1
|
Sertbas M, Ulgen KO. Genome-Scale Metabolic Modeling of Human Pancreas with Focus on Type 2 Diabetes. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:125-138. [PMID: 40068171 DOI: 10.1089/omi.2024.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Type 2 diabetes (T2D) is characterized by relative insulin deficiency due to pancreatic beta cell dysfunction and insulin resistance in different tissues. Not only beta cells but also other islet cells (alpha, delta, and pancreatic polypeptide [PP]) are critical for maintaining glucose homeostasis in the body. In this overarching context and given that a deeper understanding of T2D pathophysiology and novel molecular targets is much needed, studies that integrate experimental and computational biology approaches offer veritable prospects for innovation. In this study, we report on single-cell RNA sequencing data integration with a generic Human1 model to generate context-specific genome-scale metabolic models for alpha, beta, delta, and PP cells for nondiabetic and T2D states and, importantly, at single-cell resolution. Moreover, flux balance analysis was performed for the investigation of metabolic activities in nondiabetic and T2D pancreatic cells. By altering glucose and oxygen uptakes to the metabolic networks, we documented the ways in which hypoglycemia, hyperglycemia, and hypoxia led to changes in metabolic activities in various cellular subsystems. Reporter metabolite analysis revealed significant transcriptional changes around several metabolites involved in sphingolipid and keratan sulfate metabolism in alpha cells, fatty acid metabolism in beta cells, and myoinositol phosphate metabolism in delta cells. Taken together, by leveraging genome-scale metabolic modeling, this research bridges the gap between metabolic theory and clinical practice, offering a comprehensive framework to advance our understanding of pancreatic metabolism in T2D, and contributes new knowledge toward the development of targeted precision medicine interventions.
Collapse
Affiliation(s)
- Mustafa Sertbas
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey
| | - Kutlu O Ulgen
- Department of Chemical Engineering, Bogazici University, Istanbul, Turkey
| |
Collapse
|
2
|
Tang N, He Y, Karatela S, Zhong J, Zeng X, Lu Q, Zhao F, Cai L. Association between erythrocyte polyunsaturated fatty acids and gestational diabetes mellitus in Chinese pregnant women. Eur J Nutr 2025; 64:87. [PMID: 39932572 DOI: 10.1007/s00394-025-03603-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 01/26/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE To explore the associations of erythrocyte polyunsaturated fatty acids (PUFAs) with the risk of gestational diabetes mellitus (GDM) in pregnant women. METHODS A total of 951 women in mid-pregnancy (20-28 weeks of gestation) were recruited during 2017-2018. Erythrocyte PUFAs were measured by gas chromatography and expressed as a percentage of total fatty acids. A 75 g oral glucose tolerance test was used for GDM diagnosis. Logistic regression and restricted cubic spline models were conducted. RESULTS Among the 951 pregnant women, 180 were diagnosed with GDM (18.93%). For n-3 PUFAs, each standard deviation (SD) increase in α-linolenic acid (ALA) and docosapentaenoic acid (DPA) was associated with multivariable-adjusted odds ratios (ORs) of 0.789 (95% CI: 0.649, 0.961; P = 0.018) and 0.782 (95% CI: 0.638, 0.957; P = 0.017), respectively. However, the associations became marginally significant after post hoc false-discovery rate (FDR) correction (both PFDR=0.065). A significant nonlinear association was observed for ALA and GDM risk (P- nonlinearity=0.001). For n-6 PUFAs, γ-linolenic acid (GLA) was significantly associated with a 46.0% higher risk of GDM [OR (95%CI): 1.460 (1.195, 1.785), PFDR=0.003) per SD increase], with a significant non-linear relationship (P- nonlinearity=0.031). Arachidonic acid (AA) showed a borderline significant association with lower GDM risk after FDR correction [OR (95%CI): 0.736 (0.568, 0.953), P = 0.020, PFDR=0.065]. CONCLUSION Erythrocyte GLA was significantly associated with an increased risk of GDM in pregnant women. Furthermore, we observed for the first time significant non-linear associations of erythrocyte ALA and GLA with GDM risk. CLINICAL TRIAL REGISTRY NUMBER NCT03023293, 2019-01-11.
Collapse
Affiliation(s)
- Nu Tang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing, 404100, China
| | - Yannan He
- Institute of Nutrition & Health, Qingdao University, Qingdao, 266071, China
| | - Shamshad Karatela
- Faculty of Health and Behavioural Sciences, Pharmacy Australia Centre of Excellence, University of Queensland, Woolloongabba, QLD, Australia
- Institute of Tropical Health and Medicine (AITHM), James Cook University, Townsville, QLD, Australia
| | | | - Xiaoling Zeng
- Ausnutria Dairy (China) Co., Ltd, Changsha, Hunan Province, 410219, China
| | - Qinggui Lu
- Department of Health Care, Maternal and Child Health Care Hospital of Yuexiu District, Guangzhou, 510080, China
| | - Feng Zhao
- Center of Lipid & Chronic diseases, Suzhou Industrial Technology Research Institute, Zhejiang University, Suzhou, 215163, China.
| | - Li Cai
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Wang X, Shao Q, Gao Y. The emerging role of 12/15-lipoxygenase in ischemic stroke. Brain Res Bull 2025; 221:111194. [PMID: 39788462 DOI: 10.1016/j.brainresbull.2025.111194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/30/2024] [Accepted: 01/04/2025] [Indexed: 01/12/2025]
Abstract
The arachidonic acid metabolic pathway is a classic inflammatory pathway. 12/15-lipoxygenase (LOX), a member of the lipoxygenase family that metabolizes arachidonic acid, has been implicated in the pathogenesis of numerous central nervous system (CNS) diseases. Ischemic stroke is a devastating disease in which the occlusion of cerebral arteries leads to a series of pathophysiological changes in brain tissue, triggering an inflammatory cascade within the brain that results in neuroinflammation. Prior research has shown that 12/15-LOX levels in the brain are elevated following stroke. In this review, we elaborate on the key pathological mechanisms that unfold following ischemic stroke, including neuroinflammation, oxidative stress, neuronal apoptosis, and blood-brain barrier disruption, and present evidence demonstrating that 12/15-LOX inhibition could be used to treat ischemic stroke through various avenues. Furthermore, we list currently available inhibitors of 12/15-LOX and the preclinical or clinical applications, offering novel insights for the early diagnosis, prognosis evaluation, and targeted therapy in neurological diseases.
Collapse
Affiliation(s)
- Xuening Wang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China; Institute of Clinical Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Qiuji Shao
- Department of Cerebrovascular Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Yuan Gao
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China; Institute of Clinical Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
4
|
Almutairi A, White TD, Stephenson DJ, Stephenson BD, Gai-Tusing Y, Goel P, Phillips DW, Welner RS, Lei X, Hammock BD, Chalfant CE, Ramanadham S. Selective Reduction of Ca2+-Independent Phospholipase A2β (iPLA2β)-Derived Lipid Signaling From Macrophages Mitigates Type 1 Diabetes Development. Diabetes 2024; 73:2022-2033. [PMID: 39283670 PMCID: PMC11579405 DOI: 10.2337/db23-0770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 09/07/2024] [Indexed: 11/22/2024]
Abstract
Type 1 diabetes (T1D) is a consequence of autoimmune destruction of β-cells, and macrophages (MΦs) have a central role in initiating processes that lead to β-cell demise. We reported that Ca2+-independent phospholipase A2β (iPLA2β)-derived lipid (iDL) signaling contributes to β-cell death. Because MΦs express iPLA2β, we assessed its role in T1D development. We find that selective reduction of myeloid-iPLA2β in spontaneously diabetes-prone NOD mice 1) decreases proinflammatory eicosanoid production by MΦs, 2) favors the anti-inflammatory (M2-like) MΦ phenotype, and 3) diminishes activated CD4+ and CD8+ T-cells phenotype in the pancreatic infiltrate, prior to T1D onset. These outcomes are associated with a significant reduction in T1D. Further, inhibition of select proinflammatory lipid signaling pathways reduces M1-like MΦ polarization and adoptive transfer of M2-like MΦs reduces NOD T1D incidence, suggesting a mechanism by which iDLs impact T1D development. These findings identify MΦ-iPLA2β as a critical contributor to T1D development and potential target to counter T1D onset. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Abdulaziz Almutairi
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Basic Science, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Tayleur D. White
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel J. Stephenson
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - Benjamin D. Stephenson
- Program in Cancer Biology, UVA Comprehensive Cancer Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Ying Gai-Tusing
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Paran Goel
- Department of Medicine, Hematology & Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel W. Phillips
- Department of Medicine, Hematology & Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Robert S. Welner
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Medicine, Hematology & Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Bruce D. Hammock
- Department of Entomology and Nematology, University of California, Davis, Davis, CA
| | - Charles E. Chalfant
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
- Program in Cancer Biology, UVA Comprehensive Cancer Center, University of Virginia School of Medicine, Charlottesville, VA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
5
|
Holendová B, Stokičová L, Plecitá-Hlavatá L. Lipid Dynamics in Pancreatic β-Cells: Linking Physiology to Diabetes Onset. Antioxid Redox Signal 2024; 41:865-889. [PMID: 39495600 DOI: 10.1089/ars.2024.0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
Significance: Glucose-induced lipid metabolism is essential for preserving functional β-cells, and its disruption is linked to type 2 diabetes (T2D) development. Lipids are an integral part of the cells playing an indispensable role as structural components, energy storage molecules, and signals. Recent Advances: Glucose presence significantly impacts lipid metabolism in β-cells, where fatty acids are primarily synthesized de novo and/or are transported from the bloodstream. This process is regulated by the glycerolipid/free fatty acid cycle, which includes lipogenic and lipolytic reactions producing metabolic coupling factors crucial for insulin secretion. Disrupted lipid metabolism involving oxidative stress and inflammation is a hallmark of T2D. Critical Issues: Lipid metabolism in β-cells is complex involving multiple simultaneous processes. Exact compartmentalization and quantification of lipid metabolism and its intermediates, especially in response to glucose or chronic hyperglycemia, are essential. Current research often uses non-physiological conditions, which may not accurately reflect in vivo situations. Future Directions: Identifying and quantifying individual steps and their signaling, including redox, within the complex fatty acid and lipid metabolic pathways as well as the metabolites formed during acute versus chronic glucose stimulation, will uncover the detailed mechanisms of glucose-stimulated insulin secretion. This knowledge is crucial for understanding T2D pathogenesis and identifying pharmacological targets to prevent this disease. Antioxid. Redox Signal. 41, 865-889.
Collapse
Affiliation(s)
- Blanka Holendová
- Laboratory of Pancreatic Islet Research, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Linda Stokičová
- Laboratory of Pancreatic Islet Research, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
- Charles University, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Laboratory of Pancreatic Islet Research, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
6
|
White TD, Almutairi A, Gai-Tusing Y, Stephenson DJ, Stephenson BD, Chalfant CE, Lei X, Lu B, Hammock BD, DiLorenzo TP, Ramanadham S. Differential lipid signaling from CD4 + and CD8 + T cells contributes to type 1 diabetes development. Front Immunol 2024; 15:1444639. [PMID: 39359722 PMCID: PMC11445035 DOI: 10.3389/fimmu.2024.1444639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/14/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction We reported that Ca2+-independent phospholipase A2β (iPLA2β)-derived lipids (iDLs) contribute to type 1 diabetes (T1D) onset. As CD4+ and CD8+ T cells are critical in promoting β-cell death, we tested the hypothesis that iDL signaling from these cells participates in T1D development. Methods CD4+ and CD8+ T cells from wild-type non-obese diabetic (NOD) and NOD.iPLA2β+/- (NOD.HET) mice were administered in different combinations to immunodeficient NOD.scid. Results In mice receiving only NOD T cells, T1D onset was rapid (5 weeks), incidence 100% by 20 weeks, and islets absent. In contrast, onset was delayed 1 week and incidence reduced 40%-50% in mice receiving combinations that included NOD.HET T cells. Consistently, islets from these non-diabetic mice were devoid of infiltrate and contained insulin-positive β-cells. Reduced iPLA2β led to decreased production of proinflammatory lipids from CD4+ T cells including prostaglandins and dihydroxyeicosatrienoic acids (DHETs), products of soluble epoxide hydrolase (sEH), and inhibition of their signaling decreased (by 82%) IFNγ+CD4+ cells abundance. However, only DHETs production was reduced from CD8+ T cells and was accompanied by decreases in sEH and granzyme B. Discussion These findings suggest that differential select iDL signaling in CD4+ and CD8+ T cells contributes to T1D development, and that therapeutics targeting such signaling might be considered to counter T1D.
Collapse
Affiliation(s)
- Tayleur D. White
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Abdulaziz Almutairi
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Basic Science, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Ying Gai-Tusing
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Daniel J. Stephenson
- Cancer Biology Program, University of Virginia National Cancer Institute (UVA NCI) Comprehensive Cancer Center, University of Virginia-School of Medicine, Charlottesville, VA, United States
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA, United States
| | - Benjamin D. Stephenson
- Cancer Biology Program, University of Virginia National Cancer Institute (UVA NCI) Comprehensive Cancer Center, University of Virginia-School of Medicine, Charlottesville, VA, United States
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA, United States
- Department of Medicine, University of Virginia-School of Medicine, Charlottesville, VA, United States
- Department of Cell Biology, University of Virginia-School of Medicine, Charlottesville, VA, United States
| | - Charles E. Chalfant
- Cancer Biology Program, University of Virginia National Cancer Institute (UVA NCI) Comprehensive Cancer Center, University of Virginia-School of Medicine, Charlottesville, VA, United States
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA, United States
- Department of Medicine, University of Virginia-School of Medicine, Charlottesville, VA, United States
- Department of Cell Biology, University of Virginia-School of Medicine, Charlottesville, VA, United States
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Brian Lu
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bruce D. Hammock
- Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Teresa P. DiLorenzo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, United States
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
7
|
Zhao F, Gong Z, Yang Y, Li X, Chen D, Shi X, Yu T, Wei P. Effects of environmentally relevant concentrations of florfenicol on the glucose metabolism system, intestinal microbiome, and liver metabolome of zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 938:173417. [PMID: 38797401 DOI: 10.1016/j.scitotenv.2024.173417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Florfenicol, a widely used veterinary antibiotic, has now been frequently detected in various water environments and human urines, with high concentrations. Accordingly, the ecological risks and health hazards of florfenicol are attracting increasing attention. In recent years, antibiotic exposure has been implicated in the disruption of animal glucose metabolism. However, the specific effects of florfenicol on the glucose metabolism system and the underlying mechanisms are largely unknown. Herein, zebrafish as an animal model were exposed to environmentally relevant concentrations of florfenicol for 28 days. Using biochemical and molecular analyses, we found that exposure to florfenicol disturbed glucose homeostasis, as evidenced by the abnormal levels of blood glucose and hepatic/muscular glycogen, and the altered expression of genes involved in glycogenolysis, gluconeogenesis, glycogenesis, and glycolysis. Considering the efficient antibacterial activity of florfenicol and the crucial role of intestinal flora in host glucose metabolism, we then analyzed changes in the gut microbiome and its key metabolite short-chain fatty acids (SCFAs). Results indicated that exposure to florfenicol caused gut microbiota dysbiosis, inhibited the production of intestinal SCFAs, and ultimately affected the downstream signaling pathways of SCFA involved in glucose metabolism. Moreover, non-targeted metabolomics revealed that arachidonic acid and linoleic acid metabolic pathways may be associated with insulin sensitivity changes in florfenicol-exposed livers. Overall, this study highlighted a crucial aspect of the environmental risks of florfenicol to both non-target organisms and humans, and presented novel insights into the mechanistic elucidation of metabolic toxicity of antibiotics.
Collapse
Affiliation(s)
- Fei Zhao
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Zhilin Gong
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Yanyu Yang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Xinhui Li
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Dong Chen
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Xueqing Shi
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Tong Yu
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Penghao Wei
- School of Environmental and Municipal Engineering, Qingdao University of Technology, Qingdao 266520, China.
| |
Collapse
|
8
|
Dutta A, Hossain MA, Somadder PD, Moli MA, Ahmed K, Rahman MM, Bui FM. Exploring the therapeutic targets of stevioside in management of type 2 diabetes by network pharmacology and in-silico approach. Diabetes Metab Syndr 2024; 18:103111. [PMID: 39217825 DOI: 10.1016/j.dsx.2024.103111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 07/17/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
AIMS The main objective of the current study is to investigate the pathways and therapeutic targets linked to stevioside in the management of T2D using computational approaches. METHODS We collected RNA-seq datasets from NCBI, then employed GREIN to retrieve differentially expressed genes (DEGs). Computer-assisted techniques DAVID, STRING and NetworkAnalyst were used to explore common significant pathways and therapeutic targets associated with T2D and stevioside. Molecular docking and dynamics simulations were conducted to validate the interaction between stevioside and therapeutic targets. RESULTS Gene ontology and KEGG analysis revealed that prostaglandin synthesis, IL-17 signaling, inflammatory response, and interleukin signaling were potential pathways targeted by stevioside in T2D. Protein-protein interactions (PPI) analysis identified six common hub proteins (PPARG, PTGS2, CXCL8, CCL2, PTPRC, and EDN1). Molecular docking results showed best binding of stevioside to PPARG (-8 kcal/mol) and PTGS2 (-10.1 kcal/mol). Finally, 100 ns molecular dynamics demonstrated that the binding stability between stevioside and target protein (PPARG and PTGS2) falls within the acceptable range. CONCLUSIONS This study reveals that stevioside exhibits significant potential in controlling T2D by targeting key pathways and stably binding to PPARG and PTGS2. Further research is necessary to confirm and expand upon these significant computational results.
Collapse
Affiliation(s)
- Amit Dutta
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Md Arju Hossain
- Department of Microbiology, Primeasia University, Banani, Dhaka, 1213, Bangladesh
| | - Pratul Dipta Somadder
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Mahmuda Akter Moli
- Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Bangladesh
| | - Kawsar Ahmed
- Department of Information and Communication Technology, Mawlana Bhashani Science and Technology University (MBSTU), Santosh, Tangail, 1902, Bangladesh; Department of Electrical and Computer Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, SK, S7N 5A9, Canada; Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Daffodil Smart City (DSC), Birulia, Savar, Dhaka, 1216, Bangladesh.
| | - Md Masuder Rahman
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh.
| | - Francis M Bui
- Department of Electrical and Computer Engineering, University of Saskatchewan, 57 Campus Drive, Saskatoon, SK, S7N 5A9, Canada
| |
Collapse
|
9
|
Zhao L, Yang W, Ji W, Pan Q, Yang J, Cao X. Untargeted metabolomics uncovers metabolic dysregulation and tissue sensitivity in ACE2 knockout mice. Heliyon 2024; 10:e27472. [PMID: 38496880 PMCID: PMC10944221 DOI: 10.1016/j.heliyon.2024.e27472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) polymorphisms are associated with increased risk of type 2 diabetes mellitus (T2DM), obesity and dyslipidemia, which have been determined in various populations. Consistently, ACE2 knockout (ACE2 KO) mice display damaged energy metabolism in multiple tissues, especially the key metabolic tissues such as liver, skeletal muscle and epididymal white adipose tissue (eWAT) and show even more severe phenotype under high-fat diet (HFD) induced metabolic stress. However, the effects of ACE2 on global metabolomics profiling and the tissue sensitivity remain unclear. To understand how tissues independently and collectively respond to ACE2, we performed untargeted metabolomics in serum in ACE2 KO and control wild type (WT) mice both on normal diet (ND) and HFD, and in three key metabolic tissues (liver, skeletal muscle and eWAT) after HFD treatment. The results showed significant alterations in metabolic profiling in ACE2 KO mice. We identified 275 and 168 serum metabolites differing significantly between WT and ACE2 KO mice fed on ND and HFD, respectively. And the altered metabolites in the ACE2 KO group varied from 90 to 196 in liver, muscle and eWAT. The alterations in ND and HFD serum were most similar. Compared with WT mice, ACE2 KO mice showed an increase in N-phenylacetylglutamine (PAGln), methyl indole-3-acetate, 5-hydroxytryptophol, cholic acid, deoxycholic acid and 12(S)-HETE, while LPC (19:0) and LPE (16:1) decreased. Moreover, LPC (20:0), LPC (20:1) and PC (14:0e/6:0) were reduced in both ND and HFD serum, paralleling the decreases identified in HFD skeletal muscle. Interestingly, DL-tryptophan, indole and Gly-Phe decreased in both ND and HFD serum but were elevated in HFD liver of ACE2 KO mice. A low level of l-ergothioneine was observed among liver, muscle, and epididymal fat tissue of ACE2 KO mice. Pathway analysis demonstrated that different tissues exhibited different dysregulated metabolic pathways. In conclusion, these results revealed that ACE2 deficiency leads to an overall state of metabolic distress, which may provide a new insight into the underlying pathogenesis in metabolic disorders in both ACE2 KO mice and in patients with certain genetic variant of ACE2 gene.
Collapse
Affiliation(s)
| | | | - Wenyi Ji
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Qiuyue Pan
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jinkui Yang
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xi Cao
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
10
|
Jiang S, Si J, Mo J, Zhang S, Chen K, Gao J, Xu D, Bai L, Lan G, Liang J. Integrated Microbiome and Serum Metabolome Analysis Reveals Molecular Regulatory Mechanisms of the Average Daily Weight Gain of Yorkshire Pigs. Animals (Basel) 2024; 14:278. [PMID: 38254447 PMCID: PMC10812420 DOI: 10.3390/ani14020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
The average daily weight gain (ADG) is considered a crucial indicator for assessing growth rates in the swine industry. Therefore, investigating the gastrointestinal microbiota and serum metabolites influencing the ADG in pigs is pivotal for swine breed selection. This study involved the inclusion of 350 purebred Yorkshire pigs (age: 90 ± 2 days; body weight: 41.20 ± 4.60 kg). Concurrently, serum and fecal samples were collected during initial measurements of blood and serum indices. The pigs were categorized based on their ADG, with 27 male pigs divided into high-ADG (HADG) and low-ADG (LADG) groups based on their phenotype values. There were 12 pigs in LADG and 15 pigs in HADG. Feces and serum samples were collected on the 90th day. Microbiome and non-targeted metabolomics analyses were conducted using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC-MS). Pearson correlation, with Benjamini-Hochberg (BH) adjustment, was employed to assess the associations between these variables. The abundance of Lactobacillus and Prevotella in LADG was significantly higher than in HADG, while Erysipelothrix, Streptomyces, Dubosiella, Parolsenella, and Adlercreutzia in LADG were significantly lower than in HADG. The concentration of glutamine, etiocholanolone glucuronide, and retinoyl beta-glucuronide in LADG was significantly higher than in HADG, while arachidonic acid, allocholic acid, oleic acid, phenylalanine, and methyltestosterone in LADG were significantly lower than in HADG. The Lactobacillus-Streptomyces networks (Lactobacillus, Streptomyces, methyltestosterone, phenylalanine, oleic acid, arachidonic acid, glutamine, 3-ketosphingosine, L-octanoylcarnitine, camylofin, 4-guanidinobutyrate 3-methylcyclopentadecanone) were identified as the most influential at regulating swine weight gain. These findings suggest that the gastrointestinal tract regulates the daily weight gain of pigs through the network of Lactobacillus and Streptomyces. However, this study was limited to fecal and serum samples from growing and fattening boars. A comprehensive consideration of factors affecting the daily weight gain in pig production, including gender, parity, season, and breed, is warranted.
Collapse
Affiliation(s)
- Shan Jiang
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Jinglei Si
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
- Guangxi State Farms Yongxin Animal Husbandary Group Co., Ltd., Nanning 530022, China
| | - Jiayuan Mo
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Shuai Zhang
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Kuirong Chen
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Jiuyu Gao
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Di Xu
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Lijing Bai
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Ganqiu Lan
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Jing Liang
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| |
Collapse
|
11
|
Iizasa S, Nagao K, Tsuge K, Nagano Y, Yanagita T. Identification of genes regulated by lipids from seaweed Susabinori (Pyropia yezoensis) involved in the improvement of hepatic steatosis: Insights from RNA-Seq analysis in obese db/db mice. PLoS One 2023; 18:e0295591. [PMID: 38085726 PMCID: PMC10715663 DOI: 10.1371/journal.pone.0295591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Hepatic steatosis is an early stage in the progression of non-alcoholic fatty liver disease (NAFLD) and can lead to the development of non-alcoholic steatohepatitis (NASH), a major cause of liver-related morbidity and mortality. Identification of dietary components that can alleviate hepatic steatosis is crucial for developing effective therapeutic strategies for NAFLD. Recently, we demonstrated the impact of lipids extracted from the marine red alga Susabinori (Pyropia yezoensis) in a murine model of type 2-diabete (db/db). We found that Susabinori lipids (SNL), abundant in eicosapentaenoic acid (EPA)-containing polar lipids, protected against obesity-induced hepatic steatosis in db/db mice. To understand the specific genes or biological pathways underlying the effects of SNL, we conducted RNA-Seq analysis of the hepatic transcriptome. By performing comparative analysis of differentially expressed genes between normal mice and db/db mice consuming a control diet, as well as SNL-fed db/db mice, we identified the 15 SNL-dependent up-regulated genes that were down-regulated in db/db mice but up-regulated by SNL feeding. Gene ontology and pathway analysis on these 15 genes demonstrated a significant association with the metabolisms of arachidonic acid (AA) and linoleic acid (LA). Furthermore, we observed alterations in the expression levels of monoacylglycerol lipase (Magl) and fatty acid-binding protein 4 (Fabp4) in the SNL-fed db/db mice, both of which are implicated in AA and LA metabolism. Additionally, the livers of SNL-fed db/db mice exhibited reduced levels of AA and LA, but a high accumulation of EPA. In conclusion, the SNL diet might affect the metabolisms of AA and LA, which contribute to the improvement of hepatic steatosis. Our findings provide insights into the molecular mechanisms underlying the beneficial effects of SNL.
Collapse
Affiliation(s)
- Sayaka Iizasa
- Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Koji Nagao
- Department of Applied Biochemistry and Food Science, Saga University, Saga, Japan
| | | | - Yukio Nagano
- Analytical Research Center for Experimental Sciences, Saga University, Saga, Japan
| | - Teruyoshi Yanagita
- Department of Applied Biochemistry and Food Science, Saga University, Saga, Japan
- Department of Health and Nutrition Science, Nishikyushu University, Saga, Japan
- Saga Foods & Cosmetics Laboratory, Division of Research and Development Promotion, Saga Prefectural Regional Industry Support Center, Saga, Japan
| |
Collapse
|
12
|
Vived C, Lee-Papastavros A, Aparecida da Silva Pereira J, Yi P, MacDonald TL. β Cell Stress and Endocrine Function During T1D: What Is Next to Discover? Endocrinology 2023; 165:bqad162. [PMID: 37947352 DOI: 10.1210/endocr/bqad162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Canonically, type 1 diabetes (T1D) is a disease characterized by autoreactive T cells as perpetrators of endocrine dysfunction and β cell death in the spiral toward loss of β cell mass, hyperglycemia, and insulin dependence. β Cells have mostly been considered as bystanders in a flurry of autoimmune processes. More recently, our framework for understanding and investigating T1D has evolved. It appears increasingly likely that intracellular β cell stress is an important component of T1D etiology/pathology that perpetuates autoimmunity during the progression to T1D. Here we discuss the emerging and complex role of β cell stress in initiating, provoking, and catalyzing T1D. We outline the bridges between hyperglycemia, endoplasmic reticulum stress, oxidative stress, and autoimmunity from the viewpoint of intrinsic β cell (dys)function, and we extend this discussion to the potential role for a therapeutic β cell stress-metabolism axis in T1D. Lastly, we mention research angles that may be pursued to improve β cell endocrine function during T1D. Biology gleaned from studying T1D will certainly overlap to innovate therapeutic strategies for T2D, and also enhance the pursuit of creating optimized stem cell-derived β cells as endocrine therapy.
Collapse
Affiliation(s)
- Celia Vived
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jéssica Aparecida da Silva Pereira
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Peng Yi
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Tara L MacDonald
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
He K, Zhou X, Du H, Zhao J, Deng R, Wang J. A review on the relationship between Arachidonic acid 15-Lipoxygenase (ALOX15) and diabetes mellitus. PeerJ 2023; 11:e16239. [PMID: 37849828 PMCID: PMC10578307 DOI: 10.7717/peerj.16239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Arachidonic acid 15-lipoxygenase (ALOX15), as one of the lipoxygenase family, is mainly responsible for catalyzing the oxidation of various fatty acids to produce a variety of lipid components, contributing to the pathophysiological processes of various immune and inflammatory diseases. Studies have shown that ALOX15 and its related products are widely distributed in human tissues and related to multiple diseases such as liver, cardiovascular, cerebrovascular diseases, diabetes mellitus and other diseases. Diabetes mellitus (DM), the disease studied in this article, is a metabolic disease characterized by a chronic increase in blood glucose levels, which is significantly related to inflammation, oxidative stress, ferroptosis and other mechanisms, and it has a high incidence in the population, accompanied by a variety of complications. Figuring out how ALOX15 is involved in DM is critical to understanding its role in diseases. Therefore, ALOX15 inhibitors or combination therapy containing inhibitors may deliver a novel research direction for the treatment of DM and its complications. This article aims to review the biological effect and the possible function of ALOX15 in the pathogenesis of DM.
Collapse
Affiliation(s)
- Kaiying He
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Xiaochun Zhou
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Hongxuan Du
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jing Zhao
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Rongrong Deng
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jianqin Wang
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| |
Collapse
|
14
|
Zhang Y, Liu Y, Sun J, Zhang W, Guo Z, Ma Q. Arachidonic acid metabolism in health and disease. MedComm (Beijing) 2023; 4:e363. [PMID: 37746665 PMCID: PMC10511835 DOI: 10.1002/mco2.363] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Arachidonic acid (AA), an n-6 essential fatty acid, is a major component of mammalian cells and can be released by phospholipase A2. Accumulating evidence indicates that AA plays essential biochemical roles, as it is the direct precursor of bioactive lipid metabolites of eicosanoids such as prostaglandins, leukotrienes, and epoxyeicosatrienoic acid obtained from three distinct enzymatic metabolic pathways: the cyclooxygenase pathway, lipoxygenase pathway, and cytochrome P450 pathway. AA metabolism is involved not only in cell differentiation, tissue development, and organ function but also in the progression of diseases, such as hepatic fibrosis, neurodegeneration, obesity, diabetes, and cancers. These eicosanoids are generally considered proinflammatory molecules, as they can trigger oxidative stress and stimulate the immune response. Therefore, interventions in AA metabolic pathways are effective ways to manage inflammatory-related diseases in the clinic. Currently, inhibitors targeting enzymes related to AA metabolic pathways are an important area of drug discovery. Moreover, many advances have also been made in clinical studies of AA metabolic inhibitors in combination with chemotherapy and immunotherapy. Herein, we review the discovery of AA and focus on AA metabolism in relation to health and diseases. Furthermore, inhibitors targeting AA metabolism are summarized, and potential clinical applications are discussed.
Collapse
Affiliation(s)
- Yiran Zhang
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Yingxiang Liu
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Jin Sun
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Wei Zhang
- Department of PathologyThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Zheng Guo
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Qiong Ma
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
- Department of PathologyThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| |
Collapse
|
15
|
Miao X, Luo P, Liu J, Wang J, Chen Y. Dihydromyricetin ameliorated nonalcoholic steatohepatitis in mice by regulating the composition of serous lipids, bile acids and ileal microflora. Lipids Health Dis 2023; 22:112. [PMID: 37533083 PMCID: PMC10394885 DOI: 10.1186/s12944-023-01871-7] [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: 03/08/2023] [Accepted: 07/07/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Dihydromyricetin (DMY) is a natural flavonoid with anti-nonalcoholic steatohepatitis (NASH) activity. However, the effects of DMY on the composition of lipids and bile acids (BAs) in serum, and gut microbiota (GM) in ileum of mice with NASH are not clear. METHODS After male C57BL/6 mice was fed with methionine and choline deficiency (MCD) diet and simultaneously administered with DMY (300 mg/kg/day) by gavage for 8 weeks, the pathological changes of liver tissue were observed by Oil Red O, hematoxylin eosin and Masson staining, the levels of serum alaninea minotransferase, aspartate aminotransferase and liver triglyceride, malonic dialdehyde were detected by the detection kits, the composition and contents of serum lipids and BAs were detected by Liquid Chromatograph-Mass Spectrometry, the mRNA levels of hepatic BAs homeostasis-related genes were detected by RT-qPCR, and microbiological diversity in ileum was analyzed by 16S rDNA sequencing. RESULTS The results showed that the significant changes including 29 lipids, 4 BAs (23-nor-deoxycholic acid, ursodeoxycholic acid, 7-ketodeoxycholic acid and cholic acid), 2 BA transporters (Mrp2 and Oatp1b2) and 8 GMs between MCD and DMY groups. Among them, DMY treatment significantly down-regulated 21 lipids, 4 BAs mentioned above, the ratio of Firmicutes/Bacteroidota and the abundance of Erysipelotrichaceae, Faecalibacuium, significantly up-regulated 8 lipids and 5 GMs (Verrucomicrobiota, Bacteroidota, Actinobacteria, Akkermansiaceae and Akkermansia). CONCLUSIONS The results suggested that DMY may alleviate MCD diet-induced NASH through decreasing the serum levels of toxic BAs which regulated by liver Oatp1b2 and Mrp2, regulating the metabolism of related lipids, and up-regulating intestinal probiotics (Actinobacteria and Verrucomicrobiota at the phylum level; Akkermansiaceae at the family level; Akkermansiaat at the genus level) and inhibiting intestinal harmful bacteria (Firmicutes at the phylum level; Erysipelotrichaceae at the family level; Faecalibaculum at the genus level).
Collapse
Affiliation(s)
- Xiaolei Miao
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Ping Luo
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Jiao Liu
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Junjun Wang
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China.
| | - Yong Chen
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
16
|
Ludovico ID, Sarkar S, Elliott E, Virtanen SM, Erlund I, Ramanadham S, Mirmira RG, Metz TO, Nakayasu ES. Fatty acid-mediated signaling as a target for developing type 1 diabetes therapies. Expert Opin Ther Targets 2023; 27:793-806. [PMID: 37706269 PMCID: PMC10591803 DOI: 10.1080/14728222.2023.2259099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/11/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION Type 1 diabetes (T1D) is an autoimmune disease in which pro-inflammatory and cytotoxic signaling drive the death of the insulin-producing β cells. This complex signaling is regulated in part by fatty acids and their bioproducts, making them excellent therapeutic targets. AREAS COVERED We provide an overview of the fatty acid actions on β cells by discussing how they can cause lipotoxicity or regulate inflammatory response during insulitis. We also discuss how diet can affect the availability of fatty acids and disease development. Finally, we discuss development avenues that need further exploration. EXPERT OPINION Fatty acids, such as hydroxyl fatty acids, ω-3 fatty acids, and their downstream products, are druggable candidates that promote protective signaling. Inhibitors and antagonists of enzymes and receptors of arachidonic acid and free fatty acids, along with their derived metabolites, which cause pro-inflammatory and cytotoxic responses, have the potential to be developed as therapeutic targets also. Further, because diet is the main source of fatty acid intake in humans, balancing protective and pro-inflammatory/cytotoxic fatty acid levels through dietary therapy may have beneficial effects, delaying T1D progression. Therefore, therapeutic interventions targeting fatty acid signaling hold potential as avenues to treat T1D.
Collapse
Affiliation(s)
- Ivo Díaz Ludovico
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Emily Elliott
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Suvi M. Virtanen
- Health and Well-Being Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Faculty of Social Sciences, Unit of Health Sciences, Tampere University, Tampere, Finland
- Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Iris Erlund
- Department of Governmental Services, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Thomas O. Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| |
Collapse
|
17
|
Taghiyar S, Pourrajab F, Aarabi MH. Astaxanthin improves fatty acid dysregulation in diabetes by controlling the AMPK-SIRT1 pathway. EXCLI JOURNAL 2023; 22:502-515. [PMID: 37534224 PMCID: PMC10391612 DOI: 10.17179/excli2023-6132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/06/2023] [Indexed: 08/04/2023]
Abstract
Due to the rising prevalence of metabolic disorders, including type 2 diabetes (T2DM), new prevention and treatment strategies are needed. The aim was to examine the effect of astaxanthin (AST) on the major regulatory metabolism pathway SIRT-MAPK and fatty acid (FA) profile of plasma in patients with T2DM. This clinical trial included 68 T2DM patients randomly assigned to receive 10 mg/day of oral AST (n = 34) or placebo (n = 33) for 12 weeks. The expression level of SIRT1, AMPK activity, and the level of fatty acids in the serum were examined. The results showed that AST could modify the serum levels of saturated fatty acids (SFA) and polyunsaturated fatty acids (PUFA), particularly that of Arachidonic acid, from 11.31±0.35 to 8.52±0.72 %. Also, AST increased the expression and activity levels of SIRT1 and AMPK, respectively. Pearson analysis also revealed a significant association between AMPK activity and Linoleic acid serum (LA) levels (~ -0.604, p~0.013). AST can modify the FA profile of plasma by inducing metabolizing cells to uptake them. Also, it can activate the SIRT-AMPK pathway related to metabolism regulation. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Sana Taghiyar
- Department of Clinical Biochemistry, International Campus, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Fatemeh Pourrajab
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Hosein Aarabi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
18
|
Ortiz-Placín C, Castillejo-Rufo A, Estarás M, González A. Membrane Lipid Derivatives: Roles of Arachidonic Acid and Its Metabolites in Pancreatic Physiology and Pathophysiology. Molecules 2023; 28:4316. [PMID: 37298790 PMCID: PMC10254454 DOI: 10.3390/molecules28114316] [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: 04/27/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
One of the most important constituents of the cell membrane is arachidonic acid. Lipids forming part of the cellular membrane can be metabolized in a variety of cellular types of the body by a family of enzymes termed phospholipases: phospholipase A2, phospholipase C and phospholipase D. Phospholipase A2 is considered the most important enzyme type for the release of arachidonic acid. The latter is subsequently subjected to metabolization via different enzymes. Three enzymatic pathways, involving the enzymes cyclooxygenase, lipoxygenase and cytochrome P450, transform the lipid derivative into several bioactive compounds. Arachidonic acid itself plays a role as an intracellular signaling molecule. Additionally, its derivatives play critical roles in cell physiology and, moreover, are involved in the development of disease. Its metabolites comprise, predominantly, prostaglandins, thromboxanes, leukotrienes and hydroxyeicosatetraenoic acids. Their involvement in cellular responses leading to inflammation and/or cancer development is subject to intense study. This manuscript reviews the findings on the involvement of the membrane lipid derivative arachidonic acid and its metabolites in the development of pancreatitis, diabetes and/or pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | - Antonio González
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, 10003 Cáceres, Spain; (C.O.-P.); (A.C.-R.); (M.E.)
| |
Collapse
|
19
|
Tseng WEJ, Chang CW, Hwang JS, Ko PC, Liu CJ, Lim SN. Association of Long-term Antiseizure Medication Use and Incident Type 2 Diabetes Mellitus. Neurology 2023; 100:e2071-e2082. [PMID: 36963840 PMCID: PMC10186244 DOI: 10.1212/wnl.0000000000207222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 02/09/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Diabetes mellitus (DM) contributes significantly to metabolic syndrome and cardiovascular events, and it may be a comorbidity of epilepsy. The objective of this study was to investigate whether long-term antiseizure medication (ASM) use is associated with the risk of developing type 2 diabetes. METHODS We analyzed data from the Chang Gung Research Database. Patients aged ≥45 years who received ASM treatment from January 2001 to May 2019 were identified. Patients with DM-associated diseases and short-term ASM use were excluded. The patients were classified into nonenzyme interaction, enzyme-inducing, enzyme-inhibiting, and mixed ASM groups. The rate of incident diabetes associated with individual ASM was further analyzed. Propensity score weighting was performed to balance between-group differences. Analyses were conducted with Cox proportional regression models and stabilized inverse probability of treatment weighting (IPTW). Hazard ratios (HRs) were calculated at 3, 4, 6, and 9 years after the index date and the end of follow-up. RESULTS A total of 5,103 patients were analyzed, of whom 474 took nonenzyme interaction ASMs, 1,156 took enzyme-inducing ASMs, 336 took enzyme-inhibiting ASMs, and 3,137 took mixed ASMs. During follow-up (39,248 person-years), 663 patients developed new-onset DM, and the prevalence was 13.0%. The incidence of DM plateaued at 6-9 years after ASM initiation. Enzyme-inhibiting ASMs were significantly associated with a higher HR starting at the third year and then throughout the study period. The HRs were 1.93 (95% CI 1.33-2.80), 1.85 (95% CI 1.24-2.75), and 2.08 (95% CI 1.43-3.03) in unadjusted, adjusted, and stabilized IPTW models, respectively, at the end of follow-up. The dosing of ASM did not increase the risk of DM, and none of the individual ASM analyses reached statistical significance. DISCUSSION The long-term use of enzyme-inhibiting ASMs was associated with an increased risk of incident DM, and the risk increased with the duration of treatment. These findings may guide the choice of drugs in those requiring long-term ASM therapy, particularly in high-risk individuals. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that enzyme-inhibiting ASMs were associated with an increased risk of developing DM compared with nonenzyme interaction ASMs.
Collapse
Affiliation(s)
- Wei-En Johnny Tseng
- From the Section of Epilepsy (W.-E.J.T., C.-W.C., C.-J.L., S.-N.L.), Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, and Chang Gung University College of Medicine; PhD Program in Biomedical Engineering (W.-E.J.T.), Chang Gung University; Division of Endocrinology and Metabolism (J.-S.H.), Department of Internal Medicine, Linkou Chang Gung Memorial Hospital; and Center for Big Data Analytics and Statistics (P.-C.K.), Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Wei Chang
- From the Section of Epilepsy (W.-E.J.T., C.-W.C., C.-J.L., S.-N.L.), Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, and Chang Gung University College of Medicine; PhD Program in Biomedical Engineering (W.-E.J.T.), Chang Gung University; Division of Endocrinology and Metabolism (J.-S.H.), Department of Internal Medicine, Linkou Chang Gung Memorial Hospital; and Center for Big Data Analytics and Statistics (P.-C.K.), Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jawl-Shan Hwang
- From the Section of Epilepsy (W.-E.J.T., C.-W.C., C.-J.L., S.-N.L.), Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, and Chang Gung University College of Medicine; PhD Program in Biomedical Engineering (W.-E.J.T.), Chang Gung University; Division of Endocrinology and Metabolism (J.-S.H.), Department of Internal Medicine, Linkou Chang Gung Memorial Hospital; and Center for Big Data Analytics and Statistics (P.-C.K.), Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Po-Chuan Ko
- From the Section of Epilepsy (W.-E.J.T., C.-W.C., C.-J.L., S.-N.L.), Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, and Chang Gung University College of Medicine; PhD Program in Biomedical Engineering (W.-E.J.T.), Chang Gung University; Division of Endocrinology and Metabolism (J.-S.H.), Department of Internal Medicine, Linkou Chang Gung Memorial Hospital; and Center for Big Data Analytics and Statistics (P.-C.K.), Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Jing Liu
- From the Section of Epilepsy (W.-E.J.T., C.-W.C., C.-J.L., S.-N.L.), Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, and Chang Gung University College of Medicine; PhD Program in Biomedical Engineering (W.-E.J.T.), Chang Gung University; Division of Endocrinology and Metabolism (J.-S.H.), Department of Internal Medicine, Linkou Chang Gung Memorial Hospital; and Center for Big Data Analytics and Statistics (P.-C.K.), Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Siew-Na Lim
- From the Section of Epilepsy (W.-E.J.T., C.-W.C., C.-J.L., S.-N.L.), Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, and Chang Gung University College of Medicine; PhD Program in Biomedical Engineering (W.-E.J.T.), Chang Gung University; Division of Endocrinology and Metabolism (J.-S.H.), Department of Internal Medicine, Linkou Chang Gung Memorial Hospital; and Center for Big Data Analytics and Statistics (P.-C.K.), Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
20
|
Ağagündüz D, Icer MA, Yesildemir O, Koçak T, Kocyigit E, Capasso R. The roles of dietary lipids and lipidomics in gut-brain axis in type 2 diabetes mellitus. J Transl Med 2023; 21:240. [PMID: 37009872 PMCID: PMC10068184 DOI: 10.1186/s12967-023-04088-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/25/2023] [Indexed: 04/04/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), one of the main types of Noncommunicable diseases (NCDs), is a systemic inflammatory disease characterized by dysfunctional pancreatic β-cells and/or peripheral insulin resistance, resulting in impaired glucose and lipid metabolism. Genetic, metabolic, multiple lifestyle, and sociodemographic factors are known as related to high T2DM risk. Dietary lipids and lipid metabolism are significant metabolic modulators in T2DM and T2DM-related complications. Besides, accumulated evidence suggests that altered gut microbiota which plays an important role in the metabolic health of the host contributes significantly to T2DM involving impaired or improved glucose and lipid metabolism. At this point, dietary lipids may affect host physiology and health via interaction with the gut microbiota. Besides, increasing evidence in the literature suggests that lipidomics as novel parameters detected with holistic analytical techniques have important roles in the pathogenesis and progression of T2DM, through various mechanisms of action including gut-brain axis modulation. A better understanding of the roles of some nutrients and lipidomics in T2DM through gut microbiota interactions will help develop new strategies for the prevention and treatment of T2DM. However, this issue has not yet been entirely discussed in the literature. The present review provides up-to-date knowledge on the roles of dietary lipids and lipidomics in gut-brain axis in T2DM and some nutritional strategies in T2DM considering lipids- lipidomics and gut microbiota interactions are given.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490, Ankara, Turkey.
| | - Mehmet Arif Icer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Amasya University, 05100, Amasya, Turkey
| | - Ozge Yesildemir
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Bursa Uludag University, 16059, Bursa, Turkey
| | - Tevfik Koçak
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490, Ankara, Turkey
| | - Emine Kocyigit
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ordu University, 52200, Ordu, Turkey
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, 80055, Naples, Italy.
| |
Collapse
|
21
|
Watkins BA, Friedman AN, Kim J, Borkowski K, Kaiser S, Fiehn O, Newman JW. Blood Levels of Endocannabinoids, Oxylipins, and Metabolites Are Altered in Hemodialysis Patients. Int J Mol Sci 2022; 23:ijms23179781. [PMID: 36077177 PMCID: PMC9456435 DOI: 10.3390/ijms23179781] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
Hemodialysis patients (HDPs) have higher blood pressure, higher levels of inflammation, a higher risk of cardiovascular disease, and unusually low plasma n-3 polyunsaturated fatty acid (PUFA) levels compared to healthy subjects. The objective of our investigation was to examine the levels of endocannabinoids (eCBs) and oxylipins (OxLs) in female HDPs compared to healthy matched female controls, with the underlying hypothesis that differences in specific PUFA levels in hemodialysis patients would result in changes in eCBs and OxLs. Plasma phospholipid fatty acids were analyzed by gas chromatography. Plasma was extracted and analyzed using ultra-performance liquid chromatography followed by electrospray ionization and tandem MS for eCBs and OxLs. The global untargeted metabolite profiling of plasma was performed by GCTOF MS. Compared to the controls, HDPs showed lower levels of plasma EPA and the associated OxL metabolites 5- and 12-HEPE, 14,15-DiHETE, as well as DHA derived 19(20)-EpDPE. Meanwhile, no changes in arachidonylethanolamide or 2-arachidonylglycerol in the open circulation were detected. Higher levels of multiple N-acylethanolamides, monoacylglycerols, biomarkers of progressive kidney disease, the nitric oxide metabolism-linked citrulline, and the uremic toxins kynurenine and creatine were observed in HDP. These metabolic differences in cCBs and OxLs help explain the severe inflammatory and cardiovascular disease manifested by HDPs, and they should be explored in future studies.
Collapse
Affiliation(s)
- Bruce A. Watkins
- Department of Nutrition, University of California, Davis, CA 95616, USA
- Correspondence:
| | - Allon N. Friedman
- University Hospital, Suite 6100, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jeffrey Kim
- Department of Internal Medicine, University of California, Davis, CA 95616, USA
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California, Davis, CA 95616, USA
| | | | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA 95616, USA
| | - John W. Newman
- Obesity and Metabolism Research Unit, USDA-ARS Agriculture Research Service, Davis, CA 95616, USA
| |
Collapse
|
22
|
Tans R, Dey S, Dey NS, Cao JH, Paul PS, Calder G, O’Toole P, Kaye PM, Heeren RMA. Mass spectrometry imaging identifies altered hepatic lipid signatures during experimental Leishmania donovani infection. Front Immunol 2022; 13:862104. [PMID: 36003389 PMCID: PMC9394181 DOI: 10.3389/fimmu.2022.862104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Spatial analysis of lipids in inflammatory microenvironments is key to understand the pathogenesis of infectious disease. Granulomatous inflammation is a hallmark of leishmaniasis and changes in host and parasite lipid metabolism have been observed at the bulk tissue level in various infection models. Here, mass spectrometry imaging (MSI) is applied to spatially map hepatic lipid composition following infection with Leishmania donovani, an experimental mouse model of visceral leishmaniasis. Methods Livers from naïve and L. donovani-infected C57BL/6 mice were harvested at 14- and 20-days post-infection (n=5 per time point). 12 µm transverse sections were cut and covered with norhamane, prior to lipid analysis using MALDI-MSI. MALDI-MSI was performed in negative mode on a Rapiflex (Bruker Daltonics) at 5 and 50 µm spatial resolution and data-dependent analysis (DDA) on an Orbitrap-Elite (Thermo-Scientific) at 50 µm spatial resolution for structural identification analysis of lipids. Results Aberrant lipid abundances were observed in a heterogeneous distribution across infected mouse livers compared to naïve mouse liver. Distinctive localized correlated lipid masses were found in granulomas and surrounding parenchymal tissue. Structural identification revealed 40 different lipids common to naïve and d14/d20 infected mouse livers, whereas 15 identified lipids were only detected in infected mouse livers. For pathology-guided MSI imaging, we deduced lipids from manually annotated granulomatous and parenchyma regions of interests (ROIs), identifying 34 lipids that showed significantly different intensities between parenchyma and granulomas across all infected livers. Discussion Our results identify specific lipids that spatially correlate to the major histopathological feature of Leishmania donovani infection in the liver, viz. hepatic granulomas. In addition, we identified a three-fold increase in the number of unique phosphatidylglycerols (PGs) in infected liver tissue and provide direct evidence that arachidonic acid-containing phospholipids are localized with hepatic granulomas. These phospholipids may serve as important precursors for downstream oxylipin generation with consequences for the regulation of the inflammatory cascade. This study provides the first description of the use of MSI to define spatial-temporal lipid changes at local sites of infection induced by Leishmania donovani in mice.
Collapse
Affiliation(s)
- Roel Tans
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - Shoumit Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Nidhi Sharma Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Jian-Hua Cao
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - Prasanjit S. Paul
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - Grant Calder
- Department of Biology, University of York, York, United Kingdom
| | - Peter O’Toole
- Department of Biology, University of York, York, United Kingdom
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
- *Correspondence: Paul M. Kaye, ; Ron M. A. Heeren,
| | - Ron M. A. Heeren
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
- *Correspondence: Paul M. Kaye, ; Ron M. A. Heeren,
| |
Collapse
|
23
|
Arriaga MB, Karim F, Queiroz ATL, Araújo-Pereira M, Barreto-Duarte B, Sales C, Moosa MYS, Mazibuko M, Milne GL, Maruri F, Serezani CH, Koethe JR, Figueiredo MC, Kritski AL, Cordeiro-Santos M, Rolla VC, Sterling TR, Leslie A, Andrade BB. Effect of Dysglycemia on Urinary Lipid Mediator Profiles in Persons With Pulmonary Tuberculosis. Front Immunol 2022; 13:919802. [PMID: 35874781 PMCID: PMC9304990 DOI: 10.3389/fimmu.2022.919802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Oxidized lipid mediators such as eicosanoids play a central role in the inflammatory response associated with tuberculosis (TB) pathogenesis. Diabetes mellitus (DM) leads to marked changes in lipid mediators in persons with TB. However, the associations between diabetes-related changes in lipid mediators and clearance of M. tuberculosis (Mtb) among persons on anti-TB treatment (ATT) are unknown. Quantification of urinary eicosanoid metabolites can provide insights into the circulating lipid mediators involved in Mtb immune responses. Methods We conducted a multi-site prospective observational study among adults with drug-sensitive pulmonary TB and controls without active TB; both groups had sub-groups with or without dysglycemia at baseline. Participants were enrolled from RePORT-Brazil (Salvador site) and RePORT-South Africa (Durban site) and stratified according to TB status and baseline glycated hemoglobin levels: a) TB-dysglycemia (n=69); b) TB-normoglycemia (n=64); c) non-TB/dysglycemia (n=31); d) non-TB/non-dysglycemia (n=29). We evaluated the following urinary eicosanoid metabolites: 11α-hydroxy-9,15-dioxo-2,3,4,5-tetranor-prostane-1,20-dioic acid (major urinary metabolite of prostaglandin E2, PGE-M), tetranor-PGE1 (metabolite of PGE2, TN-E), 9α-hydroxy-11,15-dioxo-2,3,4,5-tetranor-prostane-1,20-dioic acid (metabolite of PGD2, PGD-M), 11-dehydro-thromboxane B2 (11dTxB2), 2,3-dinor-6-keto-PGF1α (prostaglandin I metabolite, PGI-M), and leukotriene E4 (LTE4). Comparisons between the study groups were performed at three time points: before ATT and 2 and 6 months after initiating therapy. Results PGE-M and LTE4 values were consistently higher at all three time-points in the TB-dysglycemia group compared to the other groups (p<0.001). In addition, there was a significant decrease in PGI-M and LTE4 levels from baseline to month 6 in the TB-dysglycemia and TB-normoglycemia groups. Finally, TB-dysglycemia was independently associated with increased concentrations of PGD-M, PGI-M, and LTE4 at baseline in a multivariable model adjusting for age, sex, BMI, and study site. These associations were not affected by HIV status. Conclusion The urinary eicosanoid metabolite profile was associated with TB-dysglycemia before and during ATT. These observations can help identify the mechanisms involved in the pathogenesis of TB-dysglycemia, and potential biomarkers of TB treatment outcomes, including among persons with dysglycemia.
Collapse
Affiliation(s)
- María B Arriaga
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil.,Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Farina Karim
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Artur T L Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Center of Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Mariana Araújo-Pereira
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil
| | - Beatriz Barreto-Duarte
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Brazil
| | - Caio Sales
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Brazil
| | | | - Matilda Mazibuko
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Ginger L Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Fernanda Maruri
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Carlos Henrique Serezani
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - John R Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Marina C Figueiredo
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Afrânio L Kritski
- Programa Acadêmico de Tuberculose da Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Cordeiro-Santos
- Fundação Medicina Tropical Dr Heitor Vieira Dourado, Manaus, Brazil.,Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil.,Universidade Federal do Amazonas, Manaus, Brazil
| | - Valeria C Rolla
- Laboratório de Pesquisa Clínica em Micobacteriose, Instituto Nacional de Infectologia Evandro Chagas, Fiocruz, Rio de Janeiro, Brazil
| | - Timothy R Sterling
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa.,Division of Infection and Immunity, University College London, London, United Kingdom
| | - Bruno B Andrade
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil.,Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Brazil.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States.,Curso de Medicina, Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| |
Collapse
|
24
|
Liu Y, Xia YY, Zhang T, Yang Y, Cannon RD, Mansell T, Novakovic B, Saffery R, Han TL, Zhang H, Baker PN. Complex Interactions Between Circulating Fatty Acid Levels, Desaturase Activities, and the Risk of Gestational Diabetes Mellitus: A Prospective Cohort Study. Front Nutr 2022; 9:919357. [PMID: 35898714 PMCID: PMC9313599 DOI: 10.3389/fnut.2022.919357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveMaternal abnormal fatty acid desaturation has previously been linked to gestational diabetes mellitus (GDM). However, few studies have investigated this relationship longitudinally throughout pregnancy. In this study, we investigated the relationship between GDM and desaturase activities across the pregnancy trimesters.MethodsA total of 661 women (GDM = 189, non-GDM = 472) were selected from the Complex Lipids in Mothers and Babies (CLIMB) cohort study. Clinical information and maternal serum were collected at 11–14, 22–28, and 32–34 weeks of gestation. Totally, 20 serum fatty acids were quantified using gas chromatography–mass spectrometry (GC-MS) analysis at each timepoint. Polyunsaturated fatty acid (PUFA) product-to-precursor ratios were used to estimate desaturase and elongase activities including delta-5 desaturase, delta-6 desaturase, stearoyl-CoA desaturase, and elongase.ResultsAfter adjusting for major potential confounders including maternal age, BMI, primiparity, smoking, and alcohol consumption, we observed a significant increase in the levels of γ-linolenic acid (GLA) and eicosatrienoic acid (DGLA) in the first trimester of women with GDM, whereas GLA and DGLA were reduced in the third trimester, when compared to the non-GDM group. Arachidonic acid (AA) showed an upward trend in the GDM group throughout pregnancy. Estimated delta-6 desaturase and delta-5 desaturase activity were elevated in the first trimester (OR = 1.40, 95% CI 1.03–1.91; OR = 0.56, 95% CI 0.32–0.96) but attenuated in the third trimester (OR = 0.78, 95% CI 0.58–1.07; OR = 2.64, 95% CI 1.46–4.78) in GDM pregnancies, respective to controls. Estimated delta-9–18 desaturase activity (OR = 3.70, 95% CI 1.49–9.19) was increased in women with GDM in later pregnancy.ConclusionsOur study highlights the potential importance of fatty acid desaturase activities, particularly estimated delta-5 desaturase and delta-9–18 desaturase in the pathophysiology of GDM. These findings may have applications for the early diagnosis and management of GDM.
Collapse
Affiliation(s)
- Yue Liu
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
- Mass Spectrometry Center of Maternal Fetal Medicine, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yin-Yin Xia
- Department of Occupational and Environmental Hygiene, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
- Mass Spectrometry Center of Maternal Fetal Medicine, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Ting Zhang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Yang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Mass Spectrometry Center of Maternal Fetal Medicine, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Richard D. Cannon
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Toby Mansell
- Molecular Immunity, Murdoch Childrens Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Boris Novakovic
- Molecular Immunity, Murdoch Childrens Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Richard Saffery
- Molecular Immunity, Murdoch Childrens Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Ting-Li Han
- Mass Spectrometry Center of Maternal Fetal Medicine, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Ting-Li Han
| | - Hua Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Mass Spectrometry Center of Maternal Fetal Medicine, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- *Correspondence: Hua Zhang
| | - Philip N. Baker
- College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
25
|
Hu B, Yang H, Chen G, Sun X, Zou X, Ma J, Yao X, Liang Q, Liu H. Structural characterization and preventive effect on non-alcoholic fatty liver disease of oligosaccharides from Bletilla striata. Food Funct 2022; 13:4757-4769. [PMID: 35389416 DOI: 10.1039/d1fo03899k] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, Bletilla striata polysaccharides were degraded into oligosaccharides. The structural features were analyzed by HPLC, HPLC-MS, FT-IR, and NMR spectroscopy. The results indicated that Bletilla striata oligosaccharides (BOs) were composed of mannose and glucose with a molar ratio of 5.2 : 1, and the main backbones of BOs contained (1→4)-linked-α-D-Man, (1→2)-linked-α-D-Man, and (1→2)-linked-α-D-Glc. By using a high-fat diet (HFD)-induced mouse model, we demonstrated that BOs had an improving effect on non-alcoholic fatty liver disease (NAFLD). Using the metabolomics assay, we found that BOs significantly regulated the hepatic metabolism of fatty acids, arachidonic acid, and other related metabolites in HFD-fed mice, accompanied by the reduction of lipid accumulation and fibrosis in liver tissues. In summary, BOs displayed high potential for the treatment of NAFLD as a functional food.
Collapse
Affiliation(s)
- Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| | - Guangming Chen
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| | - Xiongjie Sun
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| | - Xiaojuan Zou
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| | - Jun Ma
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| | - Xiaowei Yao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| | - Qiong Liang
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, P. R. China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, P. R. China.
| |
Collapse
|
26
|
Reinicke M, Shamkeeva S, Hell M, Isermann B, Ceglarek U, Heinemann ML. Targeted Lipidomics for Characterization of PUFAs and Eicosanoids in Extracellular Vesicles. Nutrients 2022; 14:nu14071319. [PMID: 35405932 PMCID: PMC9000901 DOI: 10.3390/nu14071319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/16/2022] [Accepted: 03/19/2022] [Indexed: 02/06/2023] Open
Abstract
Lipids are increasingly recognized as bioactive mediators of extracellular vesicle (EV) functions. However, while EV proteins and nucleic acids are well described, EV lipids are insufficiently understood due to lack of adequate quantitative methods. We adapted an established targeted and quantitative mass spectrometry (LC-MS/MS) method originally developed for analysis of 94 eicosanoids and seven polyunsaturated fatty acids (PUFA) in human plasma. Additionally, the influence of freeze–thaw (FT) cycles, injection volume, and extraction solvent were investigated. The modified protocol was applied to lipidomic analysis of differently polarized macrophage-derived EVs. We successfully quantified three PUFAs and eight eicosanoids within EVs. Lipid extraction showed reproducible PUFA and eicosanoid patterns. We found a particularly high impact of FT cycles on EV lipid profiles, with significant reductions of up to 70%. Thus, repeated FT will markedly influence analytical results and may alter EV functions, emphasizing the importance of a standardized sample pretreatment protocol for the analysis of bioactive lipids in EVs. EV lipid profiles differed largely depending on the polarization of the originating macrophages. Particularly, we observed major changes in the arachidonic acid pathway. We emphasize the importance of a standardized sample pretreatment protocol for the analysis of bioactive lipids in EVs.
Collapse
|
27
|
Tuomisto K, Palmu J, Long T, Watrous JD, Mercader K, Lagerborg KA, Andres A, Salmi M, Jalkanen S, Vasan RS, Inouye M, Havulinna AS, Tuomilehto J, Jousilahti P, Niiranen TJ, Cheng S, Jain M, Salomaa V. A plasma metabolite score of three eicosanoids predicts incident type 2 diabetes: a prospective study in three independent cohorts. BMJ Open Diabetes Res Care 2022; 10:10/2/e002519. [PMID: 35361620 PMCID: PMC8971778 DOI: 10.1136/bmjdrc-2021-002519] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/18/2022] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Peptide markers of inflammation have been associated with the development of type 2 diabetes. The role of upstream, lipid-derived mediators of inflammation such as eicosanoids, remains less clear. The aim of this study was to examine whether eicosanoids are associated with incident type 2 diabetes. RESEARCH DESIGN & METHODS In the FINRISK (Finnish Cardiovascular Risk Study) 2002 study, a population-based sample of Finnish men and women aged 25-74 years, we used directed, non-targeted liquid chromatography-mass spectrometry to identify 545 eicosanoids and related oxylipins in the participants' plasma samples (n=8292). We used multivariable-adjusted Cox regression to examine associations between eicosanoids and incident type 2 diabetes. The significant independent findings were replicated in the Framingham Heart Study (FHS, n=2886) and DIetary, Lifestyle and Genetic determinants of Obesity and Metabolic syndrome (DILGOM) 2007 (n=3905). Together, these three cohorts had 1070 cases of incident type 2 diabetes. RESULTS In the FINRISK 2002 cohort, 76 eicosanoids were associated individually with incident type 2 diabetes. We identified three eicosanoids independently associated with incident type 2 diabetes using stepwise Cox regression with forward selection and a Bonferroni-corrected inclusion threshold. A three-eicosanoid risk score produced an HR of 1.56 (95% CI 1.41 to 1.72) per 1 SD increment for risk of incident diabetes. The HR for comparing the top quartile with the lowest was 2.80 (95% CI 2.53 to 3.07). In the replication analyses, the three-eicosanoid risk score was significant in FHS (HR 1.24 (95% CI 1.10 to 1.39, p<0.001)) and directionally consistent in DILGOM (HR 1.12 (95% CI 0.99 to 1.27, p=0.07)). Meta-analysis of the three cohorts yielded a pooled HR of 1.31 (95% CI 1.05 to 1.56). CONCLUSIONS Plasma eicosanoid profiles predict incident type 2 diabetes and the clearest signals replicate in three independent cohorts. Our findings give new information on the biology underlying type 2 diabetes and suggest opportunities for early identification of people at risk.
Collapse
Affiliation(s)
- Karolina Tuomisto
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Joonatan Palmu
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Tao Long
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Jeramie D Watrous
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Kysha Mercader
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Kim A Lagerborg
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Allen Andres
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Marko Salmi
- MediCity, InFLAMES Flagship, and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- MediCity, InFLAMES Flagship, and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ramachandran S Vasan
- Boston University's and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, USA
- Sections of Preventive Medicine and Epidemiology, and Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Aki S Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Teemu J Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mohit Jain
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
28
|
Giurdanella G, Longo A, Distefano A, Olivieri M, Cristaldi M, Cosentino A, Agafonova A, Caporarello N, Lupo G, Anfuso CD. The Anti-Inflammatory Effect of the β1-Adrenergic Receptor Antagonist Metoprolol on High Glucose Treated Human Microvascular Retinal Endothelial Cells. Cells 2021; 11:cells11010051. [PMID: 35011613 PMCID: PMC8750370 DOI: 10.3390/cells11010051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
Hyperglycemia-induced impairment of the blood-retinal barrier represents the main pathological event in diabetic retinopathy that is elicited by a reduced cellular response to an accumulation of reactive oxygen species (ROS) and increased inflammation. The purpose of the study was to evaluate whether the selective β1-adrenoreceptor (β1-AR) antagonist metoprolol could modulate the inflammatory response to hyperglycemic conditions. For this purpose, human retinal endothelial cells (HREC) were treated with normal (5 mM) or high glucose (25 mM, HG) in the presence of metoprolol (10 μM), epinephrine (1 μM), or both compounds. Metoprolol prevented both the HG-induced reduction of cell viability (MTT assays) and the modulation of the angiogenic potential of HREC (tube formation assays) reducing the TNF-α, IL-1β, and VEGF mRNA levels (qRT-PCR). Moreover, metoprolol prevented the increase in phospho-ERK1/2, phospho-cPLA2, COX2, and protein levels (Western blot) as well as counteracting the translocation of ERK1/2 and cPLA2 (high-content screening). Metoprolol reduced ROS accumulation in HG-stimulated HREC by activating the anti-oxidative cellular response mediated by the Keap1/Nrf2/HO-1 pathway. In conclusion, metoprolol exerted a dual effect on HG-stimulated HREC, decreasing the activation of the pro-inflammatory ERK1/2/cPLA2/COX2 axis, and counteracting ROS accumulation by activating the Keap1/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Giovanni Giurdanella
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (G.G.); (A.L.); (A.D.); (A.C.); (A.A.); (C.D.A.)
| | - Anna Longo
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (G.G.); (A.L.); (A.D.); (A.C.); (A.A.); (C.D.A.)
| | - Alfio Distefano
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (G.G.); (A.L.); (A.D.); (A.C.); (A.A.); (C.D.A.)
| | - Melania Olivieri
- U.O. Clinical Pathology, Department of Hematology, AUSL Romagna, 47522 Cesena, Italy;
| | | | - Alessia Cosentino
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (G.G.); (A.L.); (A.D.); (A.C.); (A.A.); (C.D.A.)
| | - Aleksandra Agafonova
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (G.G.); (A.L.); (A.D.); (A.C.); (A.A.); (C.D.A.)
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA;
| | - Gabriella Lupo
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (G.G.); (A.L.); (A.D.); (A.C.); (A.A.); (C.D.A.)
- Correspondence:
| | - Carmelina Daniela Anfuso
- Biochemistry Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (G.G.); (A.L.); (A.D.); (A.C.); (A.A.); (C.D.A.)
| |
Collapse
|
29
|
Khan AH, Hwang SH, Barnett SD, Burkhan A, Jankiewicz WK, Hammock BD, Imig JD. Multitarget molecule, PTUPB, to treat diabetic nephropathy in rats. Br J Pharmacol 2021; 178:4468-4484. [PMID: 34255857 PMCID: PMC8863090 DOI: 10.1111/bph.15623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Diabetic nephropathy is a common complications related to high morbidity and mortality in type 2 diabetes. We investigated the action of the dual modulator, PTUPB, a soluble epoxide hydrolase and cyclooxygenase-2 inhibitor against diabetic nephropathy. EXPERIMENTAL APPROACH Sixteen-week-old type 2 diabetic and proteinuric obese ZSF1 rats were treated with vehicle, PTUPB or enalapril for 8 weeks. Measurements were made of epoxyeicosatrienoic acids, thromboxane B2 (TBX2 ) and prostaglandin E2 (PGE2 ) in the kidney of these and lean ZSF1 rats along with their blood pressure. KEY RESULT Obese ZSF1 rats were diabetic with fivefold higher fasting blood glucose levels and markedly higher HbA1c levels compared with lean ZSF1 rats. PTUPB nor enalapril reduced fasting blood glucose or HbA1c but alleviated the development of diabetic nephropathy. In PTUPB-treated obese ZSF1 rats, glomerular nephrin expression was preserved. Enalapril also alleviated diabetic nephropathy. Diabetic renal injury in obese ZSF1 rats was accompanied by renal inflammation with six to sevenfold higher urinary MCP-1 (CCR2) level and renal infiltration of CD-68 positive cells. PTUPB and enalapril significantly reduced urinary MCP-1 levels and renal mRNA expression of cytokines. Both PTUPB and enalapril lowered blood pressure. PTUPB but not enalapril decreased hyperlipidaemia and liver injury in obese ZSF1 rats. CONCLUSION AND IMPLICATIONS Overall, the dual modulator PTUPB does not treat hyperglycaemia but can effectively alleviate hypertension, diabetic nephropathy, hyperlipidaemia and liver injury in type 2 diabetic rats. Our data further demonstrate that the renal actions of PTUPB are comparable with a current standard diabetic nephropathy treatment.
Collapse
Affiliation(s)
- Abdul Hye Khan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Scott D. Barnett
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anna Burkhan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wojciech K. Jankiewicz
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Bruce D. Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - John D. Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
30
|
Ali T, Lei X, Barbour SE, Koizumi A, Chalfant CE, Ramanadham S. Alterations in β-Cell Sphingolipid Profile Associated with ER Stress and iPLA 2β: Another Contributor to β-Cell Apoptosis in Type 1 Diabetes. Molecules 2021; 26:molecules26216361. [PMID: 34770770 PMCID: PMC8587436 DOI: 10.3390/molecules26216361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) development, in part, is due to ER stress-induced β-cell apoptosis. Activation of the Ca2+-independent phospholipase A2 beta (iPLA2β) leads to the generation of pro-inflammatory eicosanoids, which contribute to β-cell death and T1D. ER stress induces iPLA2β-mediated generation of pro-apoptotic ceramides via neutral sphingomyelinase (NSMase). To gain a better understanding of the impact of iPLA2β on sphingolipids (SLs), we characterized their profile in β-cells undergoing ER stress. ESI/MS/MS analyses followed by ANOVA/Student’s t-test were used to assess differences in sphingolipids molecular species in Vector (V) control and iPLA2β-overexpressing (OE) INS-1 and Akita (AK, spontaneous model of ER stress) and WT-littermate (AK-WT) β-cells. As expected, iPLA2β induction was greater in the OE and AK cells in comparison with V and WT cells. We report here that ER stress led to elevations in pro-apoptotic and decreases in pro-survival sphingolipids and that the inactivation of iPLA2β restores the sphingolipid species toward those that promote cell survival. In view of our recent finding that the SL profile in macrophages—the initiators of autoimmune responses leading to T1D—is not significantly altered during T1D development, we posit that the iPLA2β-mediated shift in the β-cell sphingolipid profile is an important contributor to β-cell death associated with T1D.
Collapse
Affiliation(s)
- Tomader Ali
- Research Department, Imperial College London Diabetes Center, Abu Dhabi 51133, United Arab Emirates;
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Suzanne E. Barbour
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Akio Koizumi
- Department of Health and Environmental Sciences, Kyoto Graduate School of Medicine, Kyoto 606-8501, Japan;
| | - Charles E. Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA;
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: ; Tel.: +1-205-996-5973; Fax: +1-205-996-5220
| |
Collapse
|
31
|
Razdan A, Main NM, Chiu V, Shackel NA, de Souza P, Bryant K, Scott KF. Targeting the eicosanoid pathway in hepatocellular carcinoma. Am J Cancer Res 2021; 11:2456-2476. [PMID: 34249410 PMCID: PMC8263695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/06/2021] [Indexed: 06/13/2023] Open
Abstract
Liver cancer has variable incidence worldwide and high mortality. Histologically, the most common subtype of liver cancer is hepatocellular carcinoma (HCC). Approximately 30-40% of HCC patients are diagnosed at an advanced stage, and at present, there are limited treatment options for such patients. The current first-line therapy with tyrosine kinase inhibitors, sorafenib or lenvatinib, prolongs survival by a median of about 2.5-3 months after which the disease normally progresses. Additionally, many patients discontinue the use of tyrosine kinase inhibitors due to toxicity or may not be suitable candidates due to co-morbidity or frailty. It is, therefore, imperative to identify novel therapeutic targets for advanced HCC patients. Persistent injury to the liver as a result of insults such as hepatitis B or C viral (HBV or HCV) infections, alcohol abuse, and non-alcoholic fatty liver disease (NAFLD), results in chronic inflammation, which progresses to hepatic fibrosis and later, cirrhosis, provides the conditions for initiation of HCC. One of the key pathways studied for its role in inflammation and carcinogenesis is the eicosanoid pathway. In this review, we briefly outline the eicosanoid pathway, describe the mechanisms by which some pathway members either facilitate or counter the development of liver diseases, with the focus on NAFLD/hepatic fibrosis/cirrhosis, and HCC. We describe the link between the eicosanoid pathway, inflammation and these liver diseases, and identify components of the eicosanoid pathway that may be used as potential therapeutic targets in HCC.
Collapse
Affiliation(s)
- Anshuli Razdan
- School of Medicine, Western Sydney UniversitySydney, NSW, Australia
- Department of Medical Oncology, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Nathan M Main
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Vincent Chiu
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Nicholas A Shackel
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Paul de Souza
- School of Medicine, Western Sydney UniversitySydney, NSW, Australia
- Department of Medical Oncology, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
- School of Medicine, University of WollongongWollongong, NSW, Australia
| | - Katherine Bryant
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Kieran F Scott
- School of Medicine, Western Sydney UniversitySydney, NSW, Australia
- Department of Medical Oncology, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| |
Collapse
|
32
|
The Impact of the Ca 2+-Independent Phospholipase A 2β (iPLA 2β) on Immune Cells. Biomolecules 2021; 11:biom11040577. [PMID: 33920898 PMCID: PMC8071342 DOI: 10.3390/biom11040577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/31/2022] Open
Abstract
The Ca2+-independent phospholipase A2β (iPLA2β) is a member of the PLA2 family that has been proposed to have roles in multiple biological processes including membrane remodeling, cell proliferation, bone formation, male fertility, cell death, and signaling. Such involvement has led to the identification of iPLA2β activation in several diseases such as cancer, cardiovascular abnormalities, glaucoma, periodontitis, neurological disorders, diabetes, and other metabolic disorders. More recently, there has been heightened interest in the role that iPLA2β plays in promoting inflammation. Recognizing the potential contribution of iPLA2β in the development of autoimmune diseases, we review this issue in the context of an iPLA2β link with macrophages and T-cells.
Collapse
|
33
|
Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, Hu J, Fleming I, Wang DW. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Target Ther 2021; 6:94. [PMID: 33637672 PMCID: PMC7910446 DOI: 10.1038/s41392-020-00443-w] [Citation(s) in RCA: 584] [Impact Index Per Article: 146.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/04/2020] [Accepted: 10/15/2020] [Indexed: 01/31/2023] Open
Abstract
The arachidonic acid (AA) pathway plays a key role in cardiovascular biology, carcinogenesis, and many inflammatory diseases, such as asthma, arthritis, etc. Esterified AA on the inner surface of the cell membrane is hydrolyzed to its free form by phospholipase A2 (PLA2), which is in turn further metabolized by cyclooxygenases (COXs) and lipoxygenases (LOXs) and cytochrome P450 (CYP) enzymes to a spectrum of bioactive mediators that includes prostanoids, leukotrienes (LTs), epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid (diHETEs), eicosatetraenoic acids (ETEs), and lipoxins (LXs). Many of the latter mediators are considered to be novel preventive and therapeutic targets for cardiovascular diseases (CVD), cancers, and inflammatory diseases. This review sets out to summarize the physiological and pathophysiological importance of the AA metabolizing pathways and outline the molecular mechanisms underlying the actions of AA related to its three main metabolic pathways in CVD and cancer progression will provide valuable insight for developing new therapeutic drugs for CVD and anti-cancer agents such as inhibitors of EETs or 2J2. Thus, we herein present a synopsis of AA metabolism in human health, cardiovascular and cancer biology, and the signaling pathways involved in these processes. To explore the role of the AA metabolism and potential therapies, we also introduce the current newly clinical studies targeting AA metabolisms in the different disease conditions.
Collapse
Affiliation(s)
- Bei Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Lujin Wu
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jing Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jiong Hu
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China.
| |
Collapse
|
34
|
Roberts J, Pritchard AL, Treweeke AT, Rossi AG, Brace N, Cahill P, MacRury SM, Wei J, Megson IL. Why Is COVID-19 More Severe in Patients With Diabetes? The Role of Angiotensin-Converting Enzyme 2, Endothelial Dysfunction and the Immunoinflammatory System. Front Cardiovasc Med 2021; 7:629933. [PMID: 33614744 PMCID: PMC7886785 DOI: 10.3389/fcvm.2020.629933] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/15/2020] [Indexed: 01/08/2023] Open
Abstract
Meta-analyses have indicated that individuals with type 1 or type 2 diabetes are at increased risk of suffering a severe form of COVID-19 and have a higher mortality rate than the non-diabetic population. Patients with diabetes have chronic, low-level systemic inflammation, which results in global cellular dysfunction underlying the wide variety of symptoms associated with the disease, including an increased risk of respiratory infection. While the increased severity of COVID-19 amongst patients with diabetes is not yet fully understood, the common features associated with both diseases are dysregulated immune and inflammatory responses. An additional key player in COVID-19 is the enzyme, angiotensin-converting enzyme 2 (ACE2), which is essential for adhesion and uptake of virus into cells prior to replication. Changes to the expression of ACE2 in diabetes have been documented, but they vary across different organs and the importance of such changes on COVID-19 severity are still under investigation. This review will examine and summarise existing data on how immune and inflammatory processes interplay with the pathogenesis of COVID-19, with a particular focus on the impacts that diabetes, endothelial dysfunction and the expression dynamics of ACE2 have on the disease severity.
Collapse
Affiliation(s)
- Jacob Roberts
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Antonia L. Pritchard
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Andrew T. Treweeke
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Adriano G. Rossi
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicole Brace
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Paul Cahill
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Sandra M. MacRury
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Jun Wei
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| | - Ian L. Megson
- Institute for Health Research and Innovation, University of the Highlands and Islands, Inverness, United Kingdom
| |
Collapse
|
35
|
Bai X, Jia J, Kang Q, Fu Y, Zhou Y, Zhong Y, Zhang C, Li M. Integrated Metabolomics and Lipidomics Analysis Reveal Remodeling of Lipid Metabolism and Amino Acid Metabolism in Glucagon Receptor-Deficient Zebrafish. Front Cell Dev Biol 2021; 8:605979. [PMID: 33520988 PMCID: PMC7841139 DOI: 10.3389/fcell.2020.605979] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
The glucagon receptor (GCGR) is activated by glucagon and is essential for glucose, amino acid, and lipid metabolism of animals. GCGR blockade has been demonstrated to induce hypoglycemia, hyperaminoacidemia, hyperglucagonemia, decreased adiposity, hepatosteatosis, and pancreatic α cells hyperplasia in organisms. However, the mechanism of how GCGR regulates these physiological functions is not yet very clear. In our previous study, we revealed that GCGR regulated metabolic network at transcriptional level by RNA-seq using GCGR mutant zebrafish (gcgr -/-). Here, we further performed whole-organism metabolomics and lipidomics profiling on wild-type and gcgr -/- zebrafish to study the changes of metabolites. We found 107 significantly different metabolites from metabolomics analysis and 87 significantly different lipids from lipidomics analysis. Chemical substance classification and pathway analysis integrated with transcriptomics data both revealed that amino acid metabolism and lipid metabolism were remodeled in gcgr-deficient zebrafish. Similar to other studies, our study showed that gcgr -/- zebrafish exhibited decreased ureagenesis and impaired cholesterol metabolism. More interestingly, we found that the glycerophospholipid metabolism was disrupted, the arachidonic acid metabolism was up-regulated, and the tryptophan metabolism pathway was down-regulated in gcgr -/- zebrafish. Based on the omics data, we further validated our findings by revealing that gcgr -/- zebrafish exhibited dampened melatonin diel rhythmicity and increased locomotor activity. These global omics data provide us a better understanding about the role of GCGR in regulating metabolic network and new insight into GCGR physiological functions.
Collapse
Affiliation(s)
- Xuanxuan Bai
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianxin Jia
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qi Kang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yadong Fu
- Center for Circadian Clocks, Soochow University, Suzhou, China.,School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Yingbin Zhong
- Center for Circadian Clocks, Soochow University, Suzhou, China.,School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
36
|
Li Y, Wang L, Xu B, Zhao L, Li L, Xu K, Tang A, Zhou S, Song L, Zhang X, Zhan H. Based on Network Pharmacology Tools to Investigate the Molecular Mechanism of Cordyceps sinensis on the Treatment of Diabetic Nephropathy. J Diabetes Res 2021; 2021:8891093. [PMID: 33628839 PMCID: PMC7884116 DOI: 10.1155/2021/8891093] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/17/2021] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the most common complications of diabetes mellitus and is a major cause of end-stage kidney disease. Cordyceps sinensis (Cordyceps, Dong Chong Xia Cao) is a widely applied ingredient for treating patients with DN in China, while the molecular mechanisms remain unclear. This study is aimed at revealing the therapeutic mechanisms of Cordyceps in DN by undertaking a network pharmacology analysis. MATERIALS AND METHODS In this study, active ingredients and associated target proteins of Cordyceps sinensis were obtained via Traditional Chinese Medicine Systems Pharmacology Database (TCMSP) and Swiss Target Prediction platform, then reconfirmed by using PubChem databases. The collection of DN-related target genes was based on DisGeNET and GeneCards databases. A DN-Cordyceps common target interaction network was carried out via the STRING database, and the results were integrated and visualized by utilizing Cytoscape software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to determine the molecular mechanisms and therapeutic effects of Cordyceps on the treatment of DN. RESULTS Seven active ingredients were screened from Cordyceps, 293 putative target genes were identified, and 85 overlapping targets matched with DN were considered potential therapeutic targets, such as TNF, MAPK1, EGFR, ACE, and CASP3. The results of GO and KEGG analyses revealed that hub targets mainly participated in the AGE-RAGE signaling pathway in diabetic complications, TNF signaling pathway, PI3K-Akt signaling pathway, and IL-17 signaling pathway. These targets were correlated with inflammatory response, apoptosis, oxidative stress, insulin resistance, and other biological processes. CONCLUSIONS Our study showed that Cordyceps is characterized as multicomponent, multitarget, and multichannel. Cordyceps may play a crucial role in the treatment of DN by targeting TNF, MAPK1, EGFR, ACE, and CASP3 signaling and involved in the inflammatory response, apoptosis, oxidative stress, and insulin resistance.
Collapse
Affiliation(s)
- Yan Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Bojun Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Liangbin Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Li Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Keyang Xu
- Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang, China
| | - Anqi Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Shasha Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Lu Song
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Xiao Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Huakui Zhan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| |
Collapse
|
37
|
Fløyel T, Mirza AH, Kaur S, Frørup C, Yarani R, Størling J, Pociot F. The Rac2 GTPase contributes to cathepsin H-mediated protection against cytokine-induced apoptosis in insulin-secreting cells. Mol Cell Endocrinol 2020; 518:110993. [PMID: 32814070 DOI: 10.1016/j.mce.2020.110993] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 07/01/2020] [Accepted: 08/12/2020] [Indexed: 10/23/2022]
Abstract
The type 1 diabetes (T1D) risk locus on chromosome 15q25.1 harbors the candidate gene CTSH (cathepsin H). We previously demonstrated that CTSH regulates β-cell function in vitro and in vivo. CTSH overexpression protected insulin-secreting INS-1 cells against cytokine-induced apoptosis. The purpose of the present study was to identify the genes through which CTSH mediates its protective effects. Microarray analysis identified 63 annotated genes differentially expressed between CTSH-overexpressing INS-1 cells and control cells treated with interleukin-1β and interferon-γ for up to 16h. Permutation test identified 10 significant genes across all time-points: Elmod1, Fam49a, Gas7, Gna15, Msrb3, Nox1, Ptgs1, Rac2, Scn7a and Ttn. Pathway analysis identified the "Inflammation mediated by chemokine and cytokine signaling pathway" with Gna15, Ptgs1 and Rac2 as significant. Knockdown of Rac2 abolished the protective effect of CTSH overexpression on cytokine-induced apoptosis, suggesting that the small GTPase and T1D candidate gene Rac2 contributes to the anti-apoptotic effect of CTSH.
Collapse
Affiliation(s)
- Tina Fløyel
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, DK-2820, Gentofte, Denmark.
| | - Aashiq Hussain Mirza
- Department of Pharmacology, Weill Cornell Medicine, 1300 York Avenue, Box 125, New York, NY, 10065, USA.
| | - Simranjeet Kaur
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, DK-2820, Gentofte, Denmark.
| | - Caroline Frørup
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, DK-2820, Gentofte, Denmark.
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, DK-2820, Gentofte, Denmark.
| | - Joachim Størling
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, DK-2820, Gentofte, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark.
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Niels Steensens Vej 2, DK-2820, Gentofte, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
38
|
Kiziltoprak H, Koc M, Yetkin E, Tekin K, Inanc M, Ozulken K. Additive Effect of Topical Nepafenac on Mydriasis in Patients With Diabetes Mellitus. Eye Contact Lens 2020; 46:310-313. [PMID: 31503086 DOI: 10.1097/icl.0000000000000657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To evaluate the additive effect of topical nepafenac on pupil diameter (PD) in patients with diabetes mellitus (DM) and cataract. METHODS This prospective comparative study included the patients having cataract surgery with and without DM. Two consecutive PD measurements were taken using an automatic quantitative pupillometry system (MonPack One, Metrovision). A baseline measurement was taken, then one drop of nepafenac % 0.1 (Nevanac; Alcon, Fort Worth, TX) was instilled only to the eye that will be operated on (study eye). Cyclopentolate 1.0% (Sikloplejin; Abdi İbrahim, İstanbul, Turkey) was instilled to both eyes (study eye/fellow eye) 5 minutes later. The second measurement was taken at 1 hour after this application. RESULTS The DM group consisted of 43 patients, and the control group consisted of 39 participants. The baseline PDs of both eyes were similar in the DM group (P=0.070) and the control group (P=0.345). The change in pupil size from baseline to mydriasis was statistically significantly greater in the study eyes (2.69±0.53) than fellow eyes (2.54±0.61) in the DM group (P=0.009), but there was no statistically significant difference in the control group (2.94±0.63 vs. 2.86±0.58). When the groups were compared, the PD changes were similar in the study eyes between groups (P=0.065), while the PD changes in the fellow eyes were lower in the DM group (P=0.017). CONCLUSIONS Nepafenac has been shown additive effect on pupil dilation in diabetic patients before cataract surgery.
Collapse
Affiliation(s)
- Hasan Kiziltoprak
- Ophthalmology Department (H.K.), Bingol Women's Health and Children's Hospital, Bingol, Turkey; Ophthalmology Department (M.K., E.Y.), Ankara Ulucanlar Eye Training and Research Hospital, Ankara, Turkey; Ophthalmology Department (K.T., M.I.), Ercis State Hospital, Van, Turkey; and Ophthalmology Department (K.O.), TOBB ETU Hospital, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
39
|
Nelson AJ, Stephenson DJ, Bone RN, Cardona CL, Park MA, Tusing YG, Lei X, Kokotos G, Graves CL, Mathews CE, Kramer J, Hessner MJ, Chalfant CE, Ramanadham S. Lipid mediators and biomarkers associated with type 1 diabetes development. JCI Insight 2020; 5:138034. [PMID: 32814707 PMCID: PMC7455134 DOI: 10.1172/jci.insight.138034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/09/2020] [Indexed: 01/13/2023] Open
Abstract
Type 1 diabetes (T1D) is a consequence of autoimmune β cell destruction, but the role of lipids in this process is unknown. We previously reported that activation of Ca2+-independent phospholipase A2β (iPLA2β) modulates polarization of macrophages (MΦ). Hydrolysis of the sn-2 substituent of glycerophospholipids by iPLA2β can lead to the generation of oxidized lipids (eicosanoids), pro- and antiinflammatory, which can initiate and amplify immune responses triggering β cell death. As MΦ are early triggers of immune responses in islets, we examined the impact of iPLA2β-derived lipids (iDLs) in spontaneous-T1D prone nonobese diabetic mice (NOD), in the context of MΦ production and plasma abundances of eicosanoids and sphingolipids. We find that (a) MΦNOD exhibit a proinflammatory lipid landscape during the prediabetic phase; (b) early inhibition or genetic reduction of iPLA2β reduces production of select proinflammatory lipids, promotes antiinflammatory MΦ phenotype, and reduces T1D incidence; (c) such lipid changes are reflected in NOD plasma during the prediabetic phase and at T1D onset; and (d) importantly, similar lipid signatures are evidenced in plasma of human subjects at high risk for developing T1D. These findings suggest that iDLs contribute to T1D onset and identify select lipids that could be targeted for therapeutics and, in conjunction with autoantibodies, serve as early biomarkers of pre-T1D.
Collapse
Affiliation(s)
- Alexander J Nelson
- Department of Cell, Developmental, and Integrative Biology, and.,Comprehensive Diabetes Center, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Daniel J Stephenson
- Department of Cell Biology, Microbiology and Molecular Biology (CMMB), University of South Florida, Tampa, Florida, USA
| | - Robert N Bone
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher L Cardona
- Department of Cell Biology, Microbiology and Molecular Biology (CMMB), University of South Florida, Tampa, Florida, USA
| | - Margaret A Park
- Department of Cell Biology, Microbiology and Molecular Biology (CMMB), University of South Florida, Tampa, Florida, USA
| | - Ying G Tusing
- Department of Cell, Developmental, and Integrative Biology, and.,Comprehensive Diabetes Center, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology, and.,Comprehensive Diabetes Center, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Christina L Graves
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Clayton E Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Health Science Center, Gainesville, Florida, USA
| | - Joanna Kramer
- Max McGee Research Center for Juvenile Diabetes, Department of Pediatrics at Medical College of Wisconsin and Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Martin J Hessner
- Max McGee Research Center for Juvenile Diabetes, Department of Pediatrics at Medical College of Wisconsin and Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology (CMMB), University of South Florida, Tampa, Florida, USA.,Research Service, James A. Haley Veterans Hospital, Tampa, Florida, USA
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, and.,Comprehensive Diabetes Center, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| |
Collapse
|
40
|
Leuti A, Fazio D, Fava M, Piccoli A, Oddi S, Maccarrone M. Bioactive lipids, inflammation and chronic diseases. Adv Drug Deliv Rev 2020; 159:133-169. [PMID: 32628989 DOI: 10.1016/j.addr.2020.06.028] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Endogenous bioactive lipids are part of a complex network that modulates a plethora of cellular and molecular processes involved in health and disease, of which inflammation represents one of the most prominent examples. Inflammation serves as a well-conserved defence mechanism, triggered in the event of chemical, mechanical or microbial damage, that is meant to eradicate the source of damage and restore tissue function. However, excessive inflammatory signals, or impairment of pro-resolving/anti-inflammatory pathways leads to chronic inflammation, which is a hallmark of chronic pathologies. All main classes of endogenous bioactive lipids - namely eicosanoids, specialized pro-resolving lipid mediators, lysoglycerophopsholipids and endocannabinoids - have been consistently involved in the chronic inflammation that characterises pathologies such as cancer, diabetes, atherosclerosis, asthma, as well as autoimmune and neurodegenerative disorders and inflammatory bowel diseases. This review gathers the current knowledge concerning the involvement of endogenous bioactive lipids in the pathogenic processes of chronic inflammatory pathologies.
Collapse
|
41
|
Xia F, He C, Ren M, Xu FG, Wan JB. Quantitative profiling of eicosanoids derived from n-6 and n-3 polyunsaturated fatty acids by twin derivatization strategy combined with LC-MS/MS in patients with type 2 diabetes mellitus. Anal Chim Acta 2020; 1120:24-35. [DOI: 10.1016/j.aca.2020.04.064] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 01/19/2023]
|
42
|
Eicosanoids and Oxidative Stress in Diabetic Retinopathy. Antioxidants (Basel) 2020; 9:antiox9060520. [PMID: 32545552 PMCID: PMC7346161 DOI: 10.3390/antiox9060520] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is an important factor to cause the pathogenesis of diabetic retinopathy (DR) because the retina has high vascularization and long-time light exposition. Cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes can convert arachidonic acid (AA) into eicosanoids, which are important lipid mediators to regulate DR development. COX-derived metabolites appear to be significant factors causative to oxidative stress and retinal microvascular dysfunction. Several elegant studies have unraveled the importance of LOX-derived eicosanoids, including LTs and HETEs, to oxidative stress and retinal microvascular dysfunction. The role of CYP eicosanoids in DR is yet to be explored. There is clear evidence that CYP-derived epoxyeicosatrienoic acids (EETs) have detrimental effects on the retina. Our recent study showed that the renin-angiotensin system (RAS) activation augments retinal soluble epoxide hydrolase (sEH), a crucial enzyme degrading EETs. Our findings suggest that EETs blockade can enhance the ability of RAS blockade to prevent or mitigate microvascular damage in DR. This review will focus on the critical information related the function of these eicosanoids in the retina, the interaction between eicosanoids and reactive oxygen species (ROS), and the involvement of eicosanoids in DR. We also identify potential targets for the treatment of DR.
Collapse
|
43
|
Dai N, Yang C, Fan Q, Wang M, Liu X, Zhao H, Zhao C. The Anti-inflammatory Effect of Soluble Epoxide Hydrolase Inhibitor and 14, 15-EET in Kawasaki Disease Through PPARγ/STAT1 Signaling Pathway. Front Pediatr 2020; 8:451. [PMID: 32903307 PMCID: PMC7434939 DOI: 10.3389/fped.2020.00451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022] Open
Abstract
Soluble epoxide hydrolase (sEH) is responsible for rapid degradation of 14, 15-EET, which is one of the isomers of EETs and plays an important role in cardiovascular diseases. In this study, we investigated the mechanism by which sEH inhibitor AUDA played an anti-inflammatory effect in HCAECs. Our results indicated that AUDA treatment promoted PPARγ expression, while knockdown of PPARγ blocked the cell growth and STAT1 expression inhibition induced by 100 μmol/L AUDA in HCAECs. AUDA also inhibited the overexpression of TNF-α, IL-1 β, and MMP-9 induced by KD sera in HCAECs. Moreover, 30 blood samples from children with Kawasaki disease (KD) were collected with 30 healthy children as the control group. QPCR and ELISA assays were used to detect the level of 14, 15-EET, TNF-α, IL-1β, and MMP-9. We found that the level of 14, 15-EET was higher in peripheral blood of children with KD compared with healthy controls (P < 0.05). In comparison to KD children with non-coronary artery lesion (nCAL), the level of 14, 15-EET was higher in peripheral blood of KD children with coronary artery lesion (CAL) (P < 0.05). Compared with healthy control group, the expression levels of TNF-α, IL-1β, and MMP-9 in patients with KD were significantly up-regulated. Compared with nCAL KD children, the expression levels of TNF-α, IL-1β, and MMP-9 in CAL children were abnormally high (P < 0.05). Our study indicated that AUDA played an anti-inflammatory effect in HCAECs through PPARγ/STAT1 signaling pathway, and 14, 15-EET is up-regulated in children with KD, suggesting that 14, 15-EET involved in the progression of KD.
Collapse
Affiliation(s)
- Na Dai
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China.,Department of Pediatrics, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Chunyan Yang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China.,Department of Pediatrics, Liaocheng People's Hospital, Liaocheng, China
| | - Qing Fan
- Department of Pediatrics, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Minmin Wang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Xiaoyue Liu
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Haizhao Zhao
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Cuifen Zhao
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
44
|
Dos Santos LRB, Fleming I. Role of cytochrome P450-derived, polyunsaturated fatty acid mediators in diabetes and the metabolic syndrome. Prostaglandins Other Lipid Mediat 2019; 148:106407. [PMID: 31899373 DOI: 10.1016/j.prostaglandins.2019.106407] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 11/14/2019] [Accepted: 12/23/2019] [Indexed: 12/17/2022]
Abstract
Over the last decade, cases of metabolic syndrome and type II diabetes have increased exponentially. Exercise and ω-3 polyunsaturated fatty acid (PUFA)-enriched diets are usually prescribed but no therapy is effectively able to restore the impaired glucose metabolism, hypertension, and atherogenic dyslipidemia encountered by diabetic patients. PUFAs are metabolized by different enzymes into bioactive metabolites with anti- or pro-inflammatory activity. One important class of PUFA metabolizing enzymes are the cytochrome P450 (CYP) enzymes that can generate a series of bioactive products, many of which have been attributed protective/anti-inflammatory and insulin-sensitizing effects in animal models. PUFA epoxides are, however, further metabolized by the soluble epoxide hydrolase (sEH) to fatty acid diols. The biological actions of the latter are less well understood but while low concentrations may be biologically important, higher concentrations of diols derived from linoleic acid and docosahexaenoic acid have been linked with inflammation. One potential application for sEH inhibitors is in the treatment of diabetic retinopathy where sEH expression and activity is elevated as are levels of a diol of docosahexaenoic acid that can induce the destabilization of the retina vasculature.
Collapse
Affiliation(s)
- Laila R B Dos Santos
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany; German Centre for Cardiovascular Research (DZHK) Partner Site Rhein-Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany; German Centre for Cardiovascular Research (DZHK) Partner Site Rhein-Main, Germany.
| |
Collapse
|
45
|
Nelson AJ, Stephenson DJ, Cardona CL, Lei X, Almutairi A, White TD, Tusing YG, Park MA, Barbour SE, Chalfant CE, Ramanadham S. Macrophage polarization is linked to Ca 2+-independent phospholipase A 2β-derived lipids and cross-cell signaling in mice. J Lipid Res 2019; 61:143-158. [PMID: 31818877 DOI: 10.1194/jlr.ra119000281] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/27/2019] [Indexed: 12/27/2022] Open
Abstract
Phospholipases A2 (PLA2s) catalyze hydrolysis of the sn-2 substituent from glycerophospholipids to yield a free fatty acid (i.e., arachidonic acid), which can be metabolized to pro- or anti-inflammatory eicosanoids. Macrophages modulate inflammatory responses and are affected by Ca2+-independent phospholipase A2 (PLA2)β (iPLA2β). Here, we assessed the link between iPLA2β-derived lipids (iDLs) and macrophage polarization. Macrophages from WT and KO (iPLA2β-/-) mice were classically M1 pro-inflammatory phenotype activated or alternatively M2 anti-inflammatory phenotype activated, and eicosanoid production was determined by ultra-performance LC ESI-MS/MS. As a genotypic control, we performed similar analyses on macrophages from RIP.iPLA2β.Tg mice with selective iPLA2β overexpression in β-cells. Compared with WT, generation of select pro-inflammatory prostaglandins (PGs) was lower in iPLA2β-/- , and that of a specialized pro-resolving lipid mediator (SPM), resolvin D2, was higher; both changes are consistent with the M2 phenotype. Conversely, macrophages from RIP.iPLA2β.Tg mice exhibited an opposite landscape, one associated with the M1 phenotype: namely, increased production of pro-inflammatory eicosanoids (6-keto PGF1α, PGE2, leukotriene B4) and decreased ability to generate resolvin D2. These changes were not linked with secretory PLA2 or cytosolic PLA2α or with leakage of the transgene. Thus, we report previously unidentified links between select iPLA2β-derived eicosanoids, an SPM, and macrophage polarization. Importantly, our findings reveal for the first time that β-cell iPLA2β-derived signaling can predispose macrophage responses. These findings suggest that iDLs play critical roles in macrophage polarization, and we posit that they could be targeted therapeutically to counter inflammation-based disorders.
Collapse
Affiliation(s)
- Alexander J Nelson
- Department of Cell, Developmental, and Integrative Biology University of Alabama at Birmingham, Birmingham, AL 35294.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Daniel J Stephenson
- Department of Cell Biology, Microbiology, and Molecular Biology (CMMB), University of South Florida, Tampa, FL 33620
| | - Christopher L Cardona
- Department of Cell Biology, Microbiology, and Molecular Biology (CMMB), University of South Florida, Tampa, FL 33620
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology University of Alabama at Birmingham, Birmingham, AL 35294.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Abdulaziz Almutairi
- Department of Cell, Developmental, and Integrative Biology University of Alabama at Birmingham, Birmingham, AL 35294.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Tayleur D White
- Department of Cell, Developmental, and Integrative Biology University of Alabama at Birmingham, Birmingham, AL 35294.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Ying G Tusing
- Department of Cell, Developmental, and Integrative Biology University of Alabama at Birmingham, Birmingham, AL 35294.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Margaret A Park
- Department of Cell Biology, Microbiology, and Molecular Biology (CMMB), University of South Florida, Tampa, FL 33620
| | - Suzanne E Barbour
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology (CMMB), University of South Florida, Tampa, FL 33620.,Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology University of Alabama at Birmingham, Birmingham, AL 35294 .,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
46
|
The Combination of Whole Cell Lipidomics Analysis and Single Cell Confocal Imaging of Fluidity and Micropolarity Provides Insight into Stress-Induced Lipid Turnover in Subcellular Organelles of Pancreatic Beta Cells. Molecules 2019; 24:molecules24203742. [PMID: 31627330 PMCID: PMC6833103 DOI: 10.3390/molecules24203742] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 12/22/2022] Open
Abstract
Modern omics techniques reveal molecular structures and cellular networks of tissues and cells in unprecedented detail. Recent advances in single cell analysis have further revolutionized all disciplines in cellular and molecular biology. These methods have also been employed in current investigations on the structure and function of insulin secreting beta cells under normal and pathological conditions that lead to an impaired glucose tolerance and type 2 diabetes. Proteomic and transcriptomic analyses have pointed to significant alterations in protein expression and function in beta cells exposed to diabetes like conditions (e.g., high glucose and/or saturated fatty acids levels). These nutritional overload stressful conditions are often defined as glucolipotoxic due to the progressive damage they cause to the cells. Our recent studies on the rat insulinoma-derived INS-1E beta cell line point to differential effects of such conditions in the phospholipid bilayers in beta cells. This review focuses on confocal microscopy-based detection of these profound alterations in the plasma membrane and membranes of insulin granules and lipid droplets in single beta cells under such nutritional load conditions.
Collapse
|
47
|
Zhu Y, Li M, Rahman ML, Hinkle SN, Wu J, Weir NL, Lin Y, Yang H, Tsai MY, Ferrara A, Zhang C. Plasma phospholipid n-3 and n-6 polyunsaturated fatty acids in relation to cardiometabolic markers and gestational diabetes: A longitudinal study within the prospective NICHD Fetal Growth Studies. PLoS Med 2019; 16:e1002910. [PMID: 31518348 PMCID: PMC6743768 DOI: 10.1371/journal.pmed.1002910] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Despite dietary recommendations of polyunsaturated fatty acids (PUFAs) for cardiometabolic health, n-3 and n-6 PUFAs and their interplay in relation to diabetes risk remain debated. Importantly, data among pregnant women are scarce. We investigated individual plasma phospholipid n-3 and n-6 PUFAs in early to midpregnancy in relation to subsequent risk of gestational diabetes mellitus (GDM). METHODS AND FINDINGS Within the National Institute of Child Health and Human Development (NICHD) Fetal Growth Studies-Singleton Cohort (n = 2,802), individual plasma phospholipid n-3 and n-6 PUFAs levels were measured at gestational weeks (GWs) 10-14, 15-26, 23-31, and 33-39 among 107 GDM cases (ascertained on average at GW 27) and 214 non-GDM controls. Conditional logistic regression was used, adjusting for major risk factors for GDM. After adjusting for covariates, individual n-3 eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA) were inversely correlated with insulin-resistance markers, whereas individual n-6 dihomo-gamma-linolenic acid (DGLA) was positively correlated with insulin-resistance markers. At GW 15-26, a standard deviation (SD) increase in total n-3 PUFAs and individual n-3 DPA was associated with a 36% (adjusted odds ratio 0.64; 95% CI 0.42-0.96; P = 0.042) and 33% (0.67; 95% CI 0.45-0.99; P = 0.047) lower risk of GDM, respectively; however, the significance did not persist after post hoc false-discovery rate (FDR) correction (FDR-corrected P values > 0.05). Associations between total n-6 PUFAs and GDM were null, whereas associations with individual n-6 PUFAs were differential. Per SD increase, gamma-linolenic acid (GLA) at GWs 10-14 and DGLA at GWs 10-14 and 15-26 were significantly associated with a 1.40- to 1.95-fold higher risk of GDM, whereas docosatetraenoic acid (DTA) at GW 15-26 was associated with a 45% (0.55; 95% CI 0.37-0.83) lower risk of GDM (all FDR-corrected P values < 0.05). Null associations were observed for linoleic acid (LA) in either gestational window in relation to risk of GDM. Women with high (≥median) n-3 PUFAs and low (<median) n-6 PUFAs levels had a 64% (95% CI 0.14-0.95; P value = 0.039) lower risk of GDM versus women with low n-3 and high n-6 PUFAs. Limitations include the inability to distinguish between exogenous and endogenous influences on circulating PUFA levels and the lack of causality inherent in observational studies. CONCLUSIONS Our findings may suggest a potential role of primarily endogenously metabolized plasma phospholipid n-6 PUFAs including GLA, DGLA, and DTA in early to midpregnancy in the development of GDM. Null findings on primarily diet-derived n-3 EPA and DHA and n-6 LA do not provide strong evidence to suggest a beneficial role in prevention of GDM, although not excluding the potential benefit of EPA and DHA on glucose-insulin homeostasis given the inverse associations with insulin-resistance markers. Our findings highlight the importance of assessing individual circulating PUFAs to investigate their distinct pathophysiologic roles in glucose homeostasis in pregnancy.
Collapse
Affiliation(s)
- Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California, Oakland, California, United States of America
- Department of Epidemiology & Biostatistics, University of California, San Francisco, California, United States of America
- * E-mail: (CZ); (YZ)
| | - Mengying Li
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Mohammad L. Rahman
- Department of Population Medicine and Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stefanie N. Hinkle
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Jing Wu
- Glotech Inc., Bethesda, Maryland, United States of America
| | - Natalie L. Weir
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Yuan Lin
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, United States of America
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Assiamira Ferrara
- Division of Research, Kaiser Permanente Northern California, Oakland, California, United States of America
| | - Cuilin Zhang
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
- * E-mail: (CZ); (YZ)
| |
Collapse
|
48
|
Baboota RK, Shinde AB, Lemaire K, Fransen M, Vinckier S, Van Veldhoven PP, Schuit F, Baes M. Functional peroxisomes are required for β-cell integrity in mice. Mol Metab 2019; 22:71-83. [PMID: 30795913 PMCID: PMC6437690 DOI: 10.1016/j.molmet.2019.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/25/2019] [Accepted: 02/04/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives Peroxisomes play a crucial role in lipid and reactive oxygen species metabolism, but their importance for pancreatic β-cell functioning is presently unknown. To examine the contribution of peroxisomal metabolism to β-cell homeostasis in mice, we inactivated PEX5, the import receptor for peroxisomal matrix proteins, in an inducible and β-cell restricted manner (Rip-Pex5−/− mice). Methods After tamoxifen-induced recombination of the Pex5 gene at the age of 6 weeks, mice were fed either normal chow or a high-fat diet for 12 weeks and were subsequently phenotyped. Results Increased levels of very long chain fatty acids and reduced levels of plasmalogens in islets confirmed impairment of peroxisomal fatty acid oxidation and ether lipid synthesis, respectively. The Rip-Pex5−/− mice fed on either diet exhibited glucose intolerance associated with impaired insulin secretion. Ultrastructural and biochemical analysis revealed a decrease in the density of mature insulin granules and total pancreatic insulin content, which was further accompanied by mitochondrial disruptions, reduced complex I activity and massive vacuole overload in β-cells. RNAseq analysis suggested that cell death pathways were affected in islets from HFD-fed Rip-Pex5−/− mice. Consistent with this change we observed increased β-cell apoptosis in islets and a decrease in β-cell mass. Conclusions Our data indicate that normal peroxisome metabolism in β-cells is crucial to preserve their structure and function. Pex5 deletion in β-cells impairs glucose tolerance and reduces β-cell mass. Pex5-deficient β-cells display increased apoptosis. Peroxisomal loss causes mitochondrial deterioration and cytoplasmic vacuolization.
Collapse
Affiliation(s)
- Ritesh Kumar Baboota
- KU Leuven - University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Laboratory of Cell Metabolism, B-3000, Leuven, Belgium
| | - Abhijit Babaji Shinde
- KU Leuven - University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Laboratory of Cell Metabolism, B-3000, Leuven, Belgium
| | - Katleen Lemaire
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Gene Expression Unit, B-3000, Leuven, Belgium
| | - Marc Fransen
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Laboratory for Lipid Biochemistry and Protein Interactions, KU Leuven, B-3000, Leuven, Belgium
| | - Stefan Vinckier
- VIB-KULeuven Centre for Cancer Biology, Laboratory of Angiogenesis and Vascular Metabolism, B-3000, Leuven, Belgium
| | - Paul P Van Veldhoven
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Laboratory for Lipid Biochemistry and Protein Interactions, KU Leuven, B-3000, Leuven, Belgium
| | - Frans Schuit
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Gene Expression Unit, B-3000, Leuven, Belgium
| | - Myriam Baes
- KU Leuven - University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Laboratory of Cell Metabolism, B-3000, Leuven, Belgium.
| |
Collapse
|
49
|
van der Heijden YF, Abdullah F, Andrade BB, Andrews JR, Christopher DJ, Croda J, Ewing H, Haas DW, Hatherill M, Horsburgh CR, Mave V, Nakaya HI, Rolla V, Srinivasan S, Sugiyono RI, Ugarte-Gil C, Hamilton C. Building capacity for advances in tuberculosis research; proceedings of the third RePORT international meeting. Tuberculosis (Edinb) 2018; 113:153-162. [PMID: 30514497 PMCID: PMC6349374 DOI: 10.1016/j.tube.2018.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 10/28/2022]
Abstract
RePORT International is a global network of research sites in India, Brazil, Indonesia, South Africa, China, and the Philippines dedicated to collaborative tuberculosis research in the context of HIV. A standardized research protocol (the Common Protocol) guides the enrollment of participants with active pulmonary tuberculosis and contacts into observational cohorts. The establishment of harmonized clinical data and bio-repositories will allow cutting-edge, large-scale advances in the understanding of tuberculosis, including identification of novel biomarkers for progression to active tuberculosis and relapse after treatment. The RePORT International infrastructure aims to support research capacity development through enabling globally-diverse collaborations. To that end, representatives from the RePORT International network sites, funding agencies, and other stakeholders gathered together in Brazil in September 2017 to present updates on relevant research findings and discuss ideas for collaboration. Presenters emphasized research involving biomarker identification for incipient tuberculosis, host immunity and pharmacogenomics, co-morbidities such as HIV and type 2 diabetes mellitus, and tuberculosis transmission in vulnerable and high-risk populations. Currently, 962 active TB participants and 670 household contacts have contributed blood, sputum, urine and microbes to in-country biorepositories. Cross-consortium collaborations have begun sharing data and specimens to analyze molecular and cytokine predictive patterns.
Collapse
Affiliation(s)
- Yuri F van der Heijden
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, USA; Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Fareed Abdullah
- Office of AIDS and TB Research, South African Medical Research Council, Pretoria, South Africa.
| | - Bruno B Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, 40296-710, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, José Silveira Foundation, Salvador, 45204-040, Brazil; Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7925, South Africa; Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Universidade Salvador (UNIFACS), Laureate University, Salvador, Bahia, 41720-200, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, 40290-000, Brazil.
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | | | - Julio Croda
- School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil, Oswaldo Cruz Foundation, Campo Grande, Brazil.
| | - Heather Ewing
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - David W Haas
- Departments of Medicine, Pharmacology, Pathology, Microbiology & Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Internal Medicine, Meharry Medical College, Nashville, TN, USA.
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, South Africa.
| | - C Robert Horsburgh
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA; Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Vidya Mave
- Byramjee-Jeejeebhoy Government Medical College-Johns Hopkins University Clinical Research Site, Pune, India; Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Valeria Rolla
- Clinical Research Laboratory on Mycobacteria, National Institute of Infectious Diseases Evandro Chagas, Fiocruz, Brazil.
| | - Sudha Srinivasan
- Division of AIDS, National Institute of Allergy and Infectious Diseases at the National Institutes of Health, Bethesda, MD, USA.
| | - Retna Indah Sugiyono
- INA-RESPOND, National Institute of Health Research and Development, Ministry of Health, Indonesia.
| | - Cesar Ugarte-Gil
- Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru; School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru; TB Centre, London School of Hygiene and Tropical Medicina, London, UK; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Maryland, USA.
| | | |
Collapse
|
50
|
Sonnweber T, Pizzini A, Nairz M, Weiss G, Tancevski I. Arachidonic Acid Metabolites in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2018; 19:ijms19113285. [PMID: 30360467 PMCID: PMC6274989 DOI: 10.3390/ijms19113285] [Citation(s) in RCA: 297] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022] Open
Abstract
Lipid and immune pathways are crucial in the pathophysiology of metabolic and cardiovascular disease. Arachidonic acid (AA) and its derivatives link nutrient metabolism to immunity and inflammation, thus holding a key role in the emergence and progression of frequent diseases such as obesity, diabetes, non-alcoholic fatty liver disease, and cardiovascular disease. We herein present a synopsis of AA metabolism in human health, tissue homeostasis, and immunity, and explore the role of the AA metabolome in diverse pathophysiological conditions and diseases.
Collapse
Affiliation(s)
- Thomas Sonnweber
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|