulation, decreased hyperlipidemia and insulin resistance, and prevented hepatic steatohepatitis and liver injury. Notably, B. theta did not affect these indicators in low-fat diet (LFD)-fed mice and exhibited good safety. Mechanistically, B. theta regulated gut microbial composition, characterized by a decreased Firmicutes/Bacteroidetes ratio in HFD-Fed mice. It also increased gut-liver folate levels and hepatic metabolites, alleviating metabolic dysfunction. Additionally, treatment with B. theta increased the proportion of polyunsaturated fatty acid in the mouse liver, offering a widely reported benefit for NAFLD improvement. In conclusion, this study provides evidence that B. theta ameliorates NAFLD by regulating gut microbial composition, enhancing gut-liver folate and unsaturated fatty acid metabolism, highlighting the therapeutic role of B. theta as a potential probiotic for NAFLD.
Collapse
Affiliation(s)
- Hu Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xue-Kai Wang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mei Tang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Lei
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jian-Rui Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Han Sun
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Jiang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Biao Dong
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hong-Ying Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jian-Dong Jiang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zong-Gen Peng
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Seliga AK, Zabłocki K, Bandorowicz-Pikuła J. Palmitate Stimulates Expression of the von Willebrand Factor and Modulates Toll-like Receptors Level and Activity in Human Umbilical Vein Endothelial Cells (HUVECs). Int J Mol Sci 2023; 25:254. [PMID: 38203423 PMCID: PMC10779284 DOI: 10.3390/ijms25010254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
An increased concentration of palmitate in circulation is one of the most harmful factors in obesity. The von Willebrand factor (vWF), a protein involved in haemostasis, is produced and secreted by the vascular endothelium. An increased level of vWF in obese patients is associated with thrombosis and cardiovascular disease. The aim of this study was to investigate a palmitate effect on vWF in endothelial cells and understand the mechanisms of palmitate-activated signalling. Human umbilical vein endothelial cells (HUVECs) incubated in the presence of palmitate, exhibited an increased VWF gene expression, vWF protein maturation, and stimulated vWF secretion. Cardamonin, a Nuclear Factor kappa B (NF-κB) inhibitor, abolished the palmitate effect on VWF expression. The inhibition of Toll-like receptor (TLR) 2 with C29 resulted in the TLR4 overactivation in palmitate-treated cells. Palmitate, in the presence of TLR4 inhibitor TAK-242, leads to a higher expression of TLR6, CD36, and TIRAP. The silencing of TLR4 resulted in an increase in TLR2 level and vice versa. The obtained results indicate a potential mechanism of obesity-induced thrombotic complication caused by fatty acid activation of NF-κB signalling and vWF upregulation and help to identify various compensatory mechanisms related to TLR4 signal transduction.
Collapse
Affiliation(s)
| | | | - Joanna Bandorowicz-Pikuła
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology PAS, 3 Pasteur Str., 02-093 Warsaw, Poland; (A.K.S.); (K.Z.)
| |
Collapse
|
31
|
Wences Chirino T, Rangel López E, Luna Angulo A, Carrillo Mora P, Landa Solis C, Samudio Cruz MA, Fuentes Bello AC, Paniagua Pérez R, Ríos Martínez J, Sánchez Chapul L. Crosstalk between Exercise-Derived Endocannabinoidome and Kynurenines: Potential Target Therapies for Obesity and Depression Symptoms. Pharmaceuticals (Basel) 2023; 16:1421. [PMID: 37895892 PMCID: PMC10609722 DOI: 10.3390/ph16101421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
The kynurenine pathway (KP) and the endocannabinoid system (ECS) are known to be deregulated in depression and obesity; however, it has been recognized that acute physical exercise has an important modulating role inducing changes in the mobilization of their respective metabolites-endocannabinoids (eCBs) and kynurenines (KYNs)-which overlap at some points, acting as important antidepressant, anti-nociceptive, anti-inflammatory, and antioxidant biomarkers. Therefore, the aim of this review is to analyze and discuss some recently performed studies to investigate the potential interactions between both systems, particularly those related to exercise-derived endocannabinoidome and kynurenine mechanisms, and to elucidate how prescription of physical exercise could represent a new approach for the clinical management of these two conditions.
Collapse
Affiliation(s)
- Tiffany Wences Chirino
- Neuromuscular Diseases Laboratory, Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico; (T.W.C.); (A.L.A.); (A.C.F.B.)
| | - Edgar Rangel López
- Cell Reprogramming Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico;
| | - Alexandra Luna Angulo
- Neuromuscular Diseases Laboratory, Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico; (T.W.C.); (A.L.A.); (A.C.F.B.)
| | - Paul Carrillo Mora
- Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico; (P.C.M.); (M.A.S.C.)
| | - Carlos Landa Solis
- Tissue Engineering, Cell Therapy, and Regenerative Medicine Unit, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico;
| | - María Alejandra Samudio Cruz
- Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico; (P.C.M.); (M.A.S.C.)
| | - Alim C. Fuentes Bello
- Neuromuscular Diseases Laboratory, Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico; (T.W.C.); (A.L.A.); (A.C.F.B.)
| | - Rogelio Paniagua Pérez
- Biochemistry Laboratory, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico;
| | - Juan Ríos Martínez
- Health Sciences Research Institute, Mexican Navy, Mexico City 04470, Mexico;
| | - Laura Sánchez Chapul
- Neuromuscular Diseases Laboratory, Clinical Neurosciences Division, National Institute of Rehabilitation “Luis Guillermo Ibarra Ibarra”, Mexico City 14389, Mexico; (T.W.C.); (A.L.A.); (A.C.F.B.)
| |
Collapse
|
32
|
Bishehsari F, Drees M, Adnan D, Sharma D, Green S, Koshy J, Giron LB, Goldman A, Abdel-Mohsen M, Rasmussen HE, Miller GE, Keshavarzian A. Multi-omics approach to socioeconomic disparity in metabolic syndrome reveals roles of diet and microbiome. Proteomics 2023; 23:e2300023. [PMID: 37525324 DOI: 10.1002/pmic.202300023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023]
Abstract
The epidemy of metabolic syndrome (MetS) is typically preceded by adoption of a "risky" lifestyle (e.g., dietary habit) among populations. Evidence shows that those with low socioeconomic status (SES) are at an increased risk for MetS. To investigate this, we recruited 123 obese subjects (body mass index [BMI] ≥ 30) from Chicago. Multi-omic data were collected to interrogate fecal microbiota, systemic markers of inflammation and immune activation, plasma metabolites, and plasma glycans. Intestinal permeability was measured using the sugar permeability testing. Our results suggest a heterogenous metabolic dysregulation among obese populations who are at risk of MetS. Systemic inflammation, linked to poor diet, intestinal microbiome dysbiosis, and gut barrier dysfunction may explain the development of MetS in these individuals. Our analysis revealed 37 key features associated with increased numbers of MetS features. These features were used to construct a composite metabolic-inflammatory (MI) score that was able to predict progression of MetS among at-risk individuals. The MI score was correlated with several markers of poor diet quality as well as lower levels of gut microbial diversity and abnormalities in several species of bacteria. This study reveals novel targets to reduce the burden of MetS and suggests access to healthy food options as a practical intervention.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Michael Drees
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Darbaz Adnan
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Deepak Sharma
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Stefan Green
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Jane Koshy
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Leila B Giron
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Aaron Goldman
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | - Gregory E Miller
- Institute for Policy Research and Dept of Psychology, Northwestern Univ, Evanston, Illinois, USA
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
33
|
Liu D, Aziz NA, Landstra EN, Breteler MMB. The lipidomic correlates of epigenetic aging across the adult lifespan: A population-based study. Aging Cell 2023; 22:e13934. [PMID: 37496173 PMCID: PMC10497837 DOI: 10.1111/acel.13934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023] Open
Abstract
Lipid signaling is involved in longevity regulation, but which specific lipid molecular species affect human biological aging remains largely unknown. We investigated the relation between complex lipids and DNA methylation-based metrics of biological aging among 4181 participants (mean age 55.1 years (range 30.0-95.0)) from the Rhineland Study, an ongoing population-based cohort study in Bonn, Germany. The absolute concentration of 14 lipid classes, covering 964 molecular species and 267 fatty acid composites, was measured by Metabolon Complex Lipid Panel. DNA methylation-based metrics of biological aging (AgeAccelPheno and AgeAccelGrim) were calculated based on published algorithms. Epigenome-wide association analyses (EWAS) of biological aging-associated lipids and pathway analysis were performed to gain biological insights into the mechanisms underlying the effects of lipidomics on biological aging. We found that higher levels of molecular species belonging to neutral lipids, phosphatidylethanolamines, phosphatidylinositols, and dihydroceramides were associated with faster biological aging, whereas higher levels of lysophosphatidylcholine, hexosylceramide, and lactosylceramide species were associated with slower biological aging. Ceramide, phosphatidylcholine, and lysophosphatidylethanolamine species with odd-numbered fatty acid tail lengths were associated with slower biological aging, whereas those with even-numbered chain lengths were associated with faster biological aging. EWAS combined with functional pathway analysis revealed several complex lipids associated with biological aging as important regulators of known longevity and aging-related pathways.
Collapse
Affiliation(s)
- Dan Liu
- Population Health SciencesGerman Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - N. Ahmad Aziz
- Population Health SciencesGerman Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Neurology, Faculty of MedicineUniversity of BonnBonnGermany
| | - Elvire Nadieh Landstra
- Population Health SciencesGerman Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Monique M. B. Breteler
- Population Health SciencesGerman Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of MedicineUniversity of BonnBonnGermany
| |
Collapse
|
34
|
Delgado-Alarcón JM, Hernández Morante JJ, Morillas-Ruiz JM. Modification of Breakfast Fat Composition Can Modulate Cytokine and Other Inflammatory Mediators in Women: A Randomized Crossover Trial. Nutrients 2023; 15:3711. [PMID: 37686743 PMCID: PMC10489665 DOI: 10.3390/nu15173711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Previous trials have demonstrated that modifying dietary fat composition can influence the production of inflammation-related factors. Additionally, it has been suggested that not only the type of fat, but also the timing of fat intake can impact these factors. Therefore, the objective of the present study was to evaluate the effect of altering breakfast fat composition on inflammatory parameters. A 3-month crossover randomized trial was designed, involving 60 institutionalized women who alternately consumed a breakfast rich in polyunsaturated fatty acids (PUFA) (margarine), monounsaturated fatty acids (MUFA) (virgin olive oil), or saturated fatty acids (SFA) (butter), based on randomization. The following inflammatory markers were evaluated: epidermal growth factor (EGF), interferon (IFN)-α, interleukin (IL)-1α, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor (TNF)-α, C-reactive protein (CRP), and vascular/endothelial growth factor (VEGF). The results showed that the most significant effects were observed with the high-MUFA breakfast, as there was a statistically significant decrease in plasma IL-6 (p = 0.016) and VEGF values (p = 0.035). Other factors, such as IL-1α and CRP, also decreased substantially, but did not reach the statistically significant level. On the other hand, the high-PUFA breakfast induced a significant decrease in EGF levels (p < 0.001), whereas the high-SFA breakfast had no apparent effect on these factors. In conclusion, modifying breakfast fat, particularly by increasing MUFA or PUFA intake, appears to be sufficient for promoting a lower inflammatory marker synthesis profile and may be beneficial in improving cardiovascular complications.
Collapse
Affiliation(s)
- Jessica M. Delgado-Alarcón
- Department of Food Technology and Nutrition, Universidad Católica de Murcia, Campus de Los Jerónimos, Guadalupe, 30107 Murcia, Spain;
| | - Juan José Hernández Morante
- Eating Disorders Research Unit, Universidad Católica de Murcia, Campus de Los Jerónimos, Guadalupe, 30107 Murcia, Spain
| | - Juana M. Morillas-Ruiz
- Department of Food Technology and Nutrition, Universidad Católica de Murcia, Campus de Los Jerónimos, Guadalupe, 30107 Murcia, Spain;
| |
Collapse
|
35
|
Minne X, Mbuya Malaïka Mutombo J, Chandad F, Fanganiello RD, Houde VP. Porphyromonas gingivalis under palmitate-induced obesogenic microenvironment modulates the inflammatory transcriptional signature of macrophage-like cells. PLoS One 2023; 18:e0288009. [PMID: 37384642 PMCID: PMC10309636 DOI: 10.1371/journal.pone.0288009] [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: 04/11/2023] [Accepted: 06/16/2023] [Indexed: 07/01/2023] Open
Abstract
Metabolic diseases and low-grade chronic inflammation are interconnected: obese persons are at higher risk of developing periodontitis. However, the molecular mechanisms involved in the development and progression of periodontitis in an obesogenic microenvironment in response to periodontopathogens are still lacking. This study aims to investigate the combined effects of palmitate and Porphyromonas gingivalis on the secretion of pro-inflammatory cytokines and on transcriptional landscape modifications in macrophage-like cells. U937 macrophage-like cells were treated with palmitate and stimulated with P. gingivalis for 24h. Cytokines IL-1β, TNF-α and IL-6 were measured by ELISA in the culture medium and cell extracted RNA was submitted to a microarray analysis followed by Gene Ontology analyses. P. gingivalis, in presence of palmitate, potentiated IL-1β and TNF-α secretion in comparison to palmitate alone. Gene Ontology analyses also revealed that the combination palmitate-P. gingivalis potentiated the number of gene molecular functions implicated in the regulation of immune and inflammatory pathways compared to macrophages treated with palmitate alone. Our results provide the first comprehensive mapping of gene interconnections between palmitate and P. gingivalis during inflammatory responses in macrophage-like cells. These data highlight the importance of considering systemic conditions, specifically obesogenic microenvironment, in the management of periodontal disease in obese patients.
Collapse
Affiliation(s)
- Xavier Minne
- Faculty of Dentistry, Oral Ecology Research Group (GREB), Université Laval, Quebec City, Quebec, Canada
| | | | - Fatiha Chandad
- Faculty of Dentistry, Oral Ecology Research Group (GREB), Université Laval, Quebec City, Quebec, Canada
| | - Roberto D. Fanganiello
- Faculty of Dentistry, Oral Ecology Research Group (GREB), Université Laval, Quebec City, Quebec, Canada
| | - Vanessa P. Houde
- Faculty of Dentistry, Oral Ecology Research Group (GREB), Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
36
|
Otero A, Becerril S, Martín M, Cienfuegos JA, Valentí V, Moncada R, Catalán V, Gómez-Ambrosi J, Burrell MA, Frühbeck G, Rodríguez A. Effect of guanylin peptides on pancreas steatosis and function in experimental diet-induced obesity and after bariatric surgery. Front Endocrinol (Lausanne) 2023; 14:1185456. [PMID: 37274331 PMCID: PMC10233012 DOI: 10.3389/fendo.2023.1185456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction Obesity contributes to ectopic fat deposition in non-adipose organs, including the pancreas. Pancreas steatosis associates with inflammation and β-cell dysfunction, contributing to the onset of insulin resistance and type 2 diabetes. An improvement of pancreatic steatosis and indices of insulin resistance is observed following bariatric surgery, but the underlying mechanisms remain unknown. We sought to analyze whether guanylin (GUCA2A) and uroguanylin (GUCA2B), two gut hormones involved in the regulation of satiety, food preference and adiposity, are involved in the amelioration of pancreas fat accumulation after bariatric surgery. Methods Pancreas steatosis, inflammation, islet number and area were measured in male Wistar rats with diet-induced obesity (n=125) subjected to surgical (sham operation and sleeve gastrectomy) or dietary (pair-fed to the amount of food eaten by gastrectomized animals) interventions. The tissue distribution of guanylate cyclase C (GUCY2C) and the expression of the guanylin system were evaluated in rat pancreata by real-time PCR, Western-blot and immunohistochemistry. The effect of guanylin and uroguanylin on factors involved in insulin secretion and lipogenesis was determined in vitro in RIN-m5F β-cells exposed to lipotoxic conditions. Results Sleeve gastrectomy reduced pancreas steatosis and inflammation and improved insulin sensitivity and synthesis. An upregulation of GUCA2A and GUCY2C, but not GUCA2B, was observed in pancreata from rats with diet-induced obesity one month after sleeve gastrectomy. Interestingly, both guanylin and uroguanylin diminished the lipotoxicity in palmitate-treated RIN-m5F β-cells, evidenced by lower steatosis and downregulated lipogenic factors Srebf1, Mogat2 and Dgat1. Both guanylin peptides reduced insulin synthesis (Ins1 and Ins2) and release from RIN-m5F β-cells, but only guanylin upregulated Wnt4, a factor that controls β-cell proliferation and function. Discussion Together, sleeve gastrectomy reduced pancreatic steatosis and improved β-cell function. Several mechanisms, including the modulation of inflammation and lipogenesis as well as the upregulation of GUCA2A in the pancreas, might explain this beneficial effect of bariatric surgery.
Collapse
Affiliation(s)
- Aarón Otero
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Marina Martín
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Javier A. Cienfuegos
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - María A. Burrell
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
37
|
Morris I, Croes CA, Boes M, Kalkhoven E. Advanced omics techniques shed light on CD1d-mediated lipid antigen presentation to iNKT cells. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159292. [PMID: 36773690 DOI: 10.1016/j.bbalip.2023.159292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Invariant natural killer T cells (iNKT cells) can be activated through binding antigenic lipid/CD1d complexes to their TCR. Antigenic lipids are processed, loaded, and displayed in complex with CD1d by lipid antigen presenting cells (LAPCs). The mechanism of lipid antigen presentation via CD1d is highly conserved with recent work showing adipocytes are LAPCs that, besides having a role in lipid storage, can activate iNKT cells and play an important role in systemic metabolic disease. Recent studies shed light on parameters potentially dictating cytokine output and how obesity-associated metabolic disease may affect such parameters. By following a lipid antigen's journey, we identify five key areas which may dictate cytokine skew: co-stimulation, structural properties of the lipid antigen, stability of lipid antigen/CD1d complexes, intracellular and extracellular pH, and intracellular and extracellular lipid environment. Recent publications indicate that the combination of advanced omics-type approaches and machine learning may be a fruitful way to interconnect these 5 areas, with the ultimate goal to provide new insights for therapeutic exploration.
Collapse
Affiliation(s)
- Imogen Morris
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands
| | - Cresci-Anne Croes
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708WE Wageningen, the Netherlands
| | - Marianne Boes
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands; Department of Paediatric Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands.
| |
Collapse
|
38
|
Rubak YT, Lalel HJD, Sanam MUE. Physicochemical, microbiological, and sensory characteristics of " Sui Wu'u" traditional pork products from Bajawa, West Flores, Indonesia. Vet World 2023; 16:1165-1175. [PMID: 37576773 PMCID: PMC10420695 DOI: 10.14202/vetworld.2023.1165-1175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/19/2023] [Indexed: 08/15/2023] Open
Abstract
Background and Aim Sui Wu'u is a traditional meat preservation product from Bajawa, a region in East Nusa Tenggara. It is made by mixing pork with salt and corn flour, which is then stored in a bamboo container (tuku) for months. After 6 months of storage, this study examined the physicochemical, microbiological, and sensory properties of Sui Wu'u. Materials and Methods Sui Wu'u products were prepared using the traditional recipe from the Bajawa community. Fresh pork (pork belly and backfat), corn flour, and salt were purchased from local/traditional markets at proportions of 65%, 30%, and 5%, respectively. The physicochemical, amino acid, fatty acid profile, microbiological, and sensory properties of Sui Wu'u were evaluated after being stored for 6 months in a bamboo container (tuku). Results The results indicated that these Sui Wu'u were mainly characterized by high-fat levels, followed by protein. The pH value, salt content, moisture content, and water activity were 4.72%, 1.72%, 6.11%, and 0.62%, respectively. Minerals (K, P, Se, and Zn) and vitamin B6, as well as amino acids, such as leucine, phenylalanine, lysine (essential amino acids), glycine, proline, glutamic acid, and alanine (non-essential amino acids), are present in Sui Wu'u. The fatty acid profile was dominated by monounsaturated fatty acids (MUFA) (21.69%), saturated fatty acids (SFA) (17.78%), and polyunsaturated fatty acids (PUFA) (5.36%). Monounsaturated fatty acids, oleic acid (C18:1n9) was the most abundant fatty acid in Sui Wu'u, followed by palmitic acid SFA (C16:0); MUFA stearic acid (C18:0); and PUFA linoleic (C18:2n-6). The microbiological characteristics of Sui Wu'u showed no detectable microorganisms (<10 CFU/g) for Salmonella, total E. coli and total Staphylococcus, and average values of 4.4 × 105 CFU/g for total microbes, which were still below the maximum limit of microbial contamination according to the regulations of the Food and Drug Supervisory Agency of the Republic of Indonesia. The sensory assessment indicated that panelists highly preferred (rated as very like) Sui Wu'u for all sensory attributes. Conclusion The physicochemical, microbiological, and sensory characteristics of Sui Wu'u after 6 months of storage indicated that it still provides essential nutrients for the body and is quite safe for consumption. The stability of Sui Wu'u's shelf life can be attributed to the appropriate combination of pork, salt, corn flour, bamboo packaging (tuku), and storage temperature. The high-fat content in Sui Wu'u can be reduced by increasing the proportion of lean meat. Ensuring strict sanitation during the manufacturing process, using high-quality pork, salt, corn flour, and proper packaging with bamboo can further improve the safety of Sui Wu'u for consumption.
Collapse
Affiliation(s)
- Yuliana Tandi Rubak
- Department of Agrotechnology, Faculty of Agriculture, Universitas Nusa Cendana, Kupang, East Nusa Tenggara 85228, Indonesia
| | - Herianus J. D. Lalel
- Department of Agrotechnology, Faculty of Agriculture, Universitas Nusa Cendana, Kupang, East Nusa Tenggara 85228, Indonesia
| | - Maxs Urias Ebenhaizar Sanam
- Department of Animal Diseases Sciences and Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Nusa Cendana, Kupang, East Nusa Tenggara 85228, Indonesia
| |
Collapse
|
39
|
Sergi D, Sanz JM, Lazzer S, Brombo G, Zuliani G, Biolo G, Šimunič B, Pišot R, Dalla Nora E, Passaro A. Interleukin-18 Is a Potential Biomarker Linking Dietary Fatty Acid Quality and Insulin Resistance: Results from a Cross-Sectional Study in Northern Italy. Nutrients 2023; 15:nu15071782. [PMID: 37049621 PMCID: PMC10097292 DOI: 10.3390/nu15071782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
Dietary lipids are pivotal in modulating metabolic inflammation. Among the inflammatory mediators characterizing metabolic inflammation, interleukin 18 (IL-18) has been consistently associated with obesity and insulin resistance. This study aims to evaluate whether the quality of lipid intake impacts upon IL-18 plasma levels and the implications on insulin resistance computed by the homeostatic model assessment for insulin resistance (HOMA-IR). Using a cross-sectional design, this study confirmed that IL-18 correlated positively with insulin resistance and individuals with a HOMA-IR ≥ 2.5 displayed higher circulating IL-18 levels compared with their insulin-sensitive counterparts. In terms of the effect of the quality of dietary lipids on IL-18 circulating levels, the ratio between monounsaturated, omega-3, polyunsaturated and saturated fatty acids as well as the intake of eicosapentaenoic and docosahexaenoic acids correlated negatively with IL-18. Despite this, IL-18 circulating levels, but not dietary fatty acid quality, predicted insulin resistance. Nevertheless, the ratio between omega 3 and saturated fatty acids was a predictor of IL-18 plasma levels. Thus, the downregulation of IL-18 may underpin, at least partially, the beneficial metabolic effects of substituting omega 3 for saturated fatty acids with this cytokine potentially representing a biomarker linking dietary lipids and metabolic outcomes.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, I-44121 Ferrara, Italy
| | - Juana Maria Sanz
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari, 46, I-44121 Ferrara, Italy
| | - Stefano Lazzer
- Department of Medicine, University of Udine, Piazzale M. Kolbe 4, I-33100 Udine, Italy
| | - Gloria Brombo
- Medical Department, University Hospital of Ferrara Arcispedale Sant’Anna, Via A. Moro 8, I-44124 Ferrara, Italy
| | - Giovanni Zuliani
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, I-44121 Ferrara, Italy
- Medical Department, University Hospital of Ferrara Arcispedale Sant’Anna, Via A. Moro 8, I-44124 Ferrara, Italy
| | - Gianni Biolo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, I-34149 Trieste, Italy
| | - Boštjan Šimunič
- Institute for Kinesiology Research, Science and Research Centre of Koper, Garibaldijeva 1, SI-6000 Koper, Slovenia
| | - Rado Pišot
- Institute for Kinesiology Research, Science and Research Centre of Koper, Garibaldijeva 1, SI-6000 Koper, Slovenia
| | - Edoardo Dalla Nora
- Medical Department, University Hospital of Ferrara Arcispedale Sant’Anna, Via A. Moro 8, I-44124 Ferrara, Italy
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, I-44121 Ferrara, Italy
- Medical Department, University Hospital of Ferrara Arcispedale Sant’Anna, Via A. Moro 8, I-44124 Ferrara, Italy
| |
Collapse
|
40
|
Saalbach A, Seitz AT, Kohlmann J, Kalweit L, Vogt L, Selig L, Engel KM, Simon JC. Modulation of Dietary Fatty Acids in an Open-Label Study Improves Psoriasis and Dampens the Inflammatory Activation Status. Nutrients 2023; 15:nu15071698. [PMID: 37049538 PMCID: PMC10097201 DOI: 10.3390/nu15071698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Obesity and high abdominal fat mass are risk factors for developing the chronic inflammatory skin disease psoriasis. They are associated with increased incidence, prevalence and severity of the disease. A positive effect of weight loss on psoriasis activity has been shown in several studies. Obesity-related factors such as the dysregulation of glucose and lipid metabolism, the activation of adipose tissue and resultant persistent low-grade inflammation have been discussed as links of obesity and inflammatory diseases. Recently, we demonstrated a critical role of free fatty acids (FFAs) in obesity-mediated exacerbation of psoriatic skin inflammation in both mice and humans. In the present study, we translated these findings into a therapeutic intervention. An open-label study focusing on the dietary reduction of FFAs was conducted in patients with mild-to-moderate plaque psoriasis, and disease severity and serum markers of inflammation were analyzed. Here, we show that such a dietary intervention improves psoriatic disease activity independently of weight loss. Diet-related metabolic changes, such as a reduction in saturated free fatty acids (SFAs), may thus be more important than weight loss itself. Moreover, dietary intervention inhibited the overall pro-inflammatory activation status in patients, as shown by analysis of serum inflammatory parameters using the Olink platform. From our pilot study, we conclude that dietary intervention focusing on SFA reduction has the capacity to reduce disease activity and general inflammatory status in psoriasis patients.
Collapse
Affiliation(s)
- Anja Saalbach
- Department of Dermatology, Venerology and Allergology, Faculty of Medicine, Leipzig University, Philipp Rosenthal Str. 23, 04103 Leipzig, Germany
| | - Anna-Theresa Seitz
- Department of Dermatology, Venerology and Allergology, Faculty of Medicine, Leipzig University, Philipp Rosenthal Str. 23, 04103 Leipzig, Germany
| | - Johannes Kohlmann
- Department of Dermatology, Venerology and Allergology, Faculty of Medicine, Leipzig University, Philipp Rosenthal Str. 23, 04103 Leipzig, Germany
| | - Lena Kalweit
- Department of Dermatology, Venerology and Allergology, Faculty of Medicine, Leipzig University, Philipp Rosenthal Str. 23, 04103 Leipzig, Germany
| | - Lisa Vogt
- Department of Dermatology, Venerology and Allergology, Faculty of Medicine, Leipzig University, Philipp Rosenthal Str. 23, 04103 Leipzig, Germany
| | - Lars Selig
- Department of Medicine, Division of Nutritional Medicine, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Kathrin M. Engel
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, 04107 Leipzig, Germany
| | - Jan C. Simon
- Department of Dermatology, Venerology and Allergology, Faculty of Medicine, Leipzig University, Philipp Rosenthal Str. 23, 04103 Leipzig, Germany
| |
Collapse
|
41
|
Palmitic Acid Inhibits Myogenic Activity and Expression of Myosin Heavy Chain MHC IIb in Muscle Cells through Phosphorylation-Dependent MyoD Inactivation. Int J Mol Sci 2023; 24:ijms24065847. [PMID: 36982919 PMCID: PMC10054354 DOI: 10.3390/ijms24065847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Sarcopenia associated with aging and obesity is characterized by the atrophy of fast-twitch muscle fibers and an increase in intramuscular fat deposits. However, the mechanism of fast-twitch fiber-specific atrophy remains unclear. In this study, we aimed to assess the effect of palmitic acid (PA), the most common fatty acid component of human fat, on muscle fiber type, focusing on the expression of fiber-type-specific myosin heavy chain (MHC). Myotubes differentiated from C2C12 myoblasts were treated with PA. The PA treatment inhibited myotube formation and hypertrophy while reducing the gene expression of MHC IIb and IIx, specific isoforms of fast-twitch fibers. Consistent with this, a significant suppression of MHC IIb protein expression in PA-treated cells was observed. A reporter assay using plasmids containing the MHC IIb gene promoter revealed that the PA-induced reduction in MHC IIb gene expression was caused by the suppression of MyoD transcriptional activity through its phosphorylation. Treatment with a specific protein kinase C (PKC) inhibitor recovered the reduction in MHC IIb gene expression levels in PA-treated cells, suggesting the involvement of the PA-induced activation of PKC. Thus, PA selectively suppresses the mRNA and protein expression of fast-twitch MHC by modulating MyoD activity. This finding provides a potential pathogenic mechanism for age-related sarcopenia.
Collapse
|
42
|
Sonnefeld L, Rohmann N, Geisler C, Laudes M. Is human obesity an inflammatory disease of the hypothalamus? Eur J Endocrinol 2023; 188:R37-R45. [PMID: 36883605 DOI: 10.1093/ejendo/lvad030] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/23/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Obesity and its comorbidities are long-standing, challenging global health problems. Lack of exercise, overnutrition, and especially the consumption of fat-rich foods are some of the most important factors leading to an increase in prevalence in modern society. The pathophysiology of obesity as a metabolic inflammatory disease has moved into focus since new therapeutic approaches are required. The hypothalamus, a brain area responsible for energy homeostasis, has recently received special attention in this regard. Hypothalamic inflammation was identified to be associated with diet-induced obesity and new evidence suggests that it may be, beyond that, a pathological mechanism of the disease. This inflammation impairs the local signaling of insulin and leptin leading to dysfunction of the regulation of energy balance and thus, weight gain. After a high-fat diet consumption, activation of inflammatory mediators such as the nuclear factor κB or c-Jun N-terminal kinase pathway can be observed, accompanied by elevated secretion of pro-inflammatory interleukins and cytokines. Brain resident glia cells, especially microglia and astrocytes, initiate this release in response to the flux of fatty acids. The gliosis occurs rapidly before the actual weight gain. Dysregulated hypothalamic circuits change the interaction between neuronal and non-neuronal cells, contributing to the establishment of inflammatory processes. Several studies have reported reactive gliosis in obese humans. Although there is evidence for a causative role of hypothalamic inflammation in the obesity development, data on underlying molecular pathways in humans are limited. This review discusses the current state of knowledge on the relationship between hypothalamic inflammation and obesity in humans.
Collapse
Affiliation(s)
- Lena Sonnefeld
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| | - Nathalie Rohmann
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| | - Corinna Geisler
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| | - Matthias Laudes
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Medicine 1, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| |
Collapse
|
43
|
Patial V, Katoch S, Chhimwal J, Dadhich G, Sharma V, Rana A, Joshi R, Padwad Y. Catechins prevent obesity-induced kidney damage by modulating PPARγ/CD36 pathway and gut-kidney axis in rats. Life Sci 2023; 316:121437. [PMID: 36702203 DOI: 10.1016/j.lfs.2023.121437] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023]
Abstract
Obesity is an epidemic and a growing public health concern worldwide. It is one of the significant risk factors for developing chronic kidney disease. In the present study, we evaluated the preventive effect of green tea catechins (GTC) against obesity-induced kidney damage and revealed the underlying molecular mechanism of action. Various green tea catechins were quantified in the catechins-rich fraction using HPLC. In vitro, the palmitic and oleic acid-treated NRK-52E cells showed reduced fat accumulation and modulated expressions of PPARγ, CD36, and TGFβ after GTC treatment. In vivo, rats were fed with a high-fat diet (HFD), and the effect of GTC was assessed at 150 and 300 mg/kg body weight doses. HFD-fed rats showed a significant reduction in weight gain and improved serum creatinine, urea, and urine microalbumin levels after GTC treatment. The improved adipokines and insulin levels in GTC treated groups indicated the insulin-sensitizing effect. Histopathology revealed reduced degenerative changes, fibrous tissue deposition, and mesangial matrix proliferation in GTC treated groups. GTC treatment also downregulated the gene expressions of lipogenic and inflammatory factors and improved the altered expressions of CD36 and PPARγ in the kidney tissue. Further, GTC prevented gut dysbiosis in rats by promoting healthy microbes like Akkermansia muciniphila and Lactobacillus reuteri. Faecal metabolome revealed reduced saturated fatty acids, and improved amino acid levels in the GTC treated groups, which help to maintain gut health and metabolism. Overall, GTC prevented obesity-induced kidney damage by modulating PPARγ/CD36 signaling and maintaining gut health in rats.
Collapse
Affiliation(s)
- Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| | - Swati Katoch
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Garima Dadhich
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Vinesh Sharma
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Ajay Rana
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India
| | - Robin Joshi
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Yogendra Padwad
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
44
|
Karmakar E, Das N, Mukherjee B, Das P, Mukhopadhyay S, Roy SS. Lipid-induced alteration in retinoic acid signaling leads to mitochondrial dysfunction in HepG2 and Huh7 cells. Biochem Cell Biol 2023. [PMID: 36787544 DOI: 10.1139/bcb-2022-0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
A surfeit of mitochondrial reactive oxygen species (ROS) and inflammation serve as obligatory mediators of lipid-associated hepatocellular maladies. While retinoid homeostasis is essential in restoring systemic energy balance, its role in hepatic mitochondrial function remains elusive. The role of lecithin-retinol acyltransferase (LRAT) in maintenance of retinoid homeostasis is appreciated earlier; however, its role in modulating retinoic acid (RA) bioavailability upon lipid-imposition is unexplored. We identified LRAT overexpression in high-fat diet (HFD)-fed rats and palmitate-treated hepatoma cells. Elevation in LRAT expression depletes RA production and deregulates RA signaling. This altered RA metabolism enhances fat accumulation, accompanied by inflammation that leads to impaired mitochondrial function through enhanced ROS generation. Hence, LRAT inhibition could be a novel approach preventing lipid-induced mitochondrial dysfunction in hepatoma cells.
Collapse
Affiliation(s)
- Eshani Karmakar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India
| | - Nabanita Das
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India.,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Bijnor-sisendi Road, Lucknow, Uttar Pradesh, 226002, India
| | - Bidisha Mukherjee
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, 244, A.J.C. Bose Road, Kolkata, 700020, India
| | - Prosenjit Das
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India
| | - Satinath Mukhopadhyay
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, 244, A.J.C. Bose Road, Kolkata, 700020, India
| | - Sib Sankar Roy
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India.,Academy of Scientific & Innovative Research (AcSIR), India
| |
Collapse
|
45
|
Tumor and peritumoral adipose tissue crosstalk: De-differentiated adipocytes influence spread of colon carcinoma cells. Tissue Cell 2023; 80:101990. [PMID: 36542947 DOI: 10.1016/j.tice.2022.101990] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022]
Abstract
Colorectal cancer is the second leading cause of cancer and often has a fatal course. There are many studies in the literature that have described a close functional relationship between the tumor mass and surrounding tissue, or tumor stroma, which is affected by the continuous metabolic exchange that occurs at the interface between tumor and tissues in contact with it. There is much evidence that the presence of adipose tissue in stroma plays a fundamental role in modulating the tumor microenvironment and promote tumor development, growth, and angiogenesis due to its endocrine characteristics. In this analysis, we have studied the alterations of adipose tissue surrounding colorectal tumors with MRI and optical imaging in vivo techniques to monitor tumor progression and also performed histological and molecular analysis. We detected differences in the principal adipose markers expressed by adipocytes residing around the rectal colon and observed that peritumoral adipose tissue is exposed to a mesenchymal transition process that leads to the acquisition of a less differentiated phenotype of adipocyte that represents the main cellular type present in tumor stroma. The mesenchymal transition correlated with the acquisition of more aggressive tumor phenotype and could represent a valid target for tumor therapy.
Collapse
|
46
|
López-Acosta O, Ruiz-Ramírez A, Barrios-Maya MÁ, Alarcon-Aguilar J, Alarcon-Enos J, Céspedes Acuña CL, El-Hafidi M. Lipotoxicity, glucotoxicity and some strategies to protect vascular smooth muscle cell against proliferative phenotype in metabolic syndrome. Food Chem Toxicol 2023; 172:113546. [PMID: 36513245 DOI: 10.1016/j.fct.2022.113546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is a risk factor for the development of cardiovascular disease (CVD) and atherosclerosis through a mechanism that involves vascular smooth muscle cell (VSMC) proliferation, lipotoxicity and glucotoxicity. Several molecules found to be increased in MetS, including free fatty acids, fatty acid binding protein 4, leptin, resistin, oxidized lipoprotein particles, and advanced glycation end products, influence VSMC proliferation. Most of these molecules act through their receptors on VSMCs by activating several signaling pathways associated with ROS generation in various cellular compartments. ROS from NADPH-oxidase and mitochondria have been found to promote VSMC proliferation and cell cycle progression. In addition, most of the natural or synthetic substances described in this review, including pharmaceuticals with hypoglycemic and hypolipidemic properties, attenuate VSMC proliferation by their simultaneous modulation of cell signaling and their scavenging property due to the presence of a phenolic ring in their structure. This review discusses recent data in the literature on the role that several MetS-related molecules and ROS play in the change from contractile to proliferative phenotype of VSMCs. Hence the importance of proposing an appropriate strategy to prevent uncontrolled VSMC proliferation using antioxidants, hypoglycemic and hypolipidemic agents.
Collapse
Affiliation(s)
- Ocarol López-Acosta
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Angélica Ruiz-Ramírez
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Miguel-Ángel Barrios-Maya
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Javier Alarcon-Aguilar
- Laboratorio de Farmacología, Depto. de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Iztapalapa, Iztapalapa, Mexico
| | - Julio Alarcon-Enos
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile
| | - Carlos L Céspedes Acuña
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile.
| | - Mohammed El-Hafidi
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico.
| |
Collapse
|
47
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
48
|
Alizadeh F, Mirzaie Bavil F, Keyhanmanesh R, Lotfi H, Ghiasi F. Association of pro-inflammatory cytokines, inflammatory proteins with atherosclerosis index in obese male subjects. Horm Mol Biol Clin Investig 2023:hmbci-2022-0022. [PMID: 36592952 DOI: 10.1515/hmbci-2022-0022] [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: 02/17/2022] [Accepted: 07/19/2022] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Investigation the association of pro-inflammatory markers interleukin (IL)-1β and IL- 10 expression, serum levels of C-reactive protein (CRP), cyclooxygenase-2 (COX2), High-density lipoprotein (HDL), Apolipoprotein A1 (ApoA1), and ATP Binding Cassette Subfamily A Member 1 (ABCA1) inflammatory proteins with atherosclerosis index (homocysteine) in normal-weight and obese male subjects. METHODS 59 males including 30 obese (Body mass index (BMI) of ≥30 kg/m2) and 29 normal-weight (BMI of 18.5-24.9 kg/m2) were joined to this study. Plasma levels of IL-1β and IL-10 (pg/mL), CRP (pg/mL), COX-2 (ng/mL), APOA1 (mg/dL), ABCA1 (ng/mL), HDL, Cholesterol, and Triglyceride (TG) (mg/dL), and homocysteine (µmol/L) was measured. Association of these biomarkers with homocysteine was determined. RESULTS Obese subjects had higher serum levels of IL10, IL1β, CRP, COX-2, TG, and cholesterol concentrations (all p<0.05 except IL-10 and cholesterol) and low levels of HDL, APOA1, and ABCA1 (non-significant differences) in comparison to normal-weight group. Homocysteine levels were high in obese men with no significant differences between the two groups. In obese subjects, homocysteine had a significant inverse correlation with APOA1, ABCA1, and HDL, and a strong and moderate positive correlation was found with CRP and TG levels, respectively. CONCLUSIONS High level of homocysteine and its correlation with inflammation proteins and markers in obese subjects appear to be contributed with atherosclerosis development.
Collapse
Affiliation(s)
- Farhad Alizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Mirzaie Bavil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.,Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Fariba Ghiasi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Kolb H. Obese visceral fat tissue inflammation: from protective to detrimental? BMC Med 2022; 20:494. [PMID: 36575472 PMCID: PMC9795790 DOI: 10.1186/s12916-022-02672-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/21/2022] [Indexed: 12/28/2022] Open
Abstract
Obesity usually is accompanied by inflammation of fat tissue, with a prominent role of visceral fat. Chronic inflammation in obese fat tissue is of a lower grade than acute immune activation for clearing the tissue from an infectious agent. It is the loss of adipocyte metabolic homeostasis that causes activation of resident immune cells for supporting tissue functions and regaining homeostasis. Initially, the excess influx of lipids and glucose in the context of overnutrition is met by adipocyte growth and proliferation. Eventual lipid overload of hypertrophic adipocytes leads to endoplasmic reticulum stress and the secretion of a variety of signals causing increased sympathetic tone, lipolysis by adipocytes, lipid uptake by macrophages, matrix remodeling, angiogenesis, and immune cell activation. Pro-inflammatory signaling of adipocytes causes the resident immune system to release increased amounts of pro-inflammatory and other mediators resulting in enhanced tissue-protective responses. With chronic overnutrition, these protective actions are insufficient, and death of adipocytes as well as senescence of several tissue cell types is seen. This structural damage causes the expression or release of immunostimulatory cell components resulting in influx and activation of monocytes and many other immune cell types, with a contribution of stromal cells. Matrix remodeling and angiogenesis is further intensified as well as possibly detrimental fibrosis. The accumulation of senescent cells also may be detrimental via eventual spread of senescence state from affected to neighboring cells by the release of microRNA-containing vesicles. Obese visceral fat inflammation can be viewed as an initially protective response in order to cope with excess ambient nutrients and restore tissue homeostasis but may contribute to tissue damage at a later stage.
Collapse
Affiliation(s)
- Hubert Kolb
- Faculty of Medicine, University of Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany. .,West-German Centre of Diabetes and Health, Düsseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Düsseldorf, Germany.
| |
Collapse
|
50
|
Giacomozzi AS, Carrín ME, Palla CA. Muffins made with monoglyceride oleogels: Impact of fat replacement on sensory properties and fatty acid profile. J AM OIL CHEM SOC 2022. [DOI: 10.1002/aocs.12674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Anabella S. Giacomozzi
- Departamento de Ingeniería Química Universidad Nacional del Sur (UNS) Buenos Aires Argentina
- Planta Piloto de Ingeniería Química ‐ PLAPIQUI (UNS‐CONICET) Buenos Aires Argentina
| | - María E. Carrín
- Departamento de Ingeniería Química Universidad Nacional del Sur (UNS) Buenos Aires Argentina
- Planta Piloto de Ingeniería Química ‐ PLAPIQUI (UNS‐CONICET) Buenos Aires Argentina
| | - Camila A. Palla
- Departamento de Ingeniería Química Universidad Nacional del Sur (UNS) Buenos Aires Argentina
- Planta Piloto de Ingeniería Química ‐ PLAPIQUI (UNS‐CONICET) Buenos Aires Argentina
| |
Collapse
|