1
|
Wei Z, Iyer MR, Zhao B, Deng J, Mitchell CS. Artificial Intelligence-Assisted Comparative Analysis of the Overlapping Molecular Pathophysiology of Alzheimer's Disease, Amyotrophic Lateral Sclerosis, and Frontotemporal Dementia. Int J Mol Sci 2024; 25:13450. [PMID: 39769215 PMCID: PMC11678588 DOI: 10.3390/ijms252413450] [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: 10/28/2024] [Revised: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The overlapping molecular pathophysiology of Alzheimer's Disease (AD), Amyotrophic Lateral Sclerosis (ALS), and Frontotemporal Dementia (FTD) was analyzed using relationships from a knowledge graph of 33+ million biomedical journal articles. The unsupervised learning rank aggregation algorithm from SemNet 2.0 compared the most important amino acid, peptide, and protein (AAPP) nodes connected to AD, ALS, or FTD. FTD shared 99.9% of its nodes with ALS and AD; AD shared 64.2% of its nodes with FTD and ALS; and ALS shared 68.3% of its nodes with AD and FTD. The results were validated and mapped to functional biological processes using supervised human supervision and an external large language model. The overall percentages of mapped intersecting biological processes were as follows: inflammation and immune response, 19%; synapse and neurotransmission, 19%; cell cycle, 15%; protein aggregation, 12%; membrane regulation, 11%; stress response and regulation, 9%; and gene regulation, 4%. Once normalized for node count, biological mappings for cell cycle regulation and stress response were more prominent in the intersection of AD and FTD. Protein aggregation, gene regulation, and energetics were more prominent in the intersection of ALS and FTD. Synapse and neurotransmission, membrane regulation, and inflammation and immune response were greater at the intersection of AD and ALS. Given the extensive molecular pathophysiology overlap, small differences in regulation, genetic, or environmental factors likely shape the underlying expressed disease phenotype. The results help prioritize testable hypotheses for future clinical or experimental research.
Collapse
Affiliation(s)
- Zihan Wei
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Meghna R. Iyer
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Benjamin Zhao
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jennifer Deng
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cassie S. Mitchell
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Machine Learning at Georgia Tech, Atlanta, GA 30332, USA
| |
Collapse
|
2
|
Pardo M, Li C, Jabali A, Rudich Y. Cellular and metabolic impacts of repeated sub-acute exposures to biomass-burning extracts in vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 289:117491. [PMID: 39657377 DOI: 10.1016/j.ecoenv.2024.117491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
The increasing exposure to biomass-burning emissions underscores the need to understand their toxicological impacts on human health. In this study, we developed a laboratory model to evaluate the effects of single and repeated sub-acute exposures to water-soluble wood tar (WT) extracts, a product of biomass burning, on human lung, liver, and immune cells. Using representative cell lines for different tissues, we examined the cytotoxic effects under conditions mimicking sub-acute environmental exposure levels relevant to humans. Our findings indicate that repeated sub-acute exposures to water-soluble WT extracts significantly enhance the inflammatory response, evidenced by increased IL6, IL8, and TNFa cytokine levels, compared to a single exposure. Additionally, oxidative stress responses were more pronounced with increased lipid peroxidation and HMOX1, GCLC and CYP1A1 gene expression following repeated exposures. Metabolomics analyses of polar and lipid metabolites revealed changes related to energy production and consumption that emerge even after a single exposure at sub-acute levels and vary across different cell types representing the different tissues. Impaired cellular respiration, measured by oxygen consumption rate, corroborates the observed changes. These results provide important insights into the cellular mechanisms driving the response to biomass-burning exposure and highlight the potential health risks associated with sub-acute exposure to environmental pollutants.
Collapse
Affiliation(s)
- Michal Pardo
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel.
| | - Chunlin Li
- College of Environmental Science and Engineering, Tongji University, Shanghai 200072, China
| | - Amani Jabali
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel
| | - Yinon Rudich
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel
| |
Collapse
|
3
|
Zhang Y, Gong J, Hu X, He L, Lin Y, Zhang J, Meng X, Zhang Y, Mo J, Day DB, Xiang J. Glycerophospholipid metabolism changes association with ozone exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 475:134870. [PMID: 38876019 DOI: 10.1016/j.jhazmat.2024.134870] [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: 04/15/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/16/2024]
Abstract
Exposure to ozone (O3) has been associated with cardiovascular outcomes in humans, yet the underlying mechanisms of the adverse effect remain poorly understood. We aimed to investigate the association between O3 exposure and glycerophospholipid metabolism in healthy young adults. We quantified plasma concentrations of phosphatidylcholines (PCs) and lysophosphatidylcholines (lysoPCs) using a UPLC-MS/MS system. Time-weighted personal exposures were calculated to O3 and co-pollutants over 4 time windows, and we employed orthogonal partial least squares discriminant analysis to discern differences in lipids profiles between high and low O3 exposure. Linear mixed-effects models and mediation analysis were utilized to estimate the associations between O3 exposure, lipids, and cardiovascular physiology indicators. Forty-three healthy adults were included in this study, and the mean (SD) time-weighted personal exposures to O3 was 9.08 (4.06) ppb. With shorter exposure durations, O3 increases were associated with increasing PC and lysoPC levels; whereas at longer exposure times, the opposite relationship was shown. Furthermore, two specific lipids, namely lysoPC a C26:0 and lysoPC a C17:0, showed significantly positive mediating effects on associations of long-term O3 exposure with pulse wave velocity and systolic blood pressure, respectively. Alterations in specific lipids may underlie the cardiovascular effects of O3 exposure.
Collapse
Affiliation(s)
- Yi Zhang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China
| | - Jicheng Gong
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China.
| | - Xinyan Hu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China
| | - Linchen He
- College of Health, Lehigh University, Bethlehem, PA 19019, United States; Global Health Institute, Nicholas School of the Environment, Duke University, Durham, NC 27708, United States
| | - Yan Lin
- Global Health Institute, Nicholas School of the Environment, Duke University, Durham, NC 27708, United States
| | - Junfeng Zhang
- Global Health Institute, Nicholas School of the Environment, Duke University, Durham, NC 27708, United States
| | - Xin Meng
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, China
| | - Yinping Zhang
- Department of Building Science, Tsinghua University, Beijing 100084, China
| | - Jinhan Mo
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Drew B Day
- Seattle Children's Research Institute, Seattle, WA 98121, United States
| | - Jianbang Xiang
- School of Public Health, Sun Yat-Sen University, Shenzhen 518107, China
| |
Collapse
|
4
|
Huang HX, Inglese P, Tang J, Yagoubi R, Correia GDS, Horneffer-van der Sluis VM, Camuzeaux S, Wu V, Kopanitsa MV, Willumsen N, Jackson JS, Barron AM, Saito T, Saido TC, Gentlemen S, Takats Z, Matthews PM. Mass spectrometry imaging highlights dynamic patterns of lipid co-expression with Aβ plaques in mouse and human brains. J Neurochem 2024; 168:1193-1214. [PMID: 38372586 DOI: 10.1111/jnc.16042] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/13/2023] [Accepted: 12/06/2023] [Indexed: 02/20/2024]
Abstract
Lipids play crucial roles in the susceptibility and brain cellular responses to Alzheimer's disease (AD) and are increasingly considered potential soluble biomarkers in cerebrospinal fluid (CSF) and plasma. To delineate the pathological correlations of distinct lipid species, we conducted a comprehensive characterization of both spatially localized and global differences in brain lipid composition in AppNL-G-F mice with spatial and bulk mass spectrometry lipidomic profiling, using human amyloid-expressing (h-Aβ) and WT mouse brains controls. We observed age-dependent increases in lysophospholipids, bis(monoacylglycerol) phosphates, and phosphatidylglycerols around Aβ plaques in AppNL-G-F mice. Immunohistology-based co-localization identified associations between focal pro-inflammatory lipids, glial activation, and autophagic flux disruption. Likewise, in human donors with varying Braak stages, similar studies of cortical sections revealed co-expression of lysophospholipids and ceramides around Aβ plaques in AD (Braak stage V/VI) but not in earlier Braak stage controls. Our findings in mice provide evidence of temporally and spatially heterogeneous differences in lipid composition as local and global Aβ-related pathologies evolve. Observing similar lipidomic changes associated with pathological Aβ plaques in human AD tissue provides a foundation for understanding differences in CSF lipids with reported clinical stage or disease severity.
Collapse
Affiliation(s)
- Helen Xuexia Huang
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Paolo Inglese
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jiabin Tang
- Department of Brain Sciences, Imperial College London, London, UK
| | - Riad Yagoubi
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Gonçalo D S Correia
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Stephane Camuzeaux
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Vincen Wu
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Maksym V Kopanitsa
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Nanet Willumsen
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Johanna S Jackson
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Steve Gentlemen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Zoltan Takats
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Paul M Matthews
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| |
Collapse
|
5
|
Lv N, Wang L, Zeng M, Wang Y, Yu B, Zeng W, Jiang X, Suo Y. Saponins as therapeutic candidates for atherosclerosis. Phytother Res 2024; 38:1651-1680. [PMID: 38299680 DOI: 10.1002/ptr.8128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/25/2023] [Accepted: 01/06/2024] [Indexed: 02/02/2024]
Abstract
Drug development for atherosclerosis, the underlying pathological state of ischemic cardiovascular diseases, has posed a longstanding challenge. Saponins, classified as steroid or triterpenoid glycosides, have shown promising therapeutic potential in the treatment of atherosclerosis. Through an exhaustive examination of scientific literature spanning from May 2013 to May 2023, we identified 82 references evaluating 37 types of saponins in terms of their prospective impacts on atherosclerosis. These studies suggest that saponins have the potential to ameliorate atherosclerosis by regulating lipid metabolism, inhibiting inflammation, suppressing apoptosis, reducing oxidative stress, and modulating smooth muscle cell proliferation and migration, as well as regulating gut microbiota, autophagy, endothelial senescence, and angiogenesis. Notably, ginsenosides exhibit significant potential and manifest essential pharmacological attributes, including lipid-lowering, anti-inflammatory, anti-apoptotic, and anti-oxidative stress effects. This review provides a comprehensive examination of the pharmacological attributes of saponins in atherosclerosis, with particular emphasis on their role in the regulation of lipid metabolism regulation and anti-inflammatory effects. Thus, saponins may warrant further investigation as a potential therapy for atherosclerosis. However, due to various reasons such as low oral bioavailability, the clinical application of saponins in the treatment of atherosclerosis still needs further exploration.
Collapse
Affiliation(s)
- Nuan Lv
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luming Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miao Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yijing Wang
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenyun Zeng
- Oncology Department, Ganzhou people's hospital, Ganzhou, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanrong Suo
- Traditional Chinese Medicine Department, Ganzhou people's hospital, Ganzhou, China
| |
Collapse
|
6
|
Sasidharan K, Caddeo A, Jamialahmadi O, Noto FR, Tomasi M, Malvestiti F, Ciociola E, Tavaglione F, Mancina RM, Cherubini A, Bianco C, Mirarchi A, Männistö V, Pihlajamäki J, Kärjä V, Grimaudo S, Luukkonen PK, Qadri S, Yki-Järvinen H, Petta S, Manfrini S, Vespasiani-Gentilucci U, Bruni V, Valenti L, Romeo S. IL32 downregulation lowers triglycerides and type I collagen in di-lineage human primary liver organoids. Cell Rep Med 2024; 5:101352. [PMID: 38232700 PMCID: PMC10829727 DOI: 10.1016/j.xcrm.2023.101352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 09/26/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024]
Abstract
Steatotic liver disease (SLD) prevails as the most common chronic liver disease yet lack approved treatments due to incomplete understanding of pathogenesis. Recently, elevated hepatic and circulating interleukin 32 (IL-32) levels were found in individuals with severe SLD. However, the mechanistic link between IL-32 and intracellular triglyceride metabolism remains to be elucidated. We demonstrate in vitro that incubation with IL-32β protein leads to an increase in intracellular triglyceride synthesis, while downregulation of IL32 by small interfering RNA leads to lower triglyceride synthesis and secretion in organoids from human primary hepatocytes. This reduction requires the upregulation of Phospholipase A2 group IIA (PLA2G2A). Furthermore, downregulation of IL32 results in lower intracellular type I collagen levels in di-lineage human primary hepatic organoids. Finally, we identify a genetic variant of IL32 (rs76580947) associated with lower circulating IL-32 and protection against SLD measured by non-invasive tests. These data suggest that IL32 downregulation may be beneficial against SLD.
Collapse
Affiliation(s)
- Kavitha Sasidharan
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Andrea Caddeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Francesca Rita Noto
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Melissa Tomasi
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Malvestiti
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Federica Tavaglione
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Alessandro Cherubini
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristiana Bianco
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Angela Mirarchi
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Clinical Nutrition and Obesity Centre, Kuopio University Hospital, Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Stefania Grimaudo
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Panu K Luukkonen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Sami Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Silvia Manfrini
- Operative Unit of Endocrinology and Diabetes, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Endocrinology and Diabetes, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Vincenzo Bruni
- Operative Unit of Bariatric Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center Unit, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy.
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
7
|
Miyazaki T. Calpain and Cardiometabolic Diseases. Int J Mol Sci 2023; 24:16782. [PMID: 38069105 PMCID: PMC10705917 DOI: 10.3390/ijms242316782] [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: 10/13/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Calpain is defined as a member of the superfamily of cysteine proteases possessing the CysPC motif within the gene. Calpain-1 and -2, which are categorized as conventional isozymes, execute limited proteolysis in a calcium-dependent fashion. Accordingly, the calpain system participates in physiological and pathological phenomena, including cell migration, apoptosis, and synaptic plasticity. Recent investigations have unveiled the contributions of both conventional and unconventional calpains to the pathogenesis of cardiometabolic disorders. In the context of atherosclerosis, overactivation of conventional calpain attenuates the barrier function of vascular endothelial cells and decreases the immunosuppressive effects attributed to lymphatic endothelial cells. In addition, calpain-6 induces aberrant mRNA splicing in macrophages, conferring atheroprone properties. In terms of diabetes, polymorphisms of the calpain-10 gene can modify insulin secretion and glucose disposal. Moreover, conventional calpain reportedly participates in amino acid production from vascular endothelial cells to induce alteration of amino acid composition in the liver microenvironment, thereby facilitating steatohepatitis. Such multifaceted functionality of calpain underscores its potential as a promising candidate for pharmaceutical targets for the treatment of cardiometabolic diseases. Consequently, the present review highlights the pivotal role of calpains in the complications of cardiometabolic diseases and embarks upon a characterization of calpains as molecular targets.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Tokyo 142-8555, Japan
| |
Collapse
|
8
|
Kong F, Wu T, Dai J, Zhai Z, Cai J, Zhu Z, Xu Y, Sun T. Glucagon-like peptide 1 (GLP-1) receptor agonists in experimental Alzheimer's disease models: a systematic review and meta-analysis of preclinical studies. Front Pharmacol 2023; 14:1205207. [PMID: 37771725 PMCID: PMC10525376 DOI: 10.3389/fphar.2023.1205207] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023] Open
Abstract
Alzheimer's disease (AD) is a degenerative disease of the nervous system. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), a drug used to treat type 2 diabetes, have been shown to have neuroprotective effects. This systematic review and meta-analysis evaluated the effects and potential mechanisms of GLP-1 RAs in AD animal models. 26 studies were included by searching relevant studies from seven databases according to a predefined search strategy and inclusion criteria. Methodological quality was assessed using SYRCLE's risk of bias tool, and statistical analysis was performed using ReviewManger 5.3. The results showed that, in terms of behavioral tests, GLP-1 RAs could improve the learning and memory abilities of AD rodents; in terms of pathology, GLP-1 RAs could reduce Aβ deposition and phosphorylated tau levels in the brains of AD rodents. The therapeutic potential of GLP-1 RAs in AD involves a range of mechanisms that work synergistically to enhance the alleviation of various pathological manifestations associated with the condition. A total of five clinical trials were retrieved from ClinicalTrials.gov. More large-scale and high-quality preclinical trials should be conducted to more accurately assess the therapeutic effects of GLP-1 RAs on AD.
Collapse
Affiliation(s)
- Fanjing Kong
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianyu Wu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingyi Dai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenwei Zhai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Cai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhishan Zhu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Sun
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Hussein MA, Borik RM, Nafie MS, Abo-Salem HM, Boshra SA, Mohamed ZN. Structure Activity Relationship and Molecular Docking of Some Quinazolines Bearing Sulfamerazine Moiety as New 3CLpro, cPLA2, sPLA2 Inhibitors. Molecules 2023; 28:6052. [PMID: 37630304 PMCID: PMC10460087 DOI: 10.3390/molecules28166052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
The current work was conducted to synthesize several novel anti-inflammatory quinazolines having sulfamerazine moieties as new 3CLpro, cPLA2, and sPLA2 inhibitors. The thioureido derivative 3 was formed when compound 2 was treated with sulfamerazine. Also, compound 3 was reacted with NH2-NH2 in ethanol to produce the N-aminoquinazoline derivative. Additionally, derivative 4 was reacted with 4-hydroxy-3-methoxybenzaldehyde, ethyl chloroacetate, and/or diethyl oxalate to produce quinazoline derivatives 5, 6, and 12, respectively. The results of the pharmacological study indicated that the synthesized 4-6 and 12 derivatives showed good 3CLpro, cPLA2, and sPLA2 inhibitory activity. The IC50 values of the target compounds 4-6, and 12 against the SARS-CoV-2 main protease were 2.012, 3.68, 1.18, and 5.47 µM, respectively, whereas those of baicalein and ivermectin were 1.72 and 42.39 µM, respectively. The IC50 values of the target compounds 4-6, and 12 against sPLA2 were 2.84, 2.73, 1.016, and 4.45 µM, respectively, whereas those of baicalein and ivermectin were 0.89 and 109.6 µM, respectively. The IC50 values of the target compounds 4-6, and 12 against cPLA2 were 1.44, 2.08, 0.5, and 2.39 µM, respectively, whereas those of baicalein and ivermectin were 3.88 and 138.0 µM, respectively. Also, incubation of lung cells with LPS plus derivatives 4-6, and 12 caused a significant decrease in levels of sPLA2, cPLA2, IL-8, TNF-α, and NO. The inhibitory activity of the synthesized compounds was more pronounced compared to baicalein and ivermectin. In contrast to ivermectin and baicalein, bioinformatics investigations were carried out to establish the possible binding interactions between the newly synthesized compounds 2-6 and 12 and the active site of 3CLpro. Docking simulations were utilized to identify the binding affinity and binding mode of compounds 2-6 and 12 with the active sites of 3CLpro, sPLA2, and cPLA2 enzymes. Our findings demonstrated that all compounds had outstanding binding affinities, especially with the key amino acids of the target enzymes. These findings imply that compound 6 is a potential lead for the development of more effective SARS-CoV-2 Mpro inhibitors and anti-COVID-19 quinazoline derivative-based drugs. Compound 6 was shown to have more antiviral activity than baicalein and against 3CLpro. Furthermore, the IC50 value of ivermectin against the SARS-CoV-2 main protease was revealed to be 42.39 µM, indicating that it has low effectiveness.
Collapse
Affiliation(s)
- Mohammed Abdalla Hussein
- Biotechnology Department, Faculty of Applied Heath Science Technology, October 6 University, Giza 28125, Egypt;
| | - Rita M. Borik
- Chemistry Department, Faculty of Science (Female Section), Jazan University, Jazan 82621, Saudi Arabia;
| | - Mohamed S. Nafie
- Chemistry Department (Biochemistry Program), Faculty of Science, Suez Canal University, Ismailia 41522, Egypt;
| | - Heba M. Abo-Salem
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza 28125, Egypt;
| | - Sylvia A. Boshra
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Giza 28125, Egypt
| | - Zahraa N. Mohamed
- Medical Laboratory Department, Faculty of Applied Medical Sciences, October 6 University, Giza 28125, Egypt;
| |
Collapse
|
10
|
Urazov S, Chernov A, Popov O, Klenkova N, Sushentseva N, Polkovnikova I, Apalko S, Kislyuk K, Pavlovich D, Ivanov A, Shcherbak S. Secretory Phospholipase A2 and Interleukin-6 Levels as Predictive Markers of the Severity and Outcome of Patients with COVID-19 Infections. Int J Mol Sci 2023; 24:5540. [PMID: 36982611 PMCID: PMC10059025 DOI: 10.3390/ijms24065540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Coronavirus disease (COVID-19) has become a global pandemic. COVID-19 patients need immediate diagnosis and rehabilitation, which makes it urgent to identify new protein markers for a prognosis of the severity and outcome of the disease. The aim of this study was to analyze the levels of interleukin-6 (IL-6) and secretory phospholipase (sPLA2) in the blood of patients regarding the severity and outcome of COVID-19 infection. The study included clinical and biochemical data obtained from 158 patients with COVID-19 treated at St. Petersburg City Hospital No. 40. A detailed clinical blood test was performed on all patients, as well as an assessment of IL-6, sPLA2, aspartate aminotransferase (AST), total protein, albumin, lactate dehydrogenase (LDH), APTT, fibrinogen, procalcitonin, D-dimer, C-reactive protein (CRB), ferritin, and glomerular filtration rate (GFR) levels. It was found that the levels of PLA2, IL-6, APTV, AST, CRP, LDH, IL-6, D-dimer, and ferritin, as well as the number of neutrophils, significantly increased in patients with mild to severe COVID-19 infections. The levels of IL-6 were positively correlated with APTT; the levels of AST, LDH, CRP, D-dimer, and ferritin; and the number of neutrophils. The increase in the level of sPLA2 was positively correlated with the levels of CRP, LDH, D-dimer, and ferritin, the number of neutrophils, and APTT, and negatively correlated with the levels of GFR and lymphocytes. High levels of IL-6 and PLA2 significantly increase the risk of a severe course by 13.7 and 2.24 times, and increase the risk of death from COVID-19 infection by 14.82 and 5.32 times, respectively. We have shown that the blood levels of sPLA2 and IL-6 increase in cases which eventually result in death and when patients are transferred to the ICU (as the severity of COVID-19 infection increases), showing that IL-6 and sPLA2 can be considered as early predictors of aggravation of COVID-19 infections.
Collapse
Affiliation(s)
- Stanislav Urazov
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | - Alexandr Chernov
- Bioenergetics Department of Life Sciences, Ben-Gurion University, Beer Sheva 84105, Israel
- Federal State Budgetary Institution of Science “Institute of Experimental Medicine”, 197376 St. Petersburg, Russia
| | - Oleg Popov
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | - Natalya Klenkova
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | | | | | - Svetlana Apalko
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | - Kseniya Kislyuk
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | | | - Andrey Ivanov
- Center for Clinical and Laboratory Diagnostics, Federal State Budgetary Military Educational Institution of Higher Education “Military Medical Academy named after S.M. Kirov”, 194044 St. Petersburg, Russia
| | - Sergey Shcherbak
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
- Department of Postgraduate Medical Education, Federal State Budgetary Educational Institution of Higher Professional Education “Saint-Petersburg State University”, 199034 St. Petersburg, Russia
| |
Collapse
|
11
|
Zhang H, Zhang Y, Mu T, Cao J, Liu X, Yang X, Ren D, Zhao K. Response of gut microbiota and ileal transcriptome to inulin intervention in HFD induced obese mice. Int J Biol Macromol 2023; 225:861-872. [PMID: 36402387 DOI: 10.1016/j.ijbiomac.2022.11.151] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/19/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Inulin, as a dietary fiber, exerted prominent anti-obesity effects by modulating gut microbiota. However, the possible relationship and interplay of gut microbiome and function of distal intestine is still unclear now. This study aimed to investigate the possible targets of microbes and the related intestinal genes mediated by inulin. C57 BL/6 male mice were randomly allocated to chow diet (Chow) group, high-fat diet (HFD) group, and HFD supplemented with 3 % inulin (Inulin) group. Compared with HFD treatment, inulin supplementation significantly decreased the body weight, fat deposition, and fasting blood glucose level. In addition, mice treated with inulin had a remarkable alteration in the structure of cecal microbiota and transcriptomic profiling of ileum. In particular, inulin supplementation significantly reversed the HFD induced expression of Bacteroides, Allobaculum and nonrank_f_Bacteroidates_S24-7_group, and reversed the expression of genes belonging to phospholipase A2 (PLA2) family and cytochrome P450 (CYP450) family. In summary, inulin might alleviate HFD-induced fat deposition and metabolic disorders via regulating lipid metabolism of ileum, while the interaction between the sPLA2s and gut microbes might play important roles in the process.
Collapse
Affiliation(s)
- Hong Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, P. R. China; Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China
| | - Yunhui Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China
| | - Tong Mu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China
| | - Jianxin Cao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China
| | - Xiaoxia Liu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China
| | - Ke Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, P. R. China; Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Normal University, Xi'an 710062, P. R. China.
| |
Collapse
|
12
|
Lokhov PG, Balashova EE, Trifonova OP, Maslov DL, Grigoriev AI, Ponomarenko EA, Archakov AI. Mass Spectrometric Blood Metabogram: Acquisition, Characterization, and Prospects for Application. Int J Mol Sci 2023; 24:ijms24021736. [PMID: 36675249 PMCID: PMC9861083 DOI: 10.3390/ijms24021736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
In metabolomics, many metabolites are measured simultaneously in a single run. Such analytical performance opens up prospects for clinical laboratory diagnostics. In this work, a mass spectrometric metabogram was developed as a simplified and clinically applicable way of measuring the blood plasma metabolome. To develop the metabogram, blood plasma samples from healthy male volunteers (n = 48) of approximately the same age, direct infusion mass spectrometry (DIMS) of the low molecular fraction of samples, and principal component analysis (PCA) of the mass spectra were used. The seven components of the metabogram defined by PCA, which cover ~70% of blood plasma metabolome variability, were characterized using a metabolite set enrichment analysis (MSEA) and clinical test results of participating volunteers. It has been established that the components of the metabogram are functionally related groups of the blood metabolome associated with regulation, lipid-carbohydrate, and lipid-amine blood components, eicosanoids, lipid intake into the organism, and liver function thereby providing a lot of clinically relevant information. Therefore, metabogram provides the possibility to apply the metabolomics performance in the clinic. The features of the metabogram are also discussed in comparison with the thin-layer chromatography and with the analysis of blood metabolome by liquid chromatography combined with mass spectrometry.
Collapse
Affiliation(s)
- Petr G. Lokhov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
- Correspondence:
| | - Elena E. Balashova
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| | - Oxana P. Trifonova
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| | - Dmitry L. Maslov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| | - Anatoly I. Grigoriev
- Institute of Biomedical Problems, Russian Federation State Scientific Research Center, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Elena A. Ponomarenko
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| | - Alexander I. Archakov
- Institute of Biomedical Chemistry, 10 Building 8, Pogodinskaya Street, 119121 Moscow, Russia
| |
Collapse
|
13
|
Tao S, Yang M, Pan B, Wang Y, Tian F, Han D, Shao W, Yang W, Xie Y, Fang X, Xia M, Hu J, Kan H, Li W, Xu Y. Maternal exposure to ambient PM 2.5 perturbs the metabolic homeostasis of maternal serum and placenta in mice. ENVIRONMENTAL RESEARCH 2023; 216:114648. [PMID: 36341790 DOI: 10.1016/j.envres.2022.114648] [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: 07/26/2022] [Revised: 10/02/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Epidemiological and animal studies have shown that maternal fine particulate matters (PM2.5) exposure correlates with various adverse pregnancy outcomes such as low birth weight (LBW) of offspring. However, the underlying biological mechanisms have not been fully understood. In this study, female C57Bl/6 J mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CAP) during pregestational and gestational periods, and metabolomics was performed to analyze the metabolic features in maternal serum and placenta by liquid chromatography-mass spectrometry (LC-MS). The partial least squares discriminate analysis (PLS-DA) displayed evident clustering of FA- and CAP-exposed samples for both maternal serum and placenta. In addition, pathway analysis identified that vitamin digestion and absorption was perturbed in maternal serum, while metabolic pathways including arachidonic acid metabolism, serotonergic synapse, 2-oxocarboxylic acid metabolism and cAMP signaling pathway were perturbed in placenta. Further analysis indicated that CAP exposure influenced the nutrient transportation capacity of placenta, by not only changing the ratios of some critical metabolites in placenta to maternal serum but also significantly altering the expressions of nutrition transporters in placenta. These findings reaffirm the importance of protecting women from PM2.5 exposure, and also advance our understanding of the toxic actions of ambient PM2.5.
Collapse
Affiliation(s)
- Shimin Tao
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China; NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Mingjun Yang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Bin Pan
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China; NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Yuzhu Wang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Fang Tian
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Dongyang Han
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Wenpu Shao
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Wenhui Yang
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Yuanting Xie
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Xinyi Fang
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Minjie Xia
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Jingying Hu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Haidong Kan
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Weihua Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
Singh V, Rai R, Mathew BJ, Chourasia R, Singh AK, Kumar A, Chaurasiya SK. Phospholipase C: underrated players in microbial infections. Front Cell Infect Microbiol 2023; 13:1089374. [PMID: 37139494 PMCID: PMC10149971 DOI: 10.3389/fcimb.2023.1089374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/21/2023] [Indexed: 05/05/2023] Open
Abstract
During bacterial infections, one or more virulence factors are required to support the survival, growth, and colonization of the pathogen within the host, leading to the symptomatic characteristic of the disease. The outcome of bacterial infections is determined by several factors from both host as well as pathogen origin. Proteins and enzymes involved in cellular signaling are important players in determining the outcome of host-pathogen interactions. phospholipase C (PLCs) participate in cellular signaling and regulation by virtue of their ability to hydrolyze membrane phospholipids into di-acyl-glycerol (DAG) and inositol triphosphate (IP3), which further causes the activation of other signaling pathways involved in various processes, including immune response. A total of 13 PLC isoforms are known so far, differing in their structure, regulation, and tissue-specific distribution. Different PLC isoforms have been implicated in various diseases, including cancer and infectious diseases; however, their roles in infectious diseases are not clearly understood. Many studies have suggested the prominent roles of both host and pathogen-derived PLCs during infections. PLCs have also been shown to contribute towards disease pathogenesis and the onset of disease symptoms. In this review, we have discussed the contribution of PLCs as a determinant of the outcome of host-pathogen interaction and pathogenesis during bacterial infections of human importance.
Collapse
Affiliation(s)
- Vinayak Singh
- Molecular Signalling Lab, Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, Madhya Pradesh, India
| | - Rupal Rai
- Molecular Signalling Lab, Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, Madhya Pradesh, India
| | - Bijina J. Mathew
- Molecular Signalling Lab, Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, Madhya Pradesh, India
| | - Rashmi Chourasia
- Department of Chemistry, IES University, Bhopal, Madhya Pradesh, India
| | - Anirudh K. Singh
- School of Sciences, SAM Global University, Raisen, Madhya Pradesh, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, India
| | - Shivendra K. Chaurasiya
- Molecular Signalling Lab, Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, Madhya Pradesh, India
- *Correspondence: Shivendra K. Chaurasiya,
| |
Collapse
|
15
|
Yu T, Wu H, Huang Q, Dong F, Li X, Zhang Y, Duan R, Niu H, Yang T. Outdoor particulate matter exposure affects metabolome in chronic obstructive pulmonary disease: Preliminary study. Front Public Health 2023; 11:1069906. [PMID: 37026137 PMCID: PMC10070744 DOI: 10.3389/fpubh.2023.1069906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/03/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction The metabolomic changes caused by airborne fine particulate matter (PM2.5) exposure in patients with chronic obstructive pulmonary disease (COPD) remain unclear. The aim of this study was to determine whether it is possible to predict PM2.5-induced acute exacerbation of COPD (AECOPD) using metabolic markers. Methods Thirty-eight patients with COPD diagnosed by the 2018 Global Initiative for Obstructive Lung Disease were selected and divided into high exposure and low exposure groups. Questionnaire data, clinical data, and peripheral blood data were collected from the patients. Targeted metabolomics using liquid chromatography-tandem mass spectrometry was performed on the plasma samples to investigate the metabolic differences between the two groups and its correlation with the risk of acute exacerbation. Results Metabolomic analysis identified 311 metabolites in the plasma of patients with COPD, among which 21 metabolites showed significant changes between the two groups, involving seven pathways, including glycerophospholipid, alanine, aspartate, and glutamate metabolism. Among the 21 metabolites, arginine and glycochenodeoxycholic acid were positively associated with AECOPD during the three months of follow-up, with an area under the curve of 72.50% and 67.14%, respectively. Discussion PM2.5 exposure can lead to changes in multiple metabolic pathways that contribute to the development of AECOPD, and arginine is a bridge between PM2.5 exposure and AECOPD.
Collapse
Affiliation(s)
- Tao Yu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hanna Wu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Qingxia Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Fudan University, Shanghai, China
| | - Fen Dong
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Xuexin Li
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yushi Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ruirui Duan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Hongtao Niu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
- Hongtao Niu
| | - Ting Yang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Respiratory Diseases, Beijing, China
- *Correspondence: Ting Yang
| |
Collapse
|
16
|
Kurano M, Saito Y, Uranbileg B, Saigusa D, Kano K, Aoki J, Yatomi Y. Modulations of bioactive lipids and their receptors in postmortem Alzheimer's disease brains. Front Aging Neurosci 2022; 14:1066578. [PMID: 36570536 PMCID: PMC9780287 DOI: 10.3389/fnagi.2022.1066578] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background Analyses of brain samples from Alzheimer's disease (AD) patients may be expected to help us improve our understanding of the pathogenesis of AD. Bioactive lipids, including sphingolipids, glycerophospholipids, and eicosanoids/related mediators have been demonstrated to exert potent physiological actions and to be involved in the pathogenesis of various human diseases. In this cross-sectional study, we attempted to elucidate the associations of these bioactive lipids with the pathogenesis/pathology of AD through postmortem studies of human brains. Methods We measured the levels of glycerophospholipids, sphingolipids, and eicosanoids/related mediators in the brains of patients with AD (AD brains), patients with Cerad score B (Cerad-b brains), and control subjects (control brains), using a liquid chromatography-mass spectrometry method; we also measured the mRNA levels of specific receptors for these bioactive lipids in the same brain specimens. Results The levels of several species of sphingomyelins and ceramides were higher in the Cerad-b and AD brains. Levels of several species of lysophosphatidic acids (LPAs), lysophosphatidylcholine, lysophosphatidylserine, lysophosphatidylethanolamine (LPE), lysophosphatidylinositol, phosphatidylcholine, phosphatidylserine (PS), phosphatidylethanolamine (PE), phosphatidylinositol, and phosphatidylglycerol were especially high in the Cerad-b brains, while those of lysophosphatidylglycerol (LPG) were especially high in the AD brains. Several eicosanoids, including metabolites of prostaglandin E2, oxylipins, metabolites of epoxide, and metabolites of DHA and EPA, such as resolvins, were also modulated in the AD brains. Among the lipid mediators, the levels of S1P2, S1P5, LPA1, LPA2, LPA6, P2Y10, GPR174, EP1, DP1, DP2, IP, FP, and TXA2r were lower in the AD and/or Cerad-b brains. The brain levels of ceramides, LPC, LPI, PE, and PS showed strong positive correlations with the Aβ contents, while those of LPG showed rather strong positive correlations with the presence of senile plaques and neurofibrillary tangles. A discriminant analysis revealed that LPG is especially important for AD and the LPE/PE axis is important for Cerad-b. Conclusions Comprehensive lipidomics, together with the measurement of lipid receptor expression levels provided novel evidence for the associations of bioactive lipids with AD, which is expected to facilitate future translational research and reverse translational research.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan,*Correspondence: Makoto Kurano,
| | - Yuko Saito
- Tokyo Metropolitan Geriatric Hospital, Institute of Gerontology, Tokyo, Japan
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Zhang L, Wen X, Hou Y, Yang Y, Song W, Zeng Y, Sun J. Integrated metabolomics and lipidomics study of patients with atopic dermatitis in response to dupilumab. Front Immunol 2022; 13:1002536. [PMID: 36341398 PMCID: PMC9632449 DOI: 10.3389/fimmu.2022.1002536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/04/2022] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases. Dupilumab, a monoclonal antibody that targets the interleukin (IL)-4 and IL-13 receptors, has been widely used in AD because of its efficacy. However, metabolic changes occurring in patients with AD in response to dupilumab remains unknown. In this study, we integrated metabolomics and lipidomics analyses with clinical data to explore potential metabolic alterations associated with dupilumab therapeutic efficacy. In addition, we investigated whether the development of treatment side effects was linked to the dysregulation of metabolic pathways. METHODS A total of 33 patients with AD were included in the current study, with serum samples collected before and after treatment with dupilumab. Comprehensive metabolomic and lipidomic analyses have previously been developed to identify serum metabolites (including lipids) that vary among treatment groups. An orthogonal partial least squares discriminant analysis model was established to screen for differential metabolites and metabolites with variable importance in projection > 1 and p < 0.05 were considered potential metabolic biomarkers. MetaboAnalyst 5.0 was used to identify related metabolic pathways. Patients were further classified into two groups, well responders (n = 19) and poor responders (n = 14), to identify differential metabolites between the two groups. RESULTS The results revealed significant changes in serum metabolites before and after 16 weeks of dupilumab treatment. Variations in the metabolic profile were more significant in the well-responder group than in the poor-responder group. Pathway enrichment analysis revealed that differential metabolites derived from the well-responder group were mainly involved in glycerophospholipid metabolism, valine, leucine and isoleucine biosynthesis, the citrate cycle, arachidonic acid metabolism, pyrimidine metabolism, and sphingolipid metabolism. CONCLUSION Serum metabolic profiles of patients with AD varied significantly after treatment with dupilumab. Differential metabolites and their related metabolic pathways may provide clues for understanding the effects of dupilumab on patient metabolism.
Collapse
Affiliation(s)
- Lishan Zhang
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueyi Wen
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yibo Hou
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongshi Yang
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Song
- Medical Science Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yueping Zeng
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Jinlyu Sun
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Ritz B, Yan Q, He D, Wu J, Walker DI, Uppal K, Jones DP, Heck JE. Child serum metabolome and traffic-related air pollution exposure in pregnancy. ENVIRONMENTAL RESEARCH 2022; 203:111907. [PMID: 34419469 PMCID: PMC8926017 DOI: 10.1016/j.envres.2021.111907] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 05/08/2023]
Abstract
BACKGROUND Maternal exposure to traffic-related air pollution during pregnancy has been shown to increase the risk of adverse birth outcomes and childhood disorders. High-resolution metabolomics (HRM) has previously been employed to identify metabolic responses to traffic-related air pollution in adults, including pregnant women. Thus far, no studies have examined metabolic effects of air pollution exposure in utero on neonates. METHODS We retrieved stored neonatal blood spots for 241 children born in California between 1998 and 2007. These children were randomly selected from all California birth rolls to serve as birth-year matched controls for children with retinoblastoma identified from the California cancer registry for a case control study of childhood cancer. We estimated prenatal traffic-related air pollution exposure (particulate matter less than 2.5 μm (PM2.5)) during the third-trimester using the California Line Source Dispersion Model, version 4 (CALINE4) based on residential addresses recorded at birth. We employed untargeted HRM to obtain metabolic profiles, and metabolites associated with air pollution exposure were identified using partial least squares (PLS) regression and linear regressions. Biological effects were characterized using pathway enrichment analyses adjusting for potential confounders including maternal age, race/ethnicity, and education. RESULTS In total we extracted 4038 and 4957 metabolite features from neonatal blood spots in hydrophilic interaction (HILIC) chromatography (positive ion mode) and C18 reverse phase columns (negative ion mode), respectively. After controlling for confounding factors, partial least square regression (Variable Importance in Projection (VIP) ≥ 2) selected 402 HILIC positive and 182 C18 negative features as statistically significantly associated with increasing third trimester PM2.5 exposure. Using pathway enrichment analysis, we identified metabolites in oxidative stress and inflammation pathways as being altered, primarily involving lipid metabolism. CONCLUSION The metabolite features and pathways associated with air pollution exposure in neonates suggest that maternal exposure during late pregnancy contributes to oxidative stress and inflammation in newborn children.
Collapse
Affiliation(s)
- Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Neurology, UCLA School of Medicine, CA, USA.
| | - Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Di He
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Jun Wu
- Program in Public Health, UCI Susan and Henry Samueli College of Health Sciences, Irvine, CA, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Department of Medicine, Emory University, Atlanta, GA, USA
| | - Julia E Heck
- College of Health and Public Service, University of North Texas, Denton, TX, USA
| |
Collapse
|
19
|
Classification and Functional Analysis between Cancer and Normal Tissues Using Explainable Pathway Deep Learning through RNA-Sequencing Gene Expression. Int J Mol Sci 2021; 22:ijms222111531. [PMID: 34768960 PMCID: PMC8584109 DOI: 10.3390/ijms222111531] [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: 09/09/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 11/24/2022] Open
Abstract
Deep learning has proven advantageous in solving cancer diagnostic or classification problems. However, it cannot explain the rationale behind human decisions. Biological pathway databases provide well-studied relationships between genes and their pathways. As pathways comprise knowledge frameworks widely used by human researchers, representing gene-to-pathway relationships in deep learning structures may aid in their comprehension. Here, we propose a deep neural network (PathDeep), which implements gene-to-pathway relationships in its structure. We also provide an application framework measuring the contribution of pathways and genes in deep neural networks in a classification problem. We applied PathDeep to classify cancer and normal tissues based on the publicly available, large gene expression dataset. PathDeep showed higher accuracy than fully connected neural networks in distinguishing cancer from normal tissues (accuracy = 0.994) in 32 tissue samples. We identified 42 pathways related to 32 cancer tissues and 57 associated genes contributing highly to the biological functions of cancer. The most significant pathway was G-protein-coupled receptor signaling, and the most enriched function was the G1/S transition of the mitotic cell cycle, suggesting that these biological functions were the most common cancer characteristics in the 32 tissues.
Collapse
|
20
|
Yassine HN, Solomon V, Thakral A, Sheikh-Bahaei N, Chui HC, Braskie MN, Schneider LS, Talbot K. Brain energy failure in dementia syndromes: Opportunities and challenges for glucagon-like peptide-1 receptor agonists. Alzheimers Dement 2021; 18:478-497. [PMID: 34647685 PMCID: PMC8940606 DOI: 10.1002/alz.12474] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/11/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022]
Abstract
Medications for type 2 diabetes (T2DM) offer a promising path for discovery and development of effective interventions for dementia syndromes. A common feature of dementia syndromes is an energy failure due to reduced energy supply to neurons and is associated with synaptic loss and results in cognitive decline and behavioral changes. Among diabetes medications, glucagon‐like peptide‐1 (GLP‐1) receptor agonists (RAs) promote protective effects on vascular, microglial, and neuronal functions. In this review, we present evidence from animal models, imaging studies, and clinical trials that support developing GLP‐1 RAs for dementia syndromes. The review examines how changes in brain energy metabolism differ in conditions of insulin resistance and T2DM from dementia and underscores the challenges that arise from the heterogeneity of dementia syndromes. The development of GLP‐1 RAs as dementia therapies requires a deeper understanding of the regional changes in brain energy homeostasis guided by novel imaging biomarkers.
Collapse
Affiliation(s)
- Hussein N Yassine
- Department of Medicine, University of Southern California, Keck School of Medicine USC, Los Angeles, California, USA.,Department of Neurology, University of Southern California, Keck School of Medicine USC, Los Angeles, California, USA
| | - Victoria Solomon
- Department of Medicine, University of Southern California, Keck School of Medicine USC, Los Angeles, California, USA
| | - Angad Thakral
- Department of Medicine, University of Southern California, Keck School of Medicine USC, Los Angeles, California, USA
| | - Nasim Sheikh-Bahaei
- Department of Radiology, Keck School of Medicine USC, Los Angeles, California, USA
| | - Helena C Chui
- Department of Neurology, University of Southern California, Keck School of Medicine USC, Los Angeles, California, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, USC, Los Angeles, California, USA
| | - Lon S Schneider
- Department of Neurology, University of Southern California, Keck School of Medicine USC, Los Angeles, California, USA.,Department of Psychiatry and Behavioral Sciences, Keck School of Medicine USC, Los Angeles, California, USA
| | - Konrad Talbot
- Departments of Neurosurgery, Pathology and Human Anatomy, and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
21
|
Zhu K, Browne RW, Blair RH, Bonner MR, Tian M, Niu Z, Deng F, Farhat Z, Mu L. Changes in arachidonic acid (AA)- and linoleic acid (LA)-derived hydroxy metabolites and their interplay with inflammatory biomarkers in response to drastic changes in air pollution exposure. ENVIRONMENTAL RESEARCH 2021; 200:111401. [PMID: 34089746 PMCID: PMC11483949 DOI: 10.1016/j.envres.2021.111401] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/20/2021] [Accepted: 05/23/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Untargeted metabolomics analyses have indicated that fatty acids and their hydroxy derivatives may be important metabolites in the mechanism through which air pollution potentiates diseases. This study aimed to use targeted analysis to investigate how metabolites in arachidonic acid (AA) and linoleic acid (LA) pathways respond to short-term changes in air pollution exposure. We further explored how they might interact with markers of antioxidant enzymes and systemic inflammation. METHODS This study included a subset of participants (n = 53) from the Beijing Olympics Air Pollution (BoaP) study in which blood samples were collected before, during, and after the Beijing Olympics. Hydroxy fatty acids were measured by liquid chromatography/mass spectrometry (LC/MS). Native total fatty acids were measured as fatty acid methyl esters (FAMEs) using gas chromatography. A set of chemokines were measured by ELISA-based chemiluminescent assay and antioxidant enzyme activities were analyzed by kinetic enzyme assays. Changes in levels of metabolites over the three time points were examined using linear mixed-effects models, adjusting for age, sex, body mass index (BMI), and smoking status. Pearson correlation and repeated measures correlation coefficients were calculated to explore the relationships of metabolites with levels of serum chemokines and antioxidant enzymes. RESULTS 12-hydroxyeicosatetraenoic acid (12-HETE) decreased by 50.5% (95% CI: -66.5, -34.5; p < 0.0001) when air pollution dropped during the Olympics and increased by 119.4% (95% CI: 36.4, 202.3; p < 0.0001) when air pollution returned to high levels after the Olympics. In contrast, 13-hydroxyoctadecadienoic acid (13-HODE) elevated significantly (p = 0.023) during the Olympics and decreased nonsignificantly after the games (p = 0.104). Interleukin 8 (IL-8) correlated with 12-HETE (r = 0.399, BH-adjusted p = 0.004) and 13-HODE (r = 0.342, BH-adjusted p = 0.014) over the three points; it presented a positive and moderate correlation with 12-HETE during the Olympics (r = 0.583, BH-adjusted p = 0.002) and with 13-HODE before the Olympics (r = 0.543, BH-adjusted p = 0.008). CONCLUSION AA- and LA-derived hydroxy metabolites are associated with air pollution and might interact with systemic inflammation in response to air pollution exposure.
Collapse
Affiliation(s)
- Kexin Zhu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Richard W Browne
- Department of Biotechnical and Clinical Laboratory Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Rachael Hageman Blair
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Matthew R Bonner
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Mingmei Tian
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Zhongzheng Niu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Furong Deng
- Department of Occupational and Environmental Health, School of Public Health, Peking University, Beijing, China
| | - Zeinab Farhat
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Lina Mu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA.
| |
Collapse
|
22
|
Kitsiouli E, Tenopoulou M, Papadopoulos S, Lekka ME. Phospholipases A2 as biomarkers in ARDS. Biomed J 2021; 44:663-670. [PMID: 34478892 PMCID: PMC8847824 DOI: 10.1016/j.bj.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a multifactorial life-threatening lung injury, characterized by diffuse lung inflammation and increased alveolocapillary barrier permeability. The different stages of ARDS have distinctive biochemical and clinical profiles. Despite the progress of our understanding on ARDS pathobiology, the mechanisms underlying its pathogenesis are still obscure. Herein, we review the existing literature about the implications of phospholipases 2 (PLA2s), a large family of enzymes that catalyze the hydrolysis of fatty acids at the sn-2 position of glycerophospholipids, in ARDS-related pathology. We emphasize on the versatile way of participation of different PLA2s isoforms in the distinct ARDS subgroup phenotypes by either potentiating lung inflammation and damage or by preserving the normal lung. Current research supports that PLA2s are associated with the progression and the outcome of ARDS. We herein discuss the transcellular communication of PLA2s through secreted extracellular vesicles and suggest it as a new mechanism of PLA2s involvement in ARDS. Thus, the elucidation of the spatiotemporal features of PLA2s expression may give new insights and provide valuable information about the risk of an individual to develop ARDS or advance to more severe stages, and potentially identify PLA2 isoforms as biomarkers and target for pharmacological intervention.
Collapse
Affiliation(s)
- Eirini Kitsiouli
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Margarita Tenopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Stylianos Papadopoulos
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Marilena E Lekka
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
23
|
Kim JY, Jang HJ, Leem J, Kim GM. Protective Effects of Bee Venom-Derived Phospholipase A 2 against Cholestatic Liver Disease in Mice. Biomedicines 2021; 9:biomedicines9080992. [PMID: 34440196 PMCID: PMC8394029 DOI: 10.3390/biomedicines9080992] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022] Open
Abstract
Hepatocyte apoptosis and inflammation play important roles in cholestatic liver diseases. Bee venom-derived secretory phospholipase A2 (bvPLA2) has been shown to ameliorate various inflammatory diseases. However, whether bvPLA2 has a therapeutic effect against cholestatic liver disease has not been evaluated. Therefore, we investigated the effects of bvPLA2 on cholestatic liver injury and fibrosis in a murine model of 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet feeding. The administration of bvPLA2 ameliorated liver damage, cholestasis, and fibrosis in DDC diet-fed mice, as assessed by serum biochemical tests and histological examinations. In addition, bvPLA2 reduced myofibroblast accumulation, concomitant with suppression of transforming growth factor-β signaling cascade. The administration of bvPLA2 inhibited hepatocyte apoptosis in DDC diet-fed mice as represented by a reduction in the number of cells stained with terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and suppression of caspase-3 activation. Moreover, bvPLA2 reduced cytokine production along with the inhibition of the nuclear factor kappa-B pathway. The number of regulatory T-cells was increased by bvPLA2, while the number of other immune cells, including neutrophils, macrophages, and CD8+ T-cells, was decreased. Our data indicate that the administration of bvPLA2 ameliorates cholestatic liver injury and fibrosis by inhibiting hepatocyte apoptosis and inflammation.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Hyo-Jeong Jang
- Department of Pediatrics, School of Medicine, Keimyung University, Daegu 42601, Korea;
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
- Correspondence: (J.L.); (G.-M.K.)
| | - Gyun-Moo Kim
- Department of Emergency Medicine, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea
- Correspondence: (J.L.); (G.-M.K.)
| |
Collapse
|
24
|
Al-Shaer AE, Buddenbaum N, Shaikh SR. Polyunsaturated fatty acids, specialized pro-resolving mediators, and targeting inflammation resolution in the age of precision nutrition. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158936. [PMID: 33794384 PMCID: PMC8496879 DOI: 10.1016/j.bbalip.2021.158936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 12/16/2022]
Abstract
Chronic inflammation contributes toward the pathogenesis of numerous diseases including, but not limited to, obesity, autoimmunity, cardiovascular diseases, and cancers. The discovery of specialized pro-resolving mediators (SPMs), which are critical for resolving inflammation, has commenced investigation into targeting pathways of inflammation resolution to improve physiological outcomes. SPMs are predominately synthesized from the n-3 polyunsaturated fatty acids (PUFA) eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids. Therefore, one viable strategy to promote inflammation resolution would be to increase dietary intake of EPA/DHA, which are deficient in select populations. However, there are inconsistencies between the use of EPA/DHA as dietary or pharmacological supplements and improved inflammatory status. Herein, we review the literature on the relationship between the high n-6/n-3 PUFA ratio, downstream SPM biosynthesis, and inflammatory endpoints. We highlight key studies that have investigated how dietary intake of EPA/DHA increase tissue SPMs and their effects on inflammation. We also discuss the biochemical pathways by which EPA/DHA drive SPM biosynthesis and underscore mechanistic gaps in knowledge about these pathways which include a neglect for host genetics/ethnic differences in SPM metabolism, sexual dimorphism in SPM levels, and potential competition from select dietary n-6 PUFAs for enzymes of SPM synthesis. Altogether, establishing how dietary PUFAs control SPM biosynthesis in a genetic- and sex-dependent manner will drive new precision nutrition studies with EPA/DHA to prevent chronic inflammation in select populations.
Collapse
Affiliation(s)
- Abrar E Al-Shaer
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, The University of North Carolina at Chapel Hill, 170 Rosenau Hall, CB# 7400, 135 Dauer Drive, Chapel Hill, NC, USA
| | - Nicole Buddenbaum
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, The University of North Carolina at Chapel Hill, 170 Rosenau Hall, CB# 7400, 135 Dauer Drive, Chapel Hill, NC, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, The University of North Carolina at Chapel Hill, 170 Rosenau Hall, CB# 7400, 135 Dauer Drive, Chapel Hill, NC, USA.
| |
Collapse
|
25
|
Nassan FL, Wang C, Kelly RS, Lasky-Su JA, Vokonas PS, Koutrakis P, Schwartz JD. Ambient PM 2.5 species and ultrafine particle exposure and their differential metabolomic signatures. ENVIRONMENT INTERNATIONAL 2021; 151:106447. [PMID: 33639346 PMCID: PMC7994935 DOI: 10.1016/j.envint.2021.106447] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/03/2021] [Indexed: 05/09/2023]
Abstract
BACKGROUND The metabolomic signatures of short- and long-term exposure to PM2.5 have been reported and linked to inflammation and oxidative stress. However, little is known about the relative contribution of the specific PM2.5 species (hence sources) that drive these metabolomic signatures. OBJECTIVES We aimed to determine the relative contribution of the different species of PM2.5 exposure to the perturbed metabolic pathways related to changes in the plasma metabolome. METHODS We performed mass-spectrometry based metabolomic profiling of plasma samples among men from the Normative Aging Study to identify metabolic pathways associated with PM2.5 species. The exposure windows included short-term (one, seven-, and thirty-day moving average) and long-term (one year moving average). We used linear mixed-effect regression with subject-specific intercepts while simultaneously adjusting for PM2.5, NO2, O3, temperature, relative humidity, and covariates and correcting for multiple testing. We also used independent component analysis (ICA) to examine the relative contribution of patterns of PM2.5 species. RESULTS Between 2000 and 2016, 456 men provided 648 blood samples, in which 1158 metabolites were quantified. We chose 305 metabolites for the short-term and 288 metabolites for the long-term exposure in this analysis that were significantly associated (p-value < 0.01) with PM2.5 to include in our PM2.5 species analysis. On average, men were 75.0 years old and their body mass index was 27.7 kg/m2. Only 3% were current smokers. In the adjusted models, ultrafine particles (UFPs) were the most significant species of short-term PM2.5 exposure followed by nickel, vanadium, potassium, silicon, and aluminum. Black carbon, vanadium, zinc, nickel, iron, copper, and selenium were the significant species of long-term PM2.5 exposure. We identified several metabolic pathways perturbed with PM2.5 species including glycerophospholipid, sphingolipid, and glutathione. These pathways are involved in inflammation, oxidative stress, immunity, and nucleic acid damage and repair. Results were overlapped with the ICA. CONCLUSIONS We identified several significant perturbed plasma metabolites and metabolic pathways associated with exposure to PM2.5 species. These species are associated with traffic, fuel oil, and wood smoke. This is the largest study to report a metabolomic signature of PM2.5 species' exposure and the first to use ICA.
Collapse
Affiliation(s)
- Feiby L Nassan
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA.
| | - Cuicui Wang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Pantel S Vokonas
- VA Normative Aging Study, VA Boston Healthcare System, School of Medicine and School of Public Health, Boston University, Boston, MA 02215, USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA; Channing Division of Network Medicine; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02129, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
26
|
Jeong H, Lee C, Cheng C, Chou HC, Yang H, Bae H. Targeting of adipose tissue macrophages by bee venom phospholipase A2 attenuates high-fat diet-induced obesity. Int J Obes (Lond) 2021; 45:1656-1667. [PMID: 33947969 PMCID: PMC8310798 DOI: 10.1038/s41366-021-00823-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 02/24/2021] [Accepted: 04/08/2021] [Indexed: 12/26/2022]
Abstract
Background/objectives Adipose tissue macrophages (ATMs) exist in either the M1 or M2 form. The anti-inflammatory M2 ATMs accumulate in lean individuals, whereas the pro-inflammatory M1 ATMs accumulate in obese individuals. Bee venom phospholipase A2 (bvPLA2), a major component in honeybee (Apis mellifera) venom, exerts potent anti-inflammatory effects via interactions with regulatory T cells (Treg) and macrophages. This study investigated the effects of bvPLA2 on a high-fat diet (HFD)-induced obesity in mice. Subjects/methods For in vivo experiments, male C57BL/6, CD206-deficient, and Treg-depleted mice models were fed either a normal diet 41.86 kJ (ND, 10 kcal% fat) or high-fat diet 251.16 kJ (HFD, 60 kcal% fat). Each group was i.p. injected with PBS or bvPLA2 (0.5 mg/kg) every 3 days for 11 weeks. Body weight and food intake were measured weekly. Histological changes in the white adipose tissue (WAT), liver, and kidney as well as the immune phenotypes of the WAT were examined. Immune cells, cytokines, and lipid profiles were also evaluated. The direct effects of bvPLA2 on 3T3-L1 pre-adipocytes and bone marrow-derived macrophages were measured in vitro. Results bvPLA2 markedly decreased bodyweight in HFD-fed mice. bvPLA2 treatment also decreased lipid accumulation in the liver and reduced kidney inflammation in the mice. It was confirmed that bvPLA2 exerted immunomodulatory effects through the CD206 receptor. In addition, bvPLA2 decreased M1 ATM and alleviated the M1/M2 imbalance in vivo. However, bvPLA2 did not directly inhibit adipogenesis in the 3T3-L1 adipose cells in vitro. Conclusions bvPLA2 is a potential therapeutic strategy for the management of obesity by regulating adipose tissue macrophage homeostasis.
Collapse
Affiliation(s)
- Hyunju Jeong
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chanju Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chenyu Cheng
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hung Chun Chou
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - HyeJin Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Nassan FL, Kelly RS, Kosheleva A, Koutrakis P, Vokonas PS, Lasky-Su JA, Schwartz JD. Metabolomic signatures of the long-term exposure to air pollution and temperature. Environ Health 2021; 20:3. [PMID: 33413450 PMCID: PMC7788989 DOI: 10.1186/s12940-020-00683-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/01/2020] [Indexed: 05/19/2023]
Abstract
BACKGROUND Long-term exposures to air pollution has been reported to be associated with inflammation and oxidative stress. However, the underlying metabolic mechanisms remain poorly understood. OBJECTIVES We aimed to determine the changes in the blood metabolome and thus the metabolic pathways associated with long-term exposure to outdoor air pollution and ambient temperature. METHODS We quantified metabolites using mass-spectrometry based global untargeted metabolomic profiling of plasma samples among men from the Normative Aging Study (NAS). We estimated the association between long-term exposure to PM2.5, NO2, O3, and temperature (annual average of central site monitors) with metabolites and their associated metabolic pathways. We used multivariable linear mixed-effect regression models (LMEM) while simultaneously adjusting for the four exposures and potential confounding and correcting for multiple testing. As a reduction method for the intercorrelated metabolites (outcome), we further used an independent component analysis (ICA) and conducted LMEM with the same exposures. RESULTS Men (N = 456) provided 648 blood samples between 2000 and 2016 in which 1158 metabolites were quantified. On average, men were 75.0 years and had an average body mass index of 27.7 kg/m2. Almost all men (97%) were not current smokers. The adjusted analysis showed statistically significant associations with several metabolites (58 metabolites with PM2.5, 15 metabolites with NO2, and 6 metabolites with temperature) while no metabolites were associated with O3. One out of five ICA factors (factor 2) was significantly associated with PM2.5. We identified eight perturbed metabolic pathways with long-term exposure to PM2.5 and temperature: glycerophospholipid, sphingolipid, glutathione, beta-alanine, propanoate, and purine metabolism, biosynthesis of unsaturated fatty acids, and taurine and hypotaurine metabolism. These pathways are related to inflammation, oxidative stress, immunity, and nucleic acid damage and repair. CONCLUSIONS Using a global untargeted metabolomic approach, we identified several significant metabolites and metabolic pathways associated with long-term exposure to PM2.5, NO2 and temperature. This study is the largest metabolomics study of long-term air pollution, to date, the first study to report a metabolomic signature of long-term temperature exposure, and the first to use ICA in the analysis of both.
Collapse
Affiliation(s)
- Feiby L. Nassan
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Landmark Center, Room 414C, 401 Park Dr, Boston, MA 02215 USA
| | - Rachel S. Kelly
- Channing Division of Network Medicine; Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02129 USA
| | - Anna Kosheleva
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Landmark Center, Room 414C, 401 Park Dr, Boston, MA 02215 USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Landmark Center, Room 414C, 401 Park Dr, Boston, MA 02215 USA
| | - Pantel S. Vokonas
- VA Normative Aging Study, VA Boston Healthcare System, School of Medicine and School of Public Health, Boston University, Boston, MA 02215 USA
| | - Jessica A. Lasky-Su
- Channing Division of Network Medicine; Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02129 USA
| | - Joel D. Schwartz
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Landmark Center, Room 414C, 401 Park Dr, Boston, MA 02215 USA
- Channing Division of Network Medicine; Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02129 USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA 02115 USA
| |
Collapse
|
28
|
Prunonosa Cervera I, Gabriel BM, Aldiss P, Morton NM. The phospholipase A2 family's role in metabolic diseases: Focus on skeletal muscle. Physiol Rep 2021; 9:e14662. [PMID: 33433056 PMCID: PMC7802192 DOI: 10.14814/phy2.14662] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity and type 2 diabetes has increased substantially in recent years creating a global health burden. In obesity, skeletal muscle, the main tissue responsible for insulin-mediated glucose uptake, exhibits dysregulation of insulin signaling, glucose uptake, lipid metabolism, and mitochondrial function, thus, promoting type 2 diabetes. The phospholipase A2 (PLA2) enzyme family mediates lipid signaling and membrane remodeling and may play an important role in metabolic disorders such as obesity, diabetes, hyperlipidemia, and fatty liver disease. The PLA2 family consists of 16 members clustered in four groups. PLA2s hydrolyze the sn-2 ester bond of phospholipids generating free fatty acids and lysophospholipids. Differential tissue and subcellular PLA2 expression patterns and the abundance of distinct fatty acyl groups in the target phospholipid determine the impact of individual family members on metabolic functions and, potentially, diseases. Here, we update the current knowledge of the role of the PLA2 family in skeletal muscle, with a view to their potential for therapeutic targeting in metabolic diseases.
Collapse
Affiliation(s)
- Iris Prunonosa Cervera
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Brendan M. Gabriel
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
- Department of Physiology and PharmacologyIntegrative PhysiologyKarolinska InstituteStockholmSweden
- Aberdeen Cardiovascular & Diabetes CentreThe Rowett InstituteUniversity of AberdeenAberdeenUK
| | - Peter Aldiss
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Nicholas M. Morton
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
29
|
Tseng HH, Huang WR, Cheng CY, Chiu HC, Liao TL, Nielsen BL, Liu HJ. Aspirin and 5-Aminoimidazole-4-carboxamide Riboside Attenuate Bovine Ephemeral Fever Virus Replication by Inhibiting BEFV-Induced Autophagy. Front Immunol 2020; 11:556838. [PMID: 33329515 PMCID: PMC7732683 DOI: 10.3389/fimmu.2020.556838] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
Recent study in our laboratory has demonstrated that BEFV-induced autophagy via activation of the PI3K/Akt/NF-κB and Src/JNK pathways and suppression of the PI3K-AKt-mTORC1 pathway is beneficial for virus replication. In the current study, we found that both aspirin and 5-aminoimidazole-4-carboxamide-1-β-riboside (AICAR) siginificantly attenuated virus replication by inhibiting BEFV-induced autophagy via suppressing the BEFV-activated PI3K/Akt/NF-κB and Src/JNK pathways as well as inducing reversion of the BEFV-suppressed PI3K-Akt-mTORC1 pathway. AICAR reversed the BEFV-activated PI3K/Akt/NF-κB and Src/JNK pathways at the early to late stages of infection and induced reversion of the BEFV-suppressed PI3K-AKt-mTORC1 pathway at the late stage of infection. Our findings reveal that inhibition of BEFV-induced autophagy by AICAR is independent of AMPK. Furthermore, we found that AICAR transcriptionally downregulates the ATG related genes ULK1, Beclin 1, and LC3 and enhances Atg7 degradation by the proteasome pathway. Aspirin suppresses virus replication by inhibiting BEFV-induced autophagy. It directly suppressed the NF-κB pathway and reversed the BEFV-activated Src/JNK pathway at the early stage of infection and reversed the BEFV-suppressed PI3K/Akt/mTOR pathway at the late stage of infection. The current study provides mechanistic insights into the effects of aspirin and AICAR on BEFV replication through suppression of BEFV-induced autophagy.
Collapse
Affiliation(s)
- Hsu-Hung Tseng
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan.,Division of General Surgery, Taichung Hospital, Ministry of Health and Welfare, Taichung, Taiwan
| | - Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan.,The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Yuan Cheng
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Hung-Chuan Chiu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan.,The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Brent L Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan.,The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
30
|
Barberis E, Timo S, Amede E, Vanella VV, Puricelli C, Cappellano G, Raineri D, Cittone MG, Rizzi E, Pedrinelli AR, Vassia V, Casciaro FG, Priora S, Nerici I, Galbiati A, Hayden E, Falasca M, Vaschetto R, Sainaghi PP, Dianzani U, Rolla R, Chiocchetti A, Baldanzi G, Marengo E, Manfredi M. Large-Scale Plasma Analysis Revealed New Mechanisms and Molecules Associated with the Host Response to SARS-CoV-2. Int J Mol Sci 2020; 21:E8623. [PMID: 33207699 PMCID: PMC7696386 DOI: 10.3390/ijms21228623] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 01/08/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread to nearly every continent, registering over 1,250,000 deaths worldwide. The effects of SARS-CoV-2 on host targets remains largely limited, hampering our understanding of Coronavirus Disease 2019 (COVID-19) pathogenesis and the development of therapeutic strategies. The present study used a comprehensive untargeted metabolomic and lipidomic approach to capture the host response to SARS-CoV-2 infection. We found that several circulating lipids acted as potential biomarkers, such as phosphatidylcholine 14:0_22:6 (area under the curve (AUC) = 0.96), phosphatidylcholine 16:1_22:6 (AUC = 0.97), and phosphatidylethanolamine 18:1_20:4 (AUC = 0.94). Furthermore, triglycerides and free fatty acids, especially arachidonic acid (AUC = 0.99) and oleic acid (AUC = 0.98), were well correlated to the severity of the disease. An untargeted analysis of non-critical COVID-19 patients identified a strong alteration of lipids and a perturbation of phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, aminoacyl-tRNA degradation, arachidonic acid metabolism, and the tricarboxylic acid (TCA) cycle. The severity of the disease was characterized by the activation of gluconeogenesis and the metabolism of porphyrins, which play a crucial role in the progress of the infection. In addition, our study provided further evidence for considering phospholipase A2 (PLA2) activity as a potential key factor in the pathogenesis of COVID-19 and a possible therapeutic target. To date, the present study provides the largest untargeted metabolomics and lipidomics analysis of plasma from COVID-19 patients and control groups, identifying new mechanisms associated with the host response to COVID-19, potential plasma biomarkers, and therapeutic targets.
Collapse
Affiliation(s)
- Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Sara Timo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
| | - Elia Amede
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Virginia V. Vanella
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Chiara Puricelli
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Giuseppe Cappellano
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Davide Raineri
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Micol G. Cittone
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Eleonora Rizzi
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Anita R. Pedrinelli
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Veronica Vassia
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Francesco G. Casciaro
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Simona Priora
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Ilaria Nerici
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Alessandra Galbiati
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Eyal Hayden
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin University, Perth 6102, Australia;
| | - Rosanna Vaschetto
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
| | - Pier Paolo Sainaghi
- Internal and Emergency Medicine Departments, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.G.C.); (E.R.); (A.R.P.); (V.V.); (F.G.C.); (S.P.); (I.N.); (A.G.); (E.H.); (P.P.S.)
- Azienda Ospedaliero-Universitaria “Maggiore della Carità”, 28100 Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Roberta Rolla
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Annalisa Chiocchetti
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.P.); (U.D.); (R.R.)
| | - Gianluca Baldanzi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.B.); (E.A.); (V.V.V.); (R.V.); (G.B.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (G.C.); (D.R.); (A.C.); (E.M.)
| |
Collapse
|
31
|
Kaartinen MT, Arora M, Heinonen S, Rissanen A, Kaprio J, Pietiläinen KH. Transglutaminases and Obesity in Humans: Association of F13A1 to Adipocyte Hypertrophy and Adipose Tissue Immune Response. Int J Mol Sci 2020; 21:E8289. [PMID: 33167412 PMCID: PMC7663854 DOI: 10.3390/ijms21218289] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 12/11/2022] Open
Abstract
Transglutaminases TG2 and FXIII-A have recently been linked to adipose tissue biology and obesity, however, human studies for TG family members in adipocytes have not been conducted. In this study, we investigated the association of TGM family members to acquired weight gain in a rare set of monozygotic (MZ) twins discordant for body weight, i.e., heavy-lean twin pairs. We report that F13A1 is the only TGM family member showing significantly altered, higher expression in adipose tissue of the heavier twin. Our previous work linked adipocyte F13A1 to increased weight, body fat mass, adipocyte size, and pro-inflammatory pathways. Here, we explored further the link of F13A1 to adipocyte size in the MZ twins via a previously conducted TWA study that was further mined for genes that specifically associate to hypertrophic adipocytes. We report that differential expression of F13A1 (ΔHeavy-Lean) associated with 47 genes which were linked via gene enrichment analysis to immune response, leucocyte and neutrophil activation, as well as cytokine response and signaling. Our work brings further support to the role of F13A1 in the human adipose tissue pathology, suggesting a role in the cascade that links hypertrophic adipocytes with inflammation.
Collapse
Affiliation(s)
- Mari T. Kaartinen
- Faculty of Medicine (Experimental Medicine), McGill University, Montreal, QC H3A 0J7, Canada;
- Faculty of Dentistry (Biomedical Sciences), McGill University, Montreal, QC H3A 0J7, Canada
| | - Mansi Arora
- Faculty of Medicine (Experimental Medicine), McGill University, Montreal, QC H3A 0J7, Canada;
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.H.); (A.R.); (K.H.P.)
| | - Aila Rissanen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.H.); (A.R.); (K.H.P.)
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, 00100 Helsinki, Finland;
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.H.); (A.R.); (K.H.P.)
- Abdominal Center, Obesity Center, Endocrinology, University of Helsinki and Helsinki University Central Hospital, 00014 Helsinki, Finland
| |
Collapse
|
32
|
Guy AT, Ding F, Abe J, Inoue M, Hirabayashi Y, Ito Y, Kamiguchi H, Greimel P. Lysolipid Chain Length Switches Agonistic to Antagonistic G Protein-Coupled Receptor Modulation. ACS Chem Neurosci 2020; 11:3635-3645. [PMID: 33053304 DOI: 10.1021/acschemneuro.0c00521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Activation of lysolipid-sensitive G protein-coupled receptors (GPCR) depends not only on lysolipid class but also on the length and degree of saturation of their respective hydrophobic tails. Positive regulation of these signaling networks caused by the lipid chain length specificity of upstream phospholipases is firmly established. Nonagonistic lysolipid homologues, featuring incompatible lipid tails, have been suggested to indirectly modulate GPCR signaling by delaying agonist catabolism. Nonetheless, recent results seem inconsistent with this hypothesis. Utilizing a simplified lysolipid-GPCR signaling assay based on the established lysophosphatidylglucoside-GPR55 signaling axis in primary sensory neurons, we demonstrate that short-chain ligand homologues directly modulate receptor activation via a potent competitive antagonistic activity. Considering the well-documented tissue-specific concentration of lysolipid homologues, we propose that endogenous lysolipids with insufficient chain length for stable receptor activation exert an antagonistic activity, effectively representing a negative control mechanism for GPCR-associated lysolipid signaling.
Collapse
Affiliation(s)
- Adam T. Guy
- RIKEN Center for Brain Science, RIKEN, Wako City, Saitama 351-0198, Japan
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Junpei Abe
- Graduate School of Science, Osaka University, Toyonaka, 560-0043, Japan
| | - Mariko Inoue
- RIKEN Center for Brain Science, RIKEN, Wako City, Saitama 351-0198, Japan
| | - Yoshio Hirabayashi
- RIKEN Cluster for Pioneering Research, RIKEN, Wako City, Saitama 351-0198, Japan
| | - Yukishige Ito
- RIKEN Cluster for Pioneering Research, RIKEN, Wako City, Saitama 351-0198, Japan
| | - Hiroyuki Kamiguchi
- RIKEN Center for Brain Science, RIKEN, Wako City, Saitama 351-0198, Japan
| | - Peter Greimel
- RIKEN Center for Brain Science, RIKEN, Wako City, Saitama 351-0198, Japan
| |
Collapse
|
33
|
Therapeutic Potential of Porcine Liver Decomposition Product: New Insights and Perspectives for Microglia-Mediated Neuroinflammation in Neurodegenerative Diseases. Biomedicines 2020; 8:biomedicines8110446. [PMID: 33105637 PMCID: PMC7690401 DOI: 10.3390/biomedicines8110446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/23/2022] Open
Abstract
It is widely accepted that microglia-mediated inflammation contributes to the progression of neurodegenerative diseases; however, the precise mechanisms through which these cells contribute remain to be elucidated. Microglia, as the primary immune effector cells of the brain, play key roles in maintaining central nervous system (CNS) homeostasis. Microglia are located throughout the brain and spinal cord and may account for up to 15% of all cells in the brain. Activated microglia express pro-inflammatory cytokines that act on the surrounding brain and spinal cord. Microglia may also play a detrimental effect on nerve cells when they gain a chronic inflammatory function and promote neuropathologies. A key feature of microglia is its rapid morphological change upon activation, characterized by the retraction of numerous fine processes and the gradual acquisition of amoeba-like shapes. These morphological changes are also accompanied by the expression and secretion of inflammatory molecules, including cytokines, chemokines, and lipid mediators that promote systemic inflammation during neurodegeneration. This may be considered a protective response intended to limit further injury and initiate repair processes. We previously reported that porcine liver decomposition product (PLDP) induces a significant increase in the Hasegawa’s Dementia Scale-Revised (HDS-R) score and the Wechsler Memory Scale (WMS) in a randomized, double-blind, placebo-controlled study in healthy humans. In addition, the oral administration of porcine liver decomposition product enhanced visual memory and delayed recall in healthy adults. We believe that PLDP is a functional food that aids cognitive function. In this review, we provide a critical assessment of recent reports of lysophospholipids derived from PLDP, a rich source of phospholipids. We also highlight some recent findings regarding bidirectional interactions between lysophospholipids and microglia and age-related neurodegenerative diseases such as dementia and Alzheimer’s disease.
Collapse
|
34
|
Bagheri M, Tiwari HK, Murillo AL, Al-Tobasei R, Arnett DK, Kind T, Barupal DK, Fan S, Fiehn O, O'connell J, Montasser M, Aslibekyan S, Irvin MR. A lipidome-wide association study of the lipoprotein insulin resistance index. Lipids Health Dis 2020; 19:153. [PMID: 32586392 PMCID: PMC7318473 DOI: 10.1186/s12944-020-01321-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 06/10/2020] [Indexed: 12/30/2022] Open
Abstract
Background The lipoprotein insulin resistance (LPIR) score was shown to predict insulin resistance (IR) and type 2 diabetes (T2D) in healthy adults. However, the molecular basis underlying the LPIR utility for classification remains unclear. Objective To identify small molecule lipids associated with variation in the LPIR score, a weighted index of lipoproteins measured by nuclear magnetic resonance, in the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN) study (n = 980). Methods Linear mixed effects models were used to test the association between the LPIR score and 413 lipid species and their principal component analysis-derived groups. Significant associations were tested for replication with homeostatic model assessment-IR (HOMA-IR), a phenotype correlated with the LPIR score (r = 0.48, p < 0.001), in the Heredity and Phenotype Intervention (HAPI) Heart Study (n = 590). Results In GOLDN, 319 lipids were associated with the LPIR score (false discovery rate-adjusted p-values ranging from 4.59 × 10− 161 to 49.50 × 10− 3). Factors 1 (triglycerides and diglycerides/storage lipids) and 3 (mixed lipids) were positively (β = 0.025, p = 4.52 × 10− 71 and β = 0.021, p = 5.84 × 10− 41, respectively) and factor 2 (phospholipids/non-storage lipids) was inversely (β = − 0.013, p = 2.28 × 10− 18) associated with the LPIR score. These findings were replicated for HOMA-IR in the HAPI Heart Study (β = 0.10, p = 1.21 × 10− 02 for storage, β = − 0.13, p = 3.14 × 10− 04 for non-storage, and β = 0.19, p = 8.40 × 10− 07 for mixed lipids). Conclusions Non-storage lipidomics species show a significant inverse association with the LPIR metabolic dysfunction score and present a promising focus for future therapeutic and prevention studies.
Collapse
Affiliation(s)
- Minoo Bagheri
- Department of Epidemiology, University of Alabama at Birmingham, 1665 University Blvd, Birmingham, AL, 35294, USA.,Department of Cardiovascular Medicine, Vanderbilt University Medical center, Nashville, TN, USA
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anarina L Murillo
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rafet Al-Tobasei
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donna K Arnett
- Dean's Office, School of Public Health, University of Kentucky, Lexington, KY, USA
| | - Tobias Kind
- West coast metabolomics center, Davis, CA, USA
| | | | - Sili Fan
- West coast metabolomics center, Davis, CA, USA
| | | | - Jeff O'connell
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - May Montasser
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham, 1665 University Blvd, Birmingham, AL, 35294, USA.
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, 1665 University Blvd, Birmingham, AL, 35294, USA
| |
Collapse
|
35
|
Signini ÉF, Nieman DC, Silva CD, Sakaguchi CA, Catai AM. Oxylipin Response to Acute and Chronic Exercise: A Systematic Review. Metabolites 2020; 10:E264. [PMID: 32630487 PMCID: PMC7345129 DOI: 10.3390/metabo10060264] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/08/2023] Open
Abstract
Oxylipins are oxidized compounds of polyunsaturated fatty acids that play important roles in the body. Recently, metabololipidomic-based studies using advanced mass spectrometry have measured the oxylipins generated during acute and chronic physical exercise and described the related physiological effects. The objective of this systematic review was to provide a panel of the primary exercise-related oxylipins and their respective functions in healthy individuals. Searches were performed in five databases (Cochrane, PubMed, Science Direct, Scopus and Web of Science) using combinations of the Medical Subject Headings (MeSH) terms: "Humans", "Exercise", "Physical Activity", "Sports", "Oxylipins", and "Lipid Mediators". An adapted scoring system created in a previous study from our group was used to rate the quality of the studies. Nine studies were included after examining 1749 documents. Seven studies focused on the acute effect of physical exercise while two studies determined the effects of exercise training on the oxylipin profile. Numerous oxylipins are mobilized during intensive and prolonged exercise, with most related to the inflammatory process, immune function, tissue repair, cardiovascular and renal functions, and oxidative stress.
Collapse
Affiliation(s)
- Étore F. Signini
- Physical Therapy Department, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil; (C.D.S.); (C.A.S.); (A.M.C.)
| | - David C. Nieman
- North Carolina Research Campus, Appalachian State University, Kannapolis, NC 28081, USA;
| | - Claudio D. Silva
- Physical Therapy Department, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil; (C.D.S.); (C.A.S.); (A.M.C.)
| | - Camila A. Sakaguchi
- Physical Therapy Department, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil; (C.D.S.); (C.A.S.); (A.M.C.)
| | - Aparecida M. Catai
- Physical Therapy Department, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil; (C.D.S.); (C.A.S.); (A.M.C.)
| |
Collapse
|
36
|
NS398, a cyclooxygenase-2 inhibitor, reverses memory performance disrupted by imipramine in C57Bl/6J mice. Brain Res 2020; 1734:146741. [PMID: 32088181 DOI: 10.1016/j.brainres.2020.146741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 11/21/2022]
Abstract
Imipramine has been widely used as an antidepressant in the clinic over the years. Unfortunately, it produces a detrimental effect on memory. At the same time, COX-2 inhibitors engagement in the mechanisms of memory formation, and synapse plastic changes has been well documented. Our previous studies have demonstrated the contribution of cyclooxygenase-2 (COX-2) inhibition to the parameters of the mGluR5 pathway in memory formation. Because chronic administration of imipramine has been shown to affect mGluR5, the purpose of this study was to verify the hypothesis of COX-2 pathway engagement in disrupting effects of imipramine. Imipramine is currently used as a reference compound, and therefore it seems important to decipher and understand mood-related pathways, as well as cognitive changes activated during its use. This study covers the examination of spatial, and motor parameters. To this end, C57Bl/6J mice received imipramine, and NS398 (a COX-2 inhibitor) alone, or in combination for 7 or 14 days. We performed the modified Barnes maze (MBM), modified rotarod (MR) tests, and electrophysiological studies. The harmful effect of imipramine on MBM learning was improved by NS398 use. The same modulatory role of the COX-2 inhibitor in procedural learning in the MR test was found. In conclusion, our data show the involvement of the COX-2 pathway in changes in the long-term memory, and procedural memory of C57Bl/6J mice after chronic imipramine treatment.
Collapse
|
37
|
Filkin SY, Lipkin AV, Fedorov AN. Phospholipase Superfamily: Structure, Functions, and Biotechnological Applications. BIOCHEMISTRY (MOSCOW) 2020; 85:S177-S195. [DOI: 10.1134/s0006297920140096] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
38
|
Liu H, Liu W, He X, Chen X, Yang J, Wang Y, Li Y, Ren J, Xu W, Zhao Y. Characterization of a cell density-dependent sRNA, Qrr, and its roles in the regulation of the quorum sensing and metabolism in Vibrio alginolyticus. Appl Microbiol Biotechnol 2020; 104:1707-1720. [PMID: 31907574 DOI: 10.1007/s00253-019-10278-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/08/2019] [Accepted: 11/23/2019] [Indexed: 12/25/2022]
Abstract
Vibrio alginolyticus is an important fish pathogen causing pandemic diseases in marine animals. Small noncoding RNAs (sRNAs) are important posttranscriptional modulators of gene expression and involved in the pathogenesis of bacterial pathogens. Thus far, no cell density-dependent sRNA has been reported in V. alginolyticus. In this study, a cell density-dependent sRNA, Qrr, predicted based on the previous RNA-Seq analysis of V. alginolyticus cultured at low cell density (LCD) and high cell density (HCD), was characterized. The Qrr mutant showed significantly impaired growth and decreased swimming and swarming ability, and increased biofilm formation, extracellular polysaccharide content, serine protease production, and LD50 values during zebrafish infection in contrast to the wild-type strain. Qrr modulates the master regulators LuxR and AphA in quorum sensing (QS) pathways possibly at the posttranscriptional level by base pairing with the 5'-untranslated regions (5'-UTRs). Meanwhile, both LuxR and AphA could directly bind to the promoter of qrr to activate or repress its transcription, respectively. Moreover, our unbiased metabolic approaches revealed that Qrr modulates a large quantity of metabolic and lipidomic pathways, including amino acids, purine and pyrimidine derivatives, tricarboxylic acid cycle (TCA cycle) intermediates, and lipids. Collectively, this work contributes to a systematic understanding of regulatory roles of the cell density-dependent sRNA, Qrr, in V. alginolyticus.
Collapse
Affiliation(s)
- Huan Liu
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Wang Liu
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Xiaoxian He
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Xuefeng Chen
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Jinfang Yang
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Yi Wang
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Yue Li
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Jiamin Ren
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Wensheng Xu
- Laboratory of Agricultural Product Detection and Control of Spoilage Organisms and Pesticide Residue, Faculty of Food Science and Engineering, Beijing University of Agriculture, Beijing, 102206, China.
| | - Yanni Zhao
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| |
Collapse
|
39
|
Yan Q, Liew Z, Uppal K, Cui X, Ling C, Heck JE, von Ehrenstein OS, Wu J, Walker DI, Jones DP, Ritz B. Maternal serum metabolome and traffic-related air pollution exposure in pregnancy. ENVIRONMENT INTERNATIONAL 2019; 130:104872. [PMID: 31228787 PMCID: PMC7017857 DOI: 10.1016/j.envint.2019.05.066] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 05/19/2023]
Abstract
BACKGROUND Maternal exposure to traffic-related air pollution during pregnancy has been shown to increase the risk of adverse birth outcomes and neurodevelopmental disorders. By utilizing high-resolution metabolomics (HRM), we investigated perturbations of the maternal serum metabolome in response to traffic-related air pollution to identify biological mechanisms. METHODS We retrieved stored mid-pregnancy serum samples from 160 mothers who lived in the Central Valley of California known for high air particulate levels. We estimated prenatal traffic-related air pollution exposure (carbon monoxide, nitric oxides, and particulate matter <2.5 μm) during first-trimester using the California Line Source Dispersion Model, version 4 (CALINE4) based on residential addresses recorded at birth. We used liquid chromatography-high resolution mass spectrometry to obtain untargeted metabolic profiles and partial least squares discriminant analysis (PLS-DA) to select metabolic features associated with air pollution exposure. Pathway analyses were employed to identify biologic pathways related to air pollution exposure. As potential confounders we included maternal age, maternal race/ethnicity, and maternal education. RESULTS In total we extracted 4038 and 4957 metabolic features from maternal serum samples in hydrophilic interaction (HILIC) chromatography (positive ion mode) and C18 (negative ion mode) columns, respectively. After controlling for confounding factors, PLS-DA (Variable Importance in Projection (VIP) ≥2) yielded 181 and 251 metabolic features (HILIC and C18, respectively) that discriminated between the high (n = 98) and low exposed (n = 62). Pathway enrichment analysis for discriminatory features associated with air pollution indicated that in maternal serum oxidative stress and inflammation related pathways were altered, including linoleate, leukotriene, and prostaglandin pathways. CONCLUSION The metabolomic features and pathways we found to be associated with air pollution exposure suggest that maternal exposure during pregnancy induces oxidative stress and inflammation pathways previously implicated in pregnancy complications and adverse outcomes.
Collapse
Affiliation(s)
- Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Zeyan Liew
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA; Yale Center for Perinatal, Pediatric, and Environmental Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Xin Cui
- Perinatal Epidemiology and Health Outcomes Research Unit, Division of Neonatology, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA, USA; California Perinatal Quality Care Collaborative, Palo Alto, CA, USA
| | - Chenxiao Ling
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Julia E Heck
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | | | - Jun Wu
- Program in Public Health, UCI Susan and Henry Samueli College of Health Sciences, Irvine, CA, USA
| | - Douglas I Walker
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Department of Medicine, Emory University, Atlanta, GA, USA
| | - Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Neurology, UCLA School of Medicine, CA, USA.
| |
Collapse
|
40
|
Prasher P, Mudila H, Sharma M, Khati B. Developmental perspectives of the drugs targeting enzyme-instigated inflammation: a mini review. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02315-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
41
|
Differential Gene Expression in Peripheral White Blood Cells with Permissive Underfeeding and Standard Feeding in Critically Ill Patients: A Descriptive Sub-study of the PermiT Randomized Controlled Trial. Sci Rep 2018; 8:17984. [PMID: 30573851 PMCID: PMC6301949 DOI: 10.1038/s41598-018-36007-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023] Open
Abstract
The effect of short-term caloric restriction on gene expression in critically ill patients has not been studied. In this sub-study of the PermiT trial (Permissive Underfeeding or Standard Enteral Feeding in Critically Ill Adults Trial- ISRCTN68144998), we examined gene expression patterns in peripheral white blood cells (buffy coat) associated with moderate caloric restriction (permissive underfeeding) in critically ill patients compared to standard feeding. Blood samples collected on study day 1 and 14 were subjected to total RNA extraction and gene expression using microarray analysis. We enrolled 50 patients, 25 in each group. Among 1751 tested genes, 332 genes in 12 pathways were found to be significantly upregulated or downregulated between study day 1 and 14 (global p value for the pathway ≤ 0.05). Using the heatmap, the differential expression of genes from day 1 to 14 in the permissive underfeeding group was compared to the standard feeding group. We further compared gene expression signal intensity in permissive underfeeding compared standard feeding by constructing univariate and multivariate linear regression models on individual patient data. We found differential expression of several genes with permissive underfeeding, most notably those related to metabolism, autophagy and other cellular functions, indicating that moderate differences in caloric intake trigger different cellular pathways.
Collapse
|
42
|
Kuefner MS, Deng X, Stephenson EJ, Pham K, Park EA. Secretory phospholipase A
2
group IIA enhances the metabolic rate and increases glucose utilization in response to thyroid hormone. FASEB J 2018; 33:738-749. [DOI: 10.1096/fj.201800711r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Michael S. Kuefner
- Department of PharmacologyCollege of MedicineUniversity of Tennessee Health Science Center Memphis Tennessee USA
- Department of Veterans Affairs Medical Center Memphis Tennessee USA
| | - Xiong Deng
- Department of PharmacologyCollege of MedicineUniversity of Tennessee Health Science Center Memphis Tennessee USA
- Department of Veterans Affairs Medical Center Memphis Tennessee USA
| | - Erin J. Stephenson
- Department of PediatricsCollege of MedicineUniversity of Tennessee Health Science Center Memphis Tennessee USA
- Children's Foundation Research InstituteLeBonheur Children's Hospital Memphis Tennessee USA
| | - Kevin Pham
- Department of PharmacologyCollege of MedicineUniversity of Tennessee Health Science Center Memphis Tennessee USA
- Department of Veterans Affairs Medical Center Memphis Tennessee USA
| | - Edwards A. Park
- Department of PharmacologyCollege of MedicineUniversity of Tennessee Health Science Center Memphis Tennessee USA
- Department of Veterans Affairs Medical Center Memphis Tennessee USA
| |
Collapse
|