1
|
Sahoo PK, Ravi A, Liu B, Yu J, Natarajan SK. Palmitoleate protects against lipopolysaccharide-induced inflammation and inflammasome activity. J Lipid Res 2024; 65:100672. [PMID: 39396700 PMCID: PMC11585775 DOI: 10.1016/j.jlr.2024.100672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 09/16/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024] Open
Abstract
Inflammation is part of natural immune defense mechanism against any form of infection or injury. However, prolonged inflammation could perturb cell homeostasis and contribute to the development of metabolic and inflammatory diseases, including maternal obesity, diabetes, cardiovascular diseases, and metabolic dysfunction-associated steatotic liver diseases (MASLD). Polyunsaturated fatty acids have been shown to mitigate inflammatory response by generating specialized proresolving lipid mediators, which take part in resolution of inflammation. Similarly here, we show that palmitoleate, an omega-7 monounsaturated fatty acid exerts anti-inflammatory properties in response to lipopolysaccharide (LPS)-mediated inflammation. Exposure of bone marrow-derived macrophages (BMDMs) to LPS or TNFα induces robust increase in the expression of proinflammatory cytokines and supplementation of palmitoleate inhibited LPS-mediated upregulation of proinflammatory cytokines. We also observed that palmitoleate was able to block LPS + ATP-induced inflammasome activation mediated cleavage of procaspase 1 and prointerleukin-1β. Further, treatment of palmitoleate protects against LPS-induced inflammation in human THP-1-derived macrophages and trophoblasts. Coexposure of LPS and palmitate (saturated free fatty acid) induces inflammasome and cell death in BMDMs, however, treatment of palmitoleate blocked LPS and palmitate-induced cell death in BMDMs. Further, LPS and palmitate together results in the activation of mitogen-activated protein kinases and pretreatment of palmitoleate inhibited the activation of mitogen-activated protein kinases and nuclear translocation of nuclear factor kappa B in BMDMs. In conclusion, palmitoleate shows anti-inflammatory properties against LPS-induced inflammation and LPS + palmitate/ATP-induced inflammasome activity and cell death.
Collapse
Affiliation(s)
- Prakash Kumar Sahoo
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Aiswariya Ravi
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Baolong Liu
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yang ling, Shaanxi, China
| | - Jiujiu Yu
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Sathish Kumar Natarajan
- Department of Nutrition & Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; College of Allied Health Professions Medical Nutrition Education, University of Nebraska Medical Center, Omaha, NE, USA; Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
2
|
Hameete BC, Plösch T, Hogenkamp A, Groenink L. A systematic review and risk of bias analysis of in vitro studies on trophoblast response to immunological triggers. Placenta 2024:S0143-4004(24)00682-9. [PMID: 39551667 DOI: 10.1016/j.placenta.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/02/2024] [Accepted: 10/20/2024] [Indexed: 11/19/2024]
Abstract
An increasing amount of evidence suggests that immune responses may affect trophoblast functioning, which in turn may play a role in gestational disorders and fetal development. This systematic review offers the first summary of in vitro studies on the trophoblast response to immunological triggers, in conjunction with a risk of bias analysis. A search in Pubmed and Embase yielded 110 relevant studies. Primary trophoblasts were the most commonly used cell type, but trophoblast subtypes were not always defined. Similarly, the exact natures of trophoblast cell lines were sometimes unclear. Cytokines and Toll-like receptor agonists were often used as interventions, but most studies focused on a select few substances such as tumor necrosis factor-α and lipopolysaccharide. In regard to the outcome parameters, some important trophoblast functions, such as hormone production and barrier formation were underrepresented. Whether or not risk of bias was high varied strongly between types of bias. Risk of selection bias, for example, was usually low. However, none of the included studies mentioned blinding or plate randomization. Only a select few studies mentioned passage numbers, use of vehicle control or conflict of interest. In conclusion, better characterization of trophoblast subtypes and a broader range of studied interventions and outcome parameters would contribute to a more complete understanding of trophoblast responses to immune stimuli. Additionally, researchers are encouraged to replicate experiments and pay close attention when setting up and writing down methodologies, in order to improve the reproducibility and translatability of their work.
Collapse
Affiliation(s)
- Bart Christiaan Hameete
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Perinatal Neurobiology, Department of Human Medicine, School of Medicine and Health Sciences Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Astrid Hogenkamp
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Lucianne Groenink
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands.
| |
Collapse
|
3
|
Bidne KL, Zemski Berry K, Dillon M, Jansson T, Powell TL. Maternal Docosahexaenoic Acid Supplementation Alters Maternal and Fetal Docosahexaenoic Acid Status and Placenta Phospholipids in Pregnancies Complicated by High Body Mass Index. Nutrients 2024; 16:2934. [PMID: 39275250 PMCID: PMC11397315 DOI: 10.3390/nu16172934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
INTRODUCTION An optimal fetal supply of docosahexaenoic acid (DHA) is critical for normal brain development. The relationship between maternal DHA intake and DHA delivery to the fetus is complex and is dependent on placental handling of DHA. Little data exist on placental DHA levels in pregnancies supplemented with the recommended dose of 200 mg/d. Our objective was to determine how prenatal DHA at the recommended 200 mg/d impacts maternal, placental, and fetal DHA status in both normal-weight and high-BMI women compared to women taking no supplements. METHODS Maternal blood, placenta, and cord blood were collected from 30 healthy pregnant women (BMI 18.9-43.26 kg/m2) giving birth at term. Red blood cells (RBCs) and villous tissue were isolated, and lipids were extracted to determine DHA content by LC-MS/MS. Data were analyzed by supplement group (0 vs. 200 mg/d) and maternal BMI (normal weight or high BMI) using two-way ANOVA. We measured maternal choline levels in maternal and cord plasma samples. RESULTS Supplementation with 200 mg/d DHA significantly increased (p < 0.05) maternal and cord RBC DHA content only in pregnancies complicated by high BMI. We did not find any impact of choline levels on maternal or cord RBC phospholipids. There were no significant differences in total placental DHA content by supplementation or maternal BMI (p > 0.05). Placental levels of phosphatidylinositol (PI) and phosphatidic acid containing DHA species were higher (p < 0.05) in high-BMI women without DHA supplementation compared to both normal-BMI and high-BMI women taking DHA supplements. CONCLUSION Maternal DHA supplementation at recommended doses cord increased RBC DHA content only in pregnancies complicated by higher BMI. Surprisingly, we found that obesity was related to an increase in placental PI and phosphatidic acid species, which was ameliorated by DHA supplementation. Phosphatidic acid activates placental mTOR, which regulates amino acid transport and may explain previous findings of the impact of DHA on placental function. Current recommendations for DHA supplementation may not be achieving the goal of improving fetal DHA levels in normal-weight women.
Collapse
Affiliation(s)
- Katie L Bidne
- Departments of Obstetrics and Gynecology, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Karin Zemski Berry
- Departments of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Mairead Dillon
- Departments of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Thomas Jansson
- Departments of Obstetrics and Gynecology, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Theresa L Powell
- Departments of Obstetrics and Gynecology, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
- Departments of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Lu C, Zhou ZW, Jiang Y, Li J, He JB, Zhang C, Chen AF, Tao X, Peng C, Xie HH. Sodium dichloroacetate improves migration ability by suppressing LPS-induced inflammation in HTR-8/SVneo cells via the TLR4/NF-κB pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:16-23. [PMID: 38164486 PMCID: PMC10722477 DOI: 10.22038/ijbms.2023.68252.14902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 07/15/2023] [Indexed: 01/03/2024]
Abstract
Objectives Inadequate cytotrophoblast migration and invasion are speculated to result in preeclampsia, which is a pro-inflammatory condition. Sodium dichloroacetate (DCA) exerts anti-inflammatory actions. Thus,we sought to investigate the effect of DCA on the migration function of the lipopolysaccharide (LPS)-stimulated human-trophoblast-derived cell line (HTR-8/SVneo). Materials and Methods HTR-8/SVneo cells were treated with LPS to suppress cell migration. Cell migration was examined by both scratch wound healing assay and transwell migration assay. Western blotting was used to analyze the expression levels of toll-like receptor-4 (TLR4), nuclear factor-κB (NF-κB), TNF-α, IL-1β, and IL-6 in the cells. Results DCA reversed LPS-induced inhibition of migration in HTR-8/SVneo cells. Furthermore, DCA significantly suppressed LPS-induced activation of TLR4, phosphorylation of NF-κB (p65), translocation of p65 into the nucleus, and the production of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6). Treatment with inhibitors of TLR4 signal transduction (CLI095 or MD2-TLR-4-IN-1) reduced LPS-induced overexpression of pro-inflammatory cytokines, and a synergistic effect was found between TLR4 inhibitors and DCA in HTR-8/SVneo cells. Conclusion DCA improved trophoblast cell migration function by suppressing LPS-induced inflammation, at least in part, via the TLR4/NF-κB signaling pathway. This result indicates that DCA might be a potential therapeutic candidate for human pregnancy-related complications associated with trophoblast disorder.
Collapse
Affiliation(s)
- Cheng Lu
- School of Public Health and Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- These authors contributed eqully to this work
| | - Zhen-Wei Zhou
- School of Public Health and Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- These authors contributed eqully to this work
| | - Yu Jiang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
- These authors contributed eqully to this work
| | - Jianzhong Li
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Jia-Bei He
- School of Public Health and Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chuan Zhang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xia Tao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Cheng Peng
- School of Public Health and Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - He-Hui Xie
- School of Public Health and Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
5
|
Zoanni B, Aiello G, Negre-Salvayre A, Aldini G, Carini M, D'Amato A. Lipidome Investigation of Carnosine Effect on Nude Mice Skin to Prevent UV-A Damage. Int J Mol Sci 2023; 24:10009. [PMID: 37373157 DOI: 10.3390/ijms241210009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The lipid profile of skin is fundamental in the maintenance of the protective barrier against the external environment. Signaling and constitutive lipids of this large organ are involved in inflammation, metabolism, aging, and wound healing, such as phospholipids, triglycerides, FFA, and sphingomyelin. Skin exposure to ultraviolet (UV) radiation results in a photoaging process that is an accelerated form of aging. UV-A radiation deeply penetrates the dermis and promotes damage to DNA, lipids, and proteins by increasing the generation of reactive oxygen species (ROS). Carnosine, an endogenous β-alanyl-L-histidine dipeptide, demonstrated antioxidant properties that prevent photoaging and modification of skin protein profiling, making carnosine a compelling ingredient to consider for use in dermatology. The aim of this research was to investigate the modification of skin lipidome after UV-A treatment in presence or not of topic administration of carnosine. Quantitative analyses based on high-resolution mass spectrometry of nude mice skin-extracted lipids resulted in several modifications of barrier composition after UV-A radiation, with or without carnosine treatment. In total, 328 out of 683 molecules showed significant alteration-262 after UV-A radiation and 126 after UV-A and carnosine treatment versus controls. Importantly, the increased oxidized TGs after UV-A radiation, responsible of dermis photoaging, were completely reverted by carnosine application to prevent the UV-A damage. Network analyses also showed that the production of ROS and the calcium and TNF signaling were modulated by UV-A and carnosine. In conclusion, lipidome analyses attested the carnosine activity to prevent the UV-A damage, reducing the lipid oxidation, the inflammation, and the dysregulation of lipid skin barrier.
Collapse
Affiliation(s)
- Beatrice Zoanni
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Gilda Aiello
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
- Department of Human Science and Quality of Life Promotion, Telematic University San Raffaele, 00166 Rome, Italy
| | - Anne Negre-Salvayre
- Faculty of Medicine, Department of Biochemistry, INSERM U1297 and University of Toulouse, 31432 Toulouse, France
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Marina Carini
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Alfonsina D'Amato
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
6
|
Motomura K, Hara M, Ito I, Morita H, Matsumoto K. Roles of human trophoblasts' pattern recognition receptors in host defense and pregnancy complications. J Reprod Immunol 2023; 156:103811. [PMID: 36669386 DOI: 10.1016/j.jri.2023.103811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/24/2022] [Accepted: 01/12/2023] [Indexed: 01/14/2023]
Abstract
The immune system in pregnancy is able to protect pregnant mothers and fetuses from pathogenic microorganisms even while permitting the mother to tolerate the semi-allogenic fetus. Trophoblasts, which are fetal-derived placental cells, play a central role on both sides of this duality at the maternal-fetal interface. In brief, the trophoblasts express pattern recognition receptors (PRRs) and are involved in the local innate immune response in the placenta. That response eliminates pathogenic microbes but also causes tissue damage. In this review, we summarize the research findings to date regarding the roles of those human trophoblast PRRs. Multiple types of PRRs (Toll-like receptors, Nod-like receptors, and RIG-I-like receptors) are expressed in the placenta and on trophoblasts. Trophoblasts' PRRs participate in protecting the fetus against viruses, bacteria, and parasites by triggering production of proinflammatory cytokines and chemokines in the placenta. On the negative side, PRR signaling in trophoblasts can also initiate inflammation and trophoblast cell death, which can lead to placental inflammation-associated pregnancy complications such as preeclampsia, anti-phospholipid antibody syndrome, and miscarriage. Further elucidation of these dual roles of trophoblasts' PRRs may shed light on the mechanisms by which fetuses are protected against congenital infections and also give us a better understanding of the etiologies of pregnancy complications, which can help us prevent/reduce adverse prenatal/neonatal outcomes.
Collapse
Affiliation(s)
- Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan.
| | - Mariko Hara
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Department of Otorhinolaryngology, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Ikuyo Ito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Department of Pediatrics, School of Medicine, Yokohama City University, Kanagawa 236-0004, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan.
| |
Collapse
|
7
|
Udompornpitak K, Charoensappakit A, Sae-Khow K, Bhunyakarnjanarat T, Dang CP, Saisorn W, Visitchanakun P, Phuengmaung P, Palaga T, Ritprajak P, Tungsanga S, Leelahavanichkul A. Obesity Exacerbates Lupus Activity in Fc Gamma Receptor IIb Deficient Lupus Mice Partly through Saturated Fatty Acid-Induced Gut Barrier Defect and Systemic Inflammation. J Innate Immun 2022; 15:240-261. [PMID: 36219976 PMCID: PMC10643905 DOI: 10.1159/000526206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/21/2022] [Indexed: 11/19/2022] Open
Abstract
The prevalence of obesity is increasing, and the coexistence of obesity and systemic lupus erythematosus (lupus) is possible. A high-fat diet (HFD) was orally administered for 6 months in female 8-week-old Fc gamma receptor IIb deficient (FcgRIIb-/-) lupus or age and gender-matched wild-type (WT) mice. Lupus nephritis (anti-dsDNA, proteinuria, and increased creatinine), gut barrier defect (fluorescein isothiocyanate dextran), serum lipopolysaccharide (LPS), serum interleukin (IL)-6, liver injury (alanine transaminase), organ fibrosis (liver and kidney pathology), spleen apoptosis (activated caspase 3), and aorta thickness (but not weight gain and lipid profiles) were more prominent in HFD-administered FcgRIIb-/- mice than the obese WT, without injury in regular diet-administered mice (both FcgRIIb-/- and WT). In parallel, combined palmitic acid (PA; a saturated fatty acid) with LPS (PA + LPS) induced higher tumor necrotic factor-α, IL-6, and IL-10 in the supernatant, inflammatory genes (inducible nitric oxide synthase and IL-1β), reactive oxygen species (dihydroethidium), and glycolysis with reduced mitochondrial activity (extracellular flux analysis) when compared with the activation by each molecule alone in both FcgRIIb-/- and WT macrophages. However, the alterations of these parameters were more prominent in PA + LPS-administered FcgRIIb-/- than in the WT cells. In conclusion, obesity accelerated inflammation in FcgRIIb-/- mice, partly due to the more potent responses from the loss of inhibitory FcgRIIb against PA + LPS with obesity-induced gut barrier defect.
Collapse
Affiliation(s)
- Kanyarat Udompornpitak
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Awirut Charoensappakit
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kritsanawan Sae-Khow
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Cong Phi Dang
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wilasinee Saisorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Peerapat Visitchanakun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Patcharee Ritprajak
- Research Unit in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Somkanya Tungsanga
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of General Internal Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
8
|
Zhou R, He M, Fan J, Li R, Zuo Y, Li B, Gao G, Sun T. The role of hypothalamic endoplasmic reticulum stress in schizophrenia and antipsychotic-induced weight gain: A narrative review. Front Neurosci 2022; 16:947295. [PMID: 36188456 PMCID: PMC9523121 DOI: 10.3389/fnins.2022.947295] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022] Open
Abstract
Schizophrenia (SCZ) is a serious mental illness that affects 1% of people worldwide. SCZ is associated with a higher risk of developing metabolic disorders such as obesity. Antipsychotics are the main treatment for SCZ, but their side effects include significant weight gain/obesity. Despite extensive research, the underlying mechanisms by which SCZ and antipsychotic treatment induce weight gain/obesity remain unclear. Hypothalamic endoplasmic reticulum (ER) stress is one of the most important pathways that modulates inflammation, neuronal function, and energy balance. This review aimed to investigate the role of hypothalamic ER stress in SCZ and antipsychotic-induced weight gain/obesity. Preliminary evidence indicates that SCZ is associated with reduced dopamine D2 receptor (DRD2) signaling, which significantly regulates the ER stress pathway, suggesting the importance of ER stress in SCZ and its related metabolic disorders. Antipsychotics such as olanzapine activate ER stress in hypothalamic neurons. These effects may induce decreased proopiomelanocortin (POMC) processing, increased neuropeptide Y (NPY) and agouti-related protein (AgRP) expression, autophagy, and leptin and insulin resistance, resulting in hyperphagia, decreased energy expenditure, and central inflammation, thereby causing weight gain. By activating ER stress, antipsychotics such as olanzapine activate hypothalamic astrocytes and Toll-like receptor 4 signaling, thereby causing inflammation and weight gain/obesity. Moreover, evidence suggests that antipsychotic-induced ER stress may be related to their antagonistic effects on neurotransmitter receptors such as DRD2 and the histamine H1 receptor. Taken together, ER stress inhibitors could be a potential effective intervention against SCZ and antipsychotic-induced weight gain and inflammation.
Collapse
Affiliation(s)
- Ruqin Zhou
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- *Correspondence: Meng He,
| | - Jun Fan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Ruoxi Li
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufeng Zuo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Benben Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
- Guanbin Gao,
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- Taolei Sun,
| |
Collapse
|
9
|
Watkins OC, Yong HEJ, Mah TKL, Cracknell-Hazra VKB, Pillai RA, Selvam P, Sharma N, Cazenave-Gassiot A, Bendt AK, Godfrey KM, Lewis RM, Wenk MR, Chan SY. Sex-Dependent Regulation of Placental Oleic Acid and Palmitic Acid Metabolism by Maternal Glycemia and Associations with Birthweight. Int J Mol Sci 2022; 23:8685. [PMID: 35955818 PMCID: PMC9369035 DOI: 10.3390/ijms23158685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 01/27/2023] Open
Abstract
Pregnancy complications such as maternal hyperglycemia increase perinatal mortality and morbidity, but risks are higher in males than in females. We hypothesized that fetal sex-dependent differences in placental palmitic-acid (PA) and oleic-acid (OA) metabolism influence such risks. Placental explants (n = 22) were incubated with isotope-labeled fatty acids (13C-PA or 13C-OA) for 24 or 48 h and the production of forty-seven 13C-PA lipids and thirty-seven 13C-OA lipids quantified by LCMS. Linear regression was used to investigate associations between maternal glycemia, BMI and fetal sex with 13C lipids, and between 13C lipids and birthweight centile. Placental explants from females showed greater incorporation of 13C-OA and 13C-PA into almost all lipids compared to males. Fetal sex also influenced relationships with maternal glycemia, with many 13C-OA and 13C-PA acylcarnitines, 13C-PA-diacylglycerols and 13C-PA phospholipids positively associated with glycemia in females but not in males. In contrast, several 13C-OA triacylglycerols and 13C-OA phospholipids were negatively associated with glycemia in males but not in females. Birthweight centile in females was positively associated with six 13C-PA and three 13C-OA lipids (mainly acylcarnitines) and was negatively associated with eight 13C-OA lipids, while males showed few associations. Fetal sex thus influences placental lipid metabolism and could be a key modulator of the impact of maternal metabolic health on perinatal outcomes, potentially contributing toward sex-specific adaptions in which females prioritize survival.
Collapse
Affiliation(s)
- Oliver C. Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Hannah E. J. Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
| | - Tania Ken Lin Mah
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
| | - Victoria K. B. Cracknell-Hazra
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK
| | - Reshma Appukuttan Pillai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Preben Selvam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry and Precision Medicine TRP, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Anne K. Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Keith M. Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO17 1BJ, UK
| | - Rohan M. Lewis
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Markus R. Wenk
- Department of Biochemistry and Precision Medicine TRP, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
| |
Collapse
|
10
|
Li L, Gu Z, Zhang J. CTRP9 overexpression attenuates palmitic acid‑induced inflammation, apoptosis and impaired migration in HTR8/SVneo cells through AMPK/SREBP1c signaling. Exp Ther Med 2022; 24:459. [PMID: 35747146 PMCID: PMC9204553 DOI: 10.3892/etm.2022.11386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/01/2022] [Indexed: 11/11/2022] Open
Abstract
Obesity in pregnant mothers often leads to a range of obstetric complications, including miscarriage, pre-eclampsia, gestational hypertension and diabetes. C1q/TNF-related protein 9 (CTRP9) is an adipokine with an anti-inflammatory effect. The aim of the present study was to identify the role of CTRP9 in the pathogenesis of maternal obesity during pregnancy. Following treatment with palmitic acid (PA), HTR8/SVneo cell viability and CTRP9 expression were analyzed using Cell Counting Kit-8 (CCK-8), reverse transcription-quantitative PCR (RT-qPCR) and western blot analyses. The effects of CTRP9 overexpression on cell viability, apoptosis, pro-inflammatory cytokine levels and migration were assessed using CCK-8, TUNEL, RT-qPCR and Transwell assays, respectively. Subsequently, sterol-regulatory element binding protein 1c (SREBP1c) overexpression efficiency was verified using RT-qPCR, and its effects on cell viability, apoptosis, pro-inflammatory cytokines and migration damage were then examined in HTR8/SVneo cells. The results showed that CTRP9 overexpression attenuated the inhibition of cell viability and apoptosis caused by PA in HTR8/SVneo cells, reduced pro-inflammatory cytokine release, improved cell migration and regulated the protein expression level of AMP-activated protein kinase (AMPK)/SREBP1c signaling. In addition, CTRP9 inhibited SREBP1c expression through AMPK signaling, thereby attenuating the inflammation, apoptosis and inhibited migration caused by PA in HTR8/SVneo cells. In brief, CTRP9 protected against inflammation, apoptosis and migration defects in HTR8/SVneo cells exposed to PA treatment through AMPK/SREBP1c signaling, which suggested the potential role of CTRP9 in alleviating the toxicity of PA.
Collapse
Affiliation(s)
- Li Li
- Department of Obstetrics and Gynecology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Zhongyi Gu
- Department of Obstetrics and Gynecology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Junjie Zhang
- Department of Obstetrics and Gynecology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
11
|
Davis J, Mire E. Maternal obesity and developmental programming of neuropsychiatric disorders: An inflammatory hypothesis. Brain Neurosci Adv 2021; 5:23982128211003484. [PMID: 33889757 PMCID: PMC8040564 DOI: 10.1177/23982128211003484] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
Maternal obesity is associated with the development of a variety of neuropsychiatric disorders; however, the mechanisms behind this association are not fully understood. Comparison between maternal immune activation and maternal obesity reveals similarities in associated impairments and maternal cytokine profile. Here, we present a summary of recent evidence describing how inflammatory processes contribute towards the development of neuropsychiatric disorders in the offspring of obese mothers. This includes discussion on how maternal cytokine levels, fatty acids and placental inflammation may interact with foetal neurodevelopment through changes to microglial behaviour and epigenetic modification. We also propose an exosome-mediated mechanism for the disruption of brain development under maternal obesity and discuss potential intervention strategies.
Collapse
Affiliation(s)
- Jonathan Davis
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Erik Mire
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
12
|
Eastman AJ, Moore RE, Townsend SD, Gaddy JA, Aronoff DM. The Influence of Obesity and Associated Fatty Acids on Placental Inflammation. Clin Ther 2021; 43:265-278. [PMID: 33487441 DOI: 10.1016/j.clinthera.2020.12.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Maternal obesity, affecting nearly 1 in 4 pregnancies, is associated with increased circulating saturated fatty acids, such as palmitate. These fatty acids are implicated in placental inflammation, which may in turn exacerbate both maternal-fetal tolerance and responses to pathogens, such as group B Streptococcus. In this review, we address the question, "How do obesity and associated fatty acids influence placental inflammation?" METHODS In this narrative review, we searched PubMed and Google Scholar using combinations of the key words placental inflammation or pregnancy and lipids, fatty acids, obesity, palmitate, or other closely related search terms. We also used references found within these articles that may have been absent from our original search queries. We analyzed methods and key results of these articles to compare and contrast their findings, which were occasionally at odds with each other. FINDINGS Although obesity can be studied as a whole, complex phenomena with in vivo mouse models and human samples from patients with obesity, in vitro modeling often relies on the treatment of cells or tissues with ≥1 fatty acids and occasionally other compounds (eg, glucose and insulin). We found that palmitate, most commonly used in vitro to recreate hallmarks of obesity, induces apoptosis, oxidative stress, mitochondrial dysfunction, autophagy defects, and inflammasome activation in many placental cell types. We compare this to in vivo models of obesity wherever possible. We found that obesity as a whole may have more complex regulation of these phenomena (apoptosis, oxidative stress, mitochondrial dysfunction, autophagy defects, and inflammasome activation) compared with in vitro models of fatty acid treatment (primarily palmitate) because of the presence of unsaturated fatty acids (ie, oleate), which may have anti-inflammatory effects. IMPLICATIONS The interaction of unsaturated fatty acids with saturated fatty acids may ameliorate many inflammatory effects of saturated fatty acids alone, which complicates interpretation of in vitro studies that focus on a particular fatty acid in isolation. This complication may explain why certain studies of obesity in vivo have differing outcomes from studies of specific fatty acids in vitro.
Collapse
Affiliation(s)
- Alison J Eastman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca E Moore
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | | | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Tennessee Valley Healthcare Systems, Department of Veterans Affairs, Nashville, TN, USA
| | - David M Aronoff
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
13
|
Chambers M, Rees A, Cronin JG, Nair M, Jones N, Thornton CA. Macrophage Plasticity in Reproduction and Environmental Influences on Their Function. Front Immunol 2021; 11:607328. [PMID: 33519817 PMCID: PMC7840613 DOI: 10.3389/fimmu.2020.607328] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages are key components of the innate immune system and exhibit extensive plasticity and heterogeneity. They play a significant role in the non-pregnant cycling uterus and throughout gestation they contribute to various processes underpinning reproductive success including implantation, placentation and parturition. Macrophages are also present in breast milk and impart immunomodulatory benefits to the infant. For a healthy pregnancy, the maternal immune system must adapt to prevent fetal rejection and support development of the semi-allogenic fetus without compromising host defense. These functions are dependent on macrophage polarization which is governed by the local tissue microenvironmental milieu. Disruption of this microenvironment, possibly by environmental factors of infectious and non-infectious origin, can affect macrophage phenotype and function and is linked to adverse obstetric outcomes, e.g. spontaneous miscarriage and preterm birth. Determining environmental influences on cellular and molecular mechanisms that control macrophage polarization at the maternal-fetal interface and the role of this in pregnancy complications could support approaches to alleviating adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Megan Chambers
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - April Rees
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - James G Cronin
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Manju Nair
- Maternity and Child Health, Singleton Hospital, Swansea Bay University Health Board, Swansea, United Kingdom
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Catherine A Thornton
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| |
Collapse
|
14
|
Evolution of the Human Diet and Its Impact on Gut Microbiota, Immune Responses, and Brain Health. Nutrients 2021; 13:nu13010196. [PMID: 33435203 PMCID: PMC7826636 DOI: 10.3390/nu13010196] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
The relatively rapid shift from consuming preagricultural wild foods for thousands of years, to consuming postindustrial semi-processed and ultra-processed foods endemic of the Western world less than 200 years ago did not allow for evolutionary adaptation of the commensal microbial species that inhabit the human gastrointestinal (GI) tract, and this has significantly impacted gut health. The human gut microbiota, the diverse and dynamic population of microbes, has been demonstrated to have extensive and important interactions with the digestive, immune, and nervous systems. Western diet-induced dysbiosis of the gut microbiota has been shown to negatively impact human digestive physiology, to have pathogenic effects on the immune system, and, in turn, cause exaggerated neuroinflammation. Given the tremendous amount of evidence linking neuroinflammation with neural dysfunction, it is no surprise that the Western diet has been implicated in the development of many diseases and disorders of the brain, including memory impairments, neurodegenerative disorders, and depression. In this review, we discuss each of these concepts to understand how what we eat can lead to cognitive and psychiatric diseases.
Collapse
|
15
|
Faustmann G, Tiran B, Trajanoski S, Obermayer-Pietsch B, Gruber HJ, Ribalta J, Roob JM, Winklhofer-Roob BM. Activation of nuclear factor-kappa B subunits c-Rel, p65 and p50 by plasma lipids and fatty acids across the menstrual cycle. Free Radic Biol Med 2020; 160:488-500. [PMID: 32846215 DOI: 10.1016/j.freeradbiomed.2020.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
This study focused on a comprehensive analysis of the canonical activation pathway of the redox-sensitive transcription factor nuclear factor-kappa B (NF-κB) in peripheral blood mononuclear cells, addressing c-Rel, p65 and p50 activation in 28 women at early (T1) and late follicular (T2) and mid (T3) and late luteal (T4) phase of the menstrual cycle, and possible relations with fasting plasma lipids and fatty acids. For the first time, strong inverse relations of c-Rel with apolipoprotein B were observed across the cycle, while those with LDL cholesterol, triglycerides as well as saturated (SFA), particularly C14-C22 SFA, monounsaturated (MUFA), and polyunsaturated fatty acids (PUFA) clustered at T2. In contrast, p65 was positively related to LDL cholesterol and total n-6 PUFA, while p50 did not show any relations. C-Rel was not directly associated with estradiol and progesterone, but data suggested an indirect C22:5n-3-mediated effect of progesterone. Strong positive relations between estradiol and individual SFA, MUFA and n-3 PUFA at T1 were confined to C18 fatty acids; C18:3n-3 was differentially associated with estradiol (positively) and progesterone (inversely). Given specific roles of c-Rel activation in immune tolerance, inhibition of c-Rel activation by higher plasma apolipoprotein B and individual fatty acid concentrations could have clinical implications for female fertility.
Collapse
Affiliation(s)
- Gernot Faustmann
- Human Nutrition & Metabolism Research and Training Center, Institute of Molecular Biosciences, University of Graz, Graz, Austria; Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Beate Tiran
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Josep Ribalta
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili and Institut d'Investigació Sanitària Pere Virgili, Reus, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Spain
| | - Johannes M Roob
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Brigitte M Winklhofer-Roob
- Human Nutrition & Metabolism Research and Training Center, Institute of Molecular Biosciences, University of Graz, Graz, Austria.
| |
Collapse
|
16
|
Rampersaud AM, Dunk CE, Lye SJ, Renaud SJ. Palmitic acid induces inflammation in placental trophoblasts and impairs their migration toward smooth muscle cells through plasminogen activator inhibitor-1. Mol Hum Reprod 2020; 26:850-865. [PMID: 32898274 DOI: 10.1093/molehr/gaaa061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
A critical component of early human placental development includes migration of extravillous trophoblasts (EVTs) into the decidua. EVTs migrate toward and displace vascular smooth muscle cells (SMCs) surrounding several uterine structures, including spiral arteries. Shallow trophoblast invasion features in several pregnancy complications including preeclampsia. Maternal obesity is a risk factor for placental dysfunction, suggesting that factors within an obese environment may impair early placental development. Herein, we tested the hypothesis that palmitic acid, a saturated fatty acid circulating at high levels in obese women, induces an inflammatory response in EVTs that hinders their capacity to migrate toward SMCs. We found that SMCs and SMC-conditioned media stimulated migration and invasion of an EVT-like cell line, HTR8/SVneo. Palmitic acid impaired EVT migration and invasion toward SMCs, and induced expression of several vasoactive and inflammatory mediators in EVTs, including endothelin, interleukin (IL)-6, IL-8 and PAI1. PAI1 was increased in plasma of women with early-onset preeclampsia, and PAI1-deficient EVTs were protected from the anti-migratory effects of palmitic acid. Using first trimester placental explants, palmitic acid exposure decreased EVT invasion through Matrigel. Our findings reveal that palmitic acid induces an inflammatory response in EVTs and attenuates their migration through a mechanism involving PAI1. High levels of palmitic acid in pathophysiological situations like obesity may impair early placental development and predispose to placental dysfunction.
Collapse
Affiliation(s)
- Amanda M Rampersaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Caroline E Dunk
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Stephen J Lye
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
17
|
Associations of Plasma Fatty Acid Patterns during Pregnancy with Respiratory and Allergy Outcomes at School Age. Nutrients 2020; 12:nu12103057. [PMID: 33036333 PMCID: PMC7601105 DOI: 10.3390/nu12103057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/20/2022] Open
Abstract
Fatty acids might play a role in asthma and allergy development as they can modulate immune responses. We examined among 4260 mother-child pairs participating in a population-based cohort the associations of maternal plasma fatty acid patterns during pregnancy with a child's respiratory and allergy outcomes at school-age. In mid-pregnancy, 22 individual fatty acids were measured from maternal blood. Three patterns were previously identified by principal component analysis: A 'high n-6 polyunsaturated fatty acid (PUFA)', a 'monounsaturated and saturated fatty acid', and a 'high n-3 PUFA' pattern. At the age of 10 years, a child's lung function was assessed by spirometry, current asthma and physician-diagnosed inhalant allergy by questionnaire, and inhalant allergic sensitization by skin prick tests. A higher 'high n-6 PUFA' pattern was associated with a higher forced expiratory volume in 1 s/forced vital capacity and forced expiratory flow after exhaling 75% of forced vital capacity (Z-score difference (95% CI) 0.04 (0, 0.07) and 0.04 (0.01, 0.07), respectively, per SD increase in the fatty acid pattern). We observed no associations of maternal fatty acid patterns with a child's asthma or allergy outcomes. Our results showed limited associations of maternal patterns of high n-6 PUFA concentrations in pregnancy with a better lung function in school-aged children.
Collapse
|
18
|
Placental function in maternal obesity. Clin Sci (Lond) 2020; 134:961-984. [PMID: 32313958 DOI: 10.1042/cs20190266] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Maternal obesity is associated with pregnancy complications and increases the risk for the infant to develop obesity, diabetes and cardiovascular disease later in life. However, the mechanisms linking the maternal obesogenic environment to adverse short- and long-term outcomes remain poorly understood. As compared with pregnant women with normal BMI, women entering pregnancy obese have more pronounced insulin resistance, higher circulating plasma insulin, leptin, IGF-1, lipids and possibly proinflammatory cytokines and lower plasma adiponectin. Importantly, the changes in maternal levels of nutrients, growth factors and hormones in maternal obesity modulate placental function. For example, high insulin, leptin, IGF-1 and low adiponectin in obese pregnant women activate mTOR signaling in the placenta, promoting protein synthesis, mitochondrial function and nutrient transport. These changes are believed to increase fetal nutrient supply and contribute to fetal overgrowth and/or adiposity in offspring, which increases the risk to develop disease later in life. However, the majority of obese women give birth to normal weight infants and these pregnancies are also associated with activation of inflammatory signaling pathways, oxidative stress, decreased oxidative phosphorylation and lipid accumulation in the placenta. Recent bioinformatics approaches have expanded our understanding of how maternal obesity affects the placenta; however, the link between changes in placental function and adverse outcomes in obese women giving birth to normal sized infants is unclear. Interventions that specifically target placental function, such as activation of placental adiponectin receptors, may prevent the transmission of metabolic disease from obese women to the next generation.
Collapse
|
19
|
Shook LL, Kislal S, Edlow AG. Fetal brain and placental programming in maternal obesity: A review of human and animal model studies. Prenat Diagn 2020; 40:1126-1137. [PMID: 32362000 DOI: 10.1002/pd.5724] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/26/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
Both human epidemiologic and animal model studies demonstrate that prenatal and lactational exposure to maternal obesity and high-fat diet are associated with adverse neurodevelopmental outcomes in offspring. Neurodevelopmental outcomes described in offspring of obese women include cognitive impairment, autism spectrum disorder (ASD), attention deficit hyperactivity disorder, anxiety and depression, disordered eating, and propensity for reward-driven behavior, among others. This review synthesizes human and animal data linking maternal obesity and high-fat diet consumption to abnormal fetal brain development, and neurodevelopmental and psychiatric morbidity in offspring. It highlights key mechanisms by which maternal obesity and maternal diet impact fetal and offspring development, and sex differences in offspring programming. In addition, we review placental effects of maternal obesity, and the role the placenta might play as an indicator vs mediator of fetal programming.
Collapse
Affiliation(s)
- Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sezen Kislal
- Massachusetts General Hospital Research Institute, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital Research Institute, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Khambadkone SG, Cordner ZA, Tamashiro KLK. Maternal stressors and the developmental origins of neuropsychiatric risk. Front Neuroendocrinol 2020; 57:100834. [PMID: 32084515 PMCID: PMC7243665 DOI: 10.1016/j.yfrne.2020.100834] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/23/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
The maternal environment during pregnancy is critical for fetal development and perinatal perturbations can prime offspring disease risk. Here, we briefly review evidence linking two well-characterized maternal stressors - psychosocial stress and infection - to increased neuropsychiatric risk in offspring. In the current climate of increasing obesity and globalization of the Western-style diet, maternal overnutrition emerges as a pressing public health concern. We focus our attention on recent epidemiological and animal model evidence showing that, like psychosocial stress and infection, maternal overnutrition can also increase offspring neuropsychiatric risk. Using lessons learned from the psychosocial stress and infection literature, we discuss how altered maternal and placental physiology in the setting of overnutrition may contribute to abnormal fetal development and resulting neuropsychiatric outcomes. A better understanding of converging pathophysiological pathways shared between stressors may enable development of interventions against neuropsychiatric illnesses that may be beneficial across stressors.
Collapse
Affiliation(s)
- Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Hussein AM, Wang Y, Mathieu J, Margaretha L, Song C, Jones DC, Cavanaugh C, Miklas JW, Mahen E, Showalter MR, Ruzzo WL, Fiehn O, Ware CB, Blau CA, Ruohola-Baker H. Metabolic Control over mTOR-Dependent Diapause-like State. Dev Cell 2020; 52:236-250.e7. [PMID: 31991105 DOI: 10.1016/j.devcel.2019.12.018] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 09/13/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Abstract
Regulation of embryonic diapause, dormancy that interrupts the tight connection between developmental stage and time, is still poorly understood. Here, we characterize the transcriptional and metabolite profiles of mouse diapause embryos and identify unique gene expression and metabolic signatures with activated lipolysis, glycolysis, and metabolic pathways regulated by AMPK. Lipolysis is increased due to mTORC2 repression, increasing fatty acids to support cell survival. We further show that starvation in pre-implantation ICM-derived mouse ESCs induces a reversible dormant state, transcriptionally mimicking the in vivo diapause stage. During starvation, Lkb1, an upstream kinase of AMPK, represses mTOR, which induces a reversible glycolytic and epigenetically H4K16Ac-negative, diapause-like state. Diapause furthermore activates expression of glutamine transporters SLC38A1/2. We show by genetic and small molecule inhibitors that glutamine transporters are essential for the H4K16Ac-negative, diapause state. These data suggest that mTORC1/2 inhibition, regulated by amino acid levels, is causal for diapause metabolism and epigenetic state.
Collapse
Affiliation(s)
- Abdiasis M Hussein
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA
| | - Julie Mathieu
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Lilyana Margaretha
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Molecular and Cellular Biology, University of Washington, Seattle, WA 98109, USA
| | - Chaozhong Song
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Daniel C Jones
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA
| | - Christopher Cavanaugh
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Jason W Miklas
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Elisabeth Mahen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Megan R Showalter
- West Coast Metabolomics Center, University of California, Davis, Davis, CA 95616, USA
| | - Walter L Ruzzo
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA 95616, USA
| | - Carol B Ware
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - C Anthony Blau
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
22
|
Lee S, Shin J, Hong Y, Shin SM, Shin HW, Shin J, Lee SK, Park HW. Sestrin2 alleviates palmitate-induced endoplasmic reticulum stress, apoptosis, and defective invasion of human trophoblast cells. Am J Reprod Immunol 2020; 83:e13222. [PMID: 31958198 DOI: 10.1111/aji.13222] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/07/2020] [Accepted: 01/13/2020] [Indexed: 12/16/2022] Open
Abstract
PROBLEM Maternal obesity induces elevated saturated fatty acid palmitate levels in the blood and causes pregnancy complications such as gestational diabetes, preeclampsia, fetal growth abnormalities, and stillbirth. Sestrin2, a highly conserved stress-inducible protein, is involved in the cellular responses of various stress conditions and homeostatic regulation. However, the effects of Sestrin2 on trophoblast cells have not yet been investigated. Here, we investigated the role of Sestrin2 in palmitate-induced lipotoxicity and its underlying mechanisms in human first-trimester trophoblast cells (Sw.71). METHOD OF STUDY Mouse placental tissues were obtained from low-fat diet-fed mice (n = 14) and high-fat diet-fed mice (n = 14) at gestation day 17.5. Sw.71 cells were treated with palmitate or bovine serum albumin as vehicle controls. The role of Sestrin2 in palmitate-induced lipotoxicity was examined by immunocytochemistry, immunoblot analysis, quantitative real-time PCR, and invasion assay. RESULTS Expression of placental Sestrin2 was elevated in high-fat diet-fed dams compared to that of low-fat diet-fed dams. Prolonged treatment of Sw.71 cells with palmitate-induced endoplasmic reticulum (ER) stress-dependent expressions of Sestrin2 protein and mRNA, and the treatment also triggered apoptosis. Knockdown of Sestrin2 increased palmitate-mediated ER stress, inflammatory signaling, and apoptosis. Furthermore, Sestrin2 suppressed impaired trophoblast invasion caused by palmitate and attenuated palmitate-induced ER stress and inflammation via AMPK/mTORC1 pathways. CONCLUSION Our study provides the relationship between Sestrin2, AMPK/mTORC1 pathway, and trophoblast function, suggesting that Sestrin2 may be a novel potential therapeutic target for the prevention of pregnancy complications.
Collapse
Affiliation(s)
- Solji Lee
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Jiha Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Yeji Hong
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Korea
| | - Seong Min Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Hye Won Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Jongdae Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea.,Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, Korea
| | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Korea.,Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, Korea
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea.,Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, Korea
| |
Collapse
|
23
|
Effects of Diets Enriched in Linseed and Fish Oil on the Expression Pattern of Toll-Like Receptors 4 and Proinflammatory Cytokines on Gonadal Axis and Reproductive Organs in Rabbit Buck. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4327470. [PMID: 32064024 PMCID: PMC6996712 DOI: 10.1155/2020/4327470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/11/2019] [Accepted: 12/20/2019] [Indexed: 01/05/2023]
Abstract
Infections of the genital tract can perturb the fertility in humans and animals. Pathogen recognition and activation of innate immunity onset through the pattern recognition receptor activation, such as Toll-like receptor 4 (TLR4), leading to the production of proinflammatory cytokines and mediators. TLR4 is expressed both on leukocytes and nonimmune cells. Rabbit TLR4 shows great similarity to its human counterpart. Moreover, the TLR4 signalling pathway could be modulated by long-chain polyunsaturated fatty acids (LC-PUFA). The objectives of this study were (i) to determine the expression levels of TLR4 and proinflammatory cytokines in the reproductive hypothalamic-gonadal axis of the male rabbit and (ii) to evaluate if the n-3 PUFA-enriched diets can modify their expression levels in the tissues and LC-PUFA profiles in seminal plasma. Fifteen rabbit bucks (n = 5/experimental group) were fed with different diets: commercial standard (group C), rich in extruded linseed (10%, group L), and in fish oil (3%, group FO) for 110 days. TLR4, TNF-α, and IL-1β mRNA were ubiquitously expressed throughout the hypothalamic-gonadal axis. However, TLR4 mRNA expression was lower in the hypothalamus than the epididymis (P < 0.01), seminal vesicles (P < 0.01), and pituitary gland (P < 0.05). Dietary enrichment in PUFA did not modify the gene expression profile nor the histological characteristics of the tissues. Conversely in seminal plasma, rabbits fed with L and FO had lower n-6 (P < 0.05), LC-PUFA n-6 (P < 0.05), and n-6/n-3 ratio (P < 0.05) but higher n-3 (P < 0.001) and LC-PUFA n-3 (P < 0.01) compared to the control group. Our study builds a map of the gene expression of TRL4 and proinflammatory cytokines in the reproductive hypothalamic-gonadal axis of the male rabbit, fundamental step for understanding the immune defence mechanisms. Diets enriched in LC-PUFA did not affect basal gene expression but modulated sperm fatty acid composition. Finally, rabbit may be an excellent animal model to study the relationship between inflammation and infertility, and the nutritional modulation of immune functions.
Collapse
|
24
|
Zhou H, Urso CJ, Jadeja V. Saturated Fatty Acids in Obesity-Associated Inflammation. J Inflamm Res 2020; 13:1-14. [PMID: 32021375 PMCID: PMC6954080 DOI: 10.2147/jir.s229691] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/11/2019] [Indexed: 01/14/2023] Open
Abstract
Obesity is a major risk factor for the development of various pathological conditions including insulin resistance, diabetes, cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Central to these conditions is obesity-associated chronic low-grade inflammation in many tissues including adipose, liver, muscle, kidney, pancreas, and brain. There is increasing evidence that saturated fatty acids (SFAs) increase the phosphorylation of MAPKs, enhance the activation of transcription factors such as nuclear factor (NF)-κB, and elevate the expression of inflammatory genes. This paper focuses on the mechanisms by which SFAs induce inflammation. SFAs may induce the expression inflammatory genes via different pathways including toll-like receptor (TLR), protein kinase C (PKC), reactive oxygen species (ROS), NOD-like receptors (NLRs), and endoplasmic reticulum (ER) stress. These findings suggest that SFAs act as an important link between obesity and inflammation.
Collapse
Affiliation(s)
- Heping Zhou
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - C J Urso
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - Viren Jadeja
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| |
Collapse
|
25
|
A global perspective on the crosstalk between saturated fatty acids and Toll-like receptor 4 in the etiology of inflammation and insulin resistance. Prog Lipid Res 2019; 77:101020. [PMID: 31870728 DOI: 10.1016/j.plipres.2019.101020] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/16/2022]
Abstract
Obesity is featured by chronic systemic low-grade inflammation that eventually contributes to the development of insulin resistance. Toll-like receptor 4 (TLR4) is an important mediator that triggers the innate immune response by activating inflammatory signaling cascades. Human, animal and cell culture studies identified saturated fatty acids (SFAs), the dominant non-esterified fatty acid (NEFA) in the circulation of obese subjects, as non-microbial agonists that trigger the inflammatory response via activating TLR4 signaling, which acts as an important causative link between fatty acid overload, chronic low-grade inflammation and the related metabolic aberrations. The interaction between SFAs and TLR4 may be modulated through the myeloid differentiation primary response gene 88-dependent and independent signaling pathway. Greater understanding of the crosstalk between dietary SFAs and TLR4 signaling in the pathogenesis of metabolic imbalance may facilitate the design of a more efficient pharmacological strategy to alleviate the risk of developing chronic diseases elicited in part by fatty acid overload. The current review discusses recent advances in the impact of crosstalk between SFAs and TLR4 on inflammation and insulin resistance in multiple cell types, tissues and organs in the context of metabolic dysregulation.
Collapse
|
26
|
Chankeaw W, Guo YZ, Båge R, Svensson A, Andersson G, Humblot P. Elevated non-esterified fatty acids impair survival and promote lipid accumulation and pro-inflammatory cytokine production in bovine endometrial epithelial cells. Reprod Fertil Dev 2019; 30:1770-1784. [PMID: 30086824 DOI: 10.1071/rd17537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 06/11/2018] [Indexed: 12/11/2022] Open
Abstract
Elevated non-esterified fatty acids (NEFAs) are associated with negative effects on bovine theca, granulosa and oviductal cells but the effects of NEFAs on bovine endometrial epithelial cells (bEECs) are not as well documented. The objective of this study was to define the effects of NEFAs on bEECs. Postprimary bEECs were treated with 150, 300 or 500µM of either palmitic acid (PA), stearic acid (SA) or oleic acid (OA) or a mixture of NEFAs (150µM of each FA) or 0.5% final concentration of vehicle ethanol (control). Viability and proliferation of bEECs exposed to 150µM of each NEFA or a mixture of NEFAs were unaffected. Increased lipid accumulation was found in all treated groups (P<0.01). In cells exposed to 500µM of each NEFA and 300µM PA decreased cell viability (P<0.001), proliferation (P<0.05) and increased apoptosis (P<0.05) were observed. Treatment with 500µM OA, PA and SA had the strongest effects on cell viability, proliferation and apoptosis (P<0.05). Treatment with PA and OA increased interleukin-6 (IL-6) concentrations (P<0.05), whereas only the highest concentration of PA, OA and SA stimulated IL-8 production (P<0.05). These results suggest that high concentrations of NEFAs may impair endometrial function with more or less pronounced effects depending on the type of NEFA and time of exposure.
Collapse
Affiliation(s)
- W Chankeaw
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Sciences, Swedish University of Agricultural Sciences, SLU, P.O. Box 7054, 750 07 Uppsala, Sweden
| | - Y Z Guo
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Sciences, Swedish University of Agricultural Sciences, SLU, P.O. Box 7054, 750 07 Uppsala, Sweden
| | - R Båge
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Sciences, Swedish University of Agricultural Sciences, SLU, P.O. Box 7054, 750 07 Uppsala, Sweden
| | - A Svensson
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Sciences, Swedish University of Agricultural Sciences, SLU, P.O. Box 7054, 750 07 Uppsala, Sweden
| | - G Andersson
- Department of Animal Breeding and Genetics, Faculty of Veterinary Medicine and Animal Sciences, Swedish University of Agricultural Sciences, SLU, P.O. Box 7023, 750 07 Uppsala, Sweden
| | - P Humblot
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Sciences, Swedish University of Agricultural Sciences, SLU, P.O. Box 7054, 750 07 Uppsala, Sweden
| |
Collapse
|
27
|
Zhao W, Cui L, Huang X, Wang S, Li D, Li L, Sun Y, Du M. Activation of Rev-erbα attenuates lipopolysaccharide-induced inflammatory reactions in human endometrial stroma cells via suppressing TLR4-regulated NF-κB activation. Acta Biochim Biophys Sin (Shanghai) 2019; 51:908-914. [PMID: 31411318 DOI: 10.1093/abbs/gmz078] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022] Open
Abstract
Perturbation of the circadian rhythm damages the biological characteristics of cells and leads to their dysfunction. Rev-erbα, an important gene in the transcription-translation loop of circadian rhythm, is involved in regulating the balance between pro-inflammation and anti-inflammation. The disruption of this balance in human endometrial stroma cells (hESCs) destroys their biological behavior function in maintaining the menstrual cycle and embryonic implantation. Whether pharmacological modulation of Rev-erbα affects the inflammation of hESCs remains unclear. In this study, we treated hESCs with lipopolysaccharide (LPS) and found that LPS treatment increased the mRNA levels of pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6, IL-8, IL-18, and TNFα, and the secretion of IL-6. SR9009, a Rev-erbα agonist, significantly alleviated the LPS-induced production of pro-inflammatory cytokines in hESCs. Meanwhile, knockdown of Rev-erbα increased the expressions of IL-1β, IL-6, and IL-8, accompanied by an increased mRNA level of the core clock gene Bmal1. Western blot analysis showed that SR9009 inhibited the expression of toll-like receptor 4 (TLR4) and the activation of NF-κB induced by LPS. All these findings suggested that pharmacological activation of Rev-erbα attenuated the LPS-induced inflammatory response of hESCs by suppressing TLR4-regulated NF-κB activation. This study may provide a strategy for preventing inflammation-related endometrial dysfunction and infertility or recurrent implantation failure.
Collapse
Affiliation(s)
- Weijie Zhao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Liyuan Cui
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Xixi Huang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Songcun Wang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Dajin Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Liping Li
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Yan Sun
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Meirong Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| |
Collapse
|
28
|
Peng XR, Feng L, Jiang WD, Wu P, Liu Y, Jiang J, Kuang SY, Tang L, Zhou XQ. Supplementation exogenous bile acid improved growth and intestinal immune function associated with NF-κB and TOR signalling pathways in on-growing grass carp (Ctenopharyngodon idella): Enhancement the effect of protein-sparing by dietary lipid. FISH & SHELLFISH IMMUNOLOGY 2019; 92:552-569. [PMID: 31252043 DOI: 10.1016/j.fsi.2019.06.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/14/2019] [Accepted: 06/24/2019] [Indexed: 06/09/2023]
Abstract
This study investigated the effects of bile acid (BA) supplementation on growth performance, intestinal immune function and the mRNA expression of the related signalling molecules in on-growing grass carp (Ctenopharyngodon idella). A total of 540 healthy grass carp (mean weight 179.85 ± 1.34 g) were fed a normal protein and lipid (NPNL) diet containing 29% crude protein (CP) and 5% ether extract (EE), and five low-protein and high-lipid (LPHL) diets (26% CP, 6% EE) with graded levels of BA (0-320 mg/kg diet) for 50 days. The fish were then challenged with Aeromonas hydrophila for 14 days. The results indicated that compared with the NPNL diet, the LPHL diet (unsupplemented BA) suppressed the growth performance, intestinal development and enteritis resistance capability and impaired the partial intestinal immune function of on-growing grass carp. Whereas in the LPHL diet, optimal BA supplementation significantly improved fish growth performance (percent weight gain, specific growth rate, feed intake and feed efficiency) and intestinal growth and function (intestine weight, intestine length and intestosomatic index), increased beneficial bacteria Lactobacillus and Bifidobacterium amounts, decreased harmful bacteria Aeromonas and Escherichia coli amounts, elevated lysozyme and acid phosphatase activities, increased complement (C3 and C4) and immunoglobulin M contents, and upregulated β-defensin-1, hepcidin, liver expressed antimicrobial peptide 2A (LEAP-2A), LEAP-2B, Mucin2, interleukin 10 (IL-10), IL-11, transforming growth factor (TGF)-β1, TGF-β2, IL-4/13A (not IL-4/13B), TOR, S6K1 and inhibitor of κBα (IκBα) mRNA levels. In addition, optimal BA supplementation in the LPHL diet downregulated tumour necrosis factor α (TNF-α), interferon γ2 (IFN-γ2), IL-1β, IL-6, IL-8, IL-15, IL-17D, IL-12p35, IL-12p40 (rather than proximal intestine (PI) or mid intestine (MI), nuclear factor kappa B p65 (NF-κB p65) (except NF-κB p52), c-Rel, IκB kinase β (IKKβ), IKKγ (except IKKα), eIF4E-binding proteins (4E-BP)1 and 4E-BP2 mRNA levels in all three intestinal segments of on-growing grass carp (P < 0.05). These findings suggest that BA supplementation in the LPHL diet improves growth and intestinal immune function of fish. Furthermore, 240 mg/kg BA supplementation in the LPHL diet was superior to the NPNL diet in improving growth and enhancing intestinal immune function of fish. Finally, based on percent weight gain, feed intake, protecting fish against enteritis, lysozyme activity in MI and acid phosphatase activity in distal intestine (DI), the optimal BA supplementation for on-growing grass carp were estimated to be 168.98, 170.23, 166.67, 176.50 and 191.97 mg/kg diet, respectively.
Collapse
Affiliation(s)
- Xiu-Rong Peng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-resistant Nutrition, Sichuan Province, China.
| |
Collapse
|
29
|
Khan MA, Pace-Asciak C, Al-Hassan JM, Afzal M, Liu YF, Oommen S, Paul BM, Nair D, Palaniyar N. Furanoid F-Acid F6 Uniquely Induces NETosis Compared to C16 and C18 Fatty Acids in Human Neutrophils. Biomolecules 2018; 8:biom8040144. [PMID: 30428625 PMCID: PMC6315434 DOI: 10.3390/biom8040144] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 01/01/2023] Open
Abstract
Various biomolecules induce neutrophil extracellular trap (NET) formation or NETosis. However, the effect of fatty acids on NETosis has not been clearly established. In this study, we focused on the NETosis-inducing ability of several lipid molecules. We extracted the lipid molecules present in Arabian Gulf catfish (Arius bilineatus, Val) skin gel, which has multiple therapeutic activities. Gas chromatography⁻mass spectrometry (GC-MS) analysis of the lipid fraction-3 from the gel with NETosis-inducing activity contained fatty acids including a furanoid F-acid (F6; 12,15-epoxy-13,14-dimethyleicosa-12,14-dienoic acid) and common long-chain fatty acids such as palmitic acid (PA; C16:0), palmitoleic acid (PO; C16:1), stearic acid (SA; C18:0), and oleic acid (OA; C18:1). Using pure molecules, we show that all of these fatty acids induce NETosis to different degrees in a dose-dependent fashion. Notably, F6 induces a unique form of NETosis that is rapid and induces reactive oxygen species (ROS) production by both NADPH oxidase (NOX) and mitochondria. F6 also induces citrullination of histone. By contrast, the common fatty acids (PA, PO, SA, and OA) only induce NOX-dependent NETosis. The activation of the kinases such as ERK (extracellular signal-regulated kinase) and JNK (c-Jun N-terminal kinase) is important for long-chain fatty acid-induced NETosis, whereas, in F-acid-induced NETosis, Akt is additionally needed. Nevertheless, NETosis induced by all of these compounds requires the final chromatin decondensation step of transcriptional firing. These findings are useful for understanding F-acid- and other fatty acid-induced NETosis and to establish the active ingredients with therapeutic potential for regulating diseases involving NET formation.
Collapse
Affiliation(s)
- Meraj A Khan
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
| | - Cecil Pace-Asciak
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
- Departments of Pharmacology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Jassim M Al-Hassan
- Department of Biological Sciences, Faculty of Science, Kuwait University, Safat 13060, Kuwait.
| | - Mohammad Afzal
- Department of Biological Sciences, Faculty of Science, Kuwait University, Safat 13060, Kuwait.
| | - Yuan Fang Liu
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
| | - Sosamma Oommen
- Department of Zoology, CMS College, Kottayam 686001, India.
| | - Bincy M Paul
- Department of Biological Sciences, Faculty of Science, Kuwait University, Safat 13060, Kuwait.
| | - Divya Nair
- Department of Biological Sciences, Faculty of Science, Kuwait University, Safat 13060, Kuwait.
| | - Nades Palaniyar
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
- Departments of Lab Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5G 0A4, Canada.
| |
Collapse
|
30
|
Stirm L, Kovárová M, Perschbacher S, Michlmaier R, Fritsche L, Siegel-Axel D, Schleicher E, Peter A, Pauluschke-Fröhlich J, Brucker S, Abele H, Wallwiener D, Preissl H, Wadsack C, Häring HU, Fritsche A, Ensenauer R, Desoye G, Staiger H. BMI-Independent Effects of Gestational Diabetes on Human Placenta. J Clin Endocrinol Metab 2018; 103:3299-3309. [PMID: 29931171 DOI: 10.1210/jc.2018-00397] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/15/2018] [Indexed: 11/19/2022]
Abstract
PURPOSE Recently, alterations in maternal lipid metabolism were associated with gestational diabetes mellitus (GDM). However, detailed plasma lipid profiles and their relevance for placental and fetal metabolism are currently not understood. METHODS Maternal and placental lipid profiles were characterized in women with GDM and women with normal glucose tolerance (NGT). Inflammatory gene expression was compared in placentas and primary term trophoblasts between the groups. In addition, trophoblasts were stimulated with nonesterified fatty acids (NEFAs), and effects on gene expression were quantified. Finally, placental macrophage content and cord blood concentrations of inflammatory parameters and NEFAs were compared between women with GDM and women with NGT with similar body mass index (BMI). RESULTS Palmitate and stearate levels were elevated in both maternal plasma and placental tissue of women with GDM. Placental GDM-associated elevations of IL6, IL8, and TLR2 expression were reflected in trophoblasts derived from women with GDM. Stimulation of primary trophoblasts with palmitate led to increased mRNA expression and protein release of the cytokine IL6 and the chemokine IL8. In line with this, elevated amounts of CD68-positive cells were quantified in the placental tissue of women with GDM. No GDM-associated elevations in a range of inflammatory parameters and NEFAs in cord blood of NGT vs GDM neonates was found. CONCLUSIONS GDM, independently of BMI, altered maternal plasma NEFAs and the placental lipid profile. GDM was associated with trophoblast and whole-placenta lipoinflammation; however, this was not accompanied by elevated concentrations of inflammatory cytokines or NEFAs in neonatal cord blood.
Collapse
Affiliation(s)
- Laura Stirm
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
| | - Markéta Kovárová
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany
| | - Sarah Perschbacher
- Institute for Social Pediatrics and Adolescent Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Renate Michlmaier
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Louise Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
| | - Dorothea Siegel-Axel
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany
| | - Erwin Schleicher
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany
| | | | - Sara Brucker
- Department of Obstetrics and Gynaecology, University Hospital Tübingen, Tübingen, Germany
| | - Harald Abele
- Department of Obstetrics and Gynaecology, University Hospital Tübingen, Tübingen, Germany
| | - Diethelm Wallwiener
- Department of Obstetrics and Gynaecology, University Hospital Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Munich, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany
| | - Regina Ensenauer
- Institute for Social Pediatrics and Adolescent Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Division of Experimental Pediatrics and Metabolism, University Children's Hospital, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Harald Staiger
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Tübingen, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
31
|
Monmai C, Go SH, Shin IS, You SG, Lee H, Kang SB, Park WJ. Immune-Enhancement and Anti-Inflammatory Activities of Fatty Acids Extracted from Halocynthia aurantium Tunic in RAW264.7 Cells. Mar Drugs 2018; 16:md16090309. [PMID: 30200438 PMCID: PMC6163248 DOI: 10.3390/md16090309] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/12/2018] [Accepted: 08/27/2018] [Indexed: 12/27/2022] Open
Abstract
Halocynthia aurantium, an edible ascidian species, has not been studied scientifically, even though tunicates and ascidians are well-known to contain several unique and biologically active materials. The current study investigated the fatty acid profiles of the H. aurantium tunic and its immune-regulatory effects on RAW264.7 macrophage cells. Results of the fatty acid profile analysis showed a difference in ratios, depending on the fatty acids being analysed, including those of saturated fatty acids (SFA), monounsaturated fatty acids (MUFA), and polyunsaturated fatty acids (PUFA). In particular, omega-3 fatty acids, such as eicosatrienoic acid n-3 (ETA n-3), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), were much higher than omega-6 fatty acids. Moreover, the H. aurantium tunic fatty acids, significantly and dose-dependently, increased the NO and prostaglandin E2 (PGE2) production in RAW264.7 cells, for immune-enhancement without cytotoxicity. In addition, these fatty acids regulated the transcription of immune-associated genes, including iNOS, IL-1β, IL-6, COX-2, and TNF-α. These actions were activated and deactivated via Mitogen-activated protein kinase (MAPK)and NF-κB signaling, to regulate the immune responses. Conversely, the H. aurantium tunic fatty acids effectively suppressed the inflammatory cytokine expressions, including iNOS, IL-1β, IL-6, COX-2, and TNF-α, in LPS-stimulated RAW264.7 cells. Productions of COX-2 and PGE2, which are key biomarkers for inflammation, were also significantly reduced. These results elucidated the immune-enhancement and anti-inflammatory mechanisms of the H. aurantium tunic fatty acids in macrophage cells. Moreover, the H. aurantium tunic might be a potential fatty acid source for immune-modulation.
Collapse
Affiliation(s)
- Chaiwat Monmai
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Korea.
| | - Seok Hyeon Go
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Korea.
| | - Ii-Shik Shin
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Korea.
| | - Sang Guan You
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Korea.
| | - Hyungjae Lee
- Department of Food Engineering, Dankook University, Cheonan, Chungnam 31116, Korea.
| | - Seok Beom Kang
- Citrus Research Station, National Institute of Horticultural and Herbal Science, RDA, Seogwipo 63607, Korea.
| | - Woo Jung Park
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon 25457, Korea.
| |
Collapse
|
32
|
Alvarado FL, Calabuig-Navarro V, Haghiac M, Puchowicz M, Tsai PJS, O'Tierney-Ginn P. Maternal obesity is not associated with placental lipid accumulation in women with high omega-3 fatty acid levels. Placenta 2018; 69:96-101. [PMID: 30213493 PMCID: PMC8439553 DOI: 10.1016/j.placenta.2018.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/10/2018] [Accepted: 07/28/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Placentas of obese women have higher lipid content compared to lean women. We have previously shown that supplementation of overweight and obese women with omega-3 fatty acids decreases placental esterification pathways and total lipid content in a mid-western population (Ohio). We hypothesized that placental lipid accumulation and inflammation would be similar between lean and obese women living in a region of high omega-3 intake, such as Hawaii. METHODS Fifty-five healthy, normal glucose tolerant women from Honolulu Hawaii, dichotomized based on pre-pregnancy BMI into lean (BMI <25 kg/m2, n = 29) and obese (BMI >30 kg/m2, n = 26), were recruited at scheduled term cesarean delivery. Maternal plasma DHA levels were analyzed by mass spectrometry. Expression of key genes involved in fatty acid oxidation and esterification were measured in placental tissue using qPCR. Total lipids were extracted from placental tissue via the Folch method. TNF-α concentration was measured by enzyme-linked immunosorbent assay in placental lysates. RESULTS DHA levels were higher in lean women compared to obese women (P = 0.02). However, DHA levels in obese women in Hawaii were eight times higher compared to obese Ohioan women (P=<0.0001). Placental lipid content and expression of key genes involved in fatty acid oxidation and esterification were similar (P > 0.05) between lean and obese women in Hawaii. Furthermore, TNF-α placental lysates were not different between lean and obese women. CONCLUSIONS Though obese women in Hawaii have lower DHA levels compared to their lean counterparts, these levels remain over eight times as high as obese Ohioan women. These relatively high plasma omega-3 levels in obese women in Hawaii may suppress placental lipid esterification/storage and inflammation to the same levels of lean women, as seen previously in vitro.
Collapse
Affiliation(s)
- Fernanda L Alvarado
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, OH, United States
| | - Virtu Calabuig-Navarro
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, OH, United States; Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Maricela Haghiac
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, OH, United States
| | - Michelle Puchowicz
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Pai-Jong S Tsai
- Department of Obstetrics, Gynecology, and Women's Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Perrie O'Tierney-Ginn
- Center for Reproductive Health, MetroHealth Medical Center, Cleveland, OH, United States; Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
33
|
Jiang WJ, Peng YC, Yang KM. Cellular signaling pathways regulating β-cell proliferation as a promising therapeutic target in the treatment of diabetes. Exp Ther Med 2018; 16:3275-3285. [PMID: 30233674 PMCID: PMC6143874 DOI: 10.3892/etm.2018.6603] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 07/27/2018] [Indexed: 12/30/2022] Open
Abstract
It is established that a decrease in β-cell number and deficiency in the function of existing β-cells contribute to type 1 and type 2 diabetes mellitus. Therefore, a major focus of current research is to identify novel methods of improving the number and function of β-cells, so as to prevent and/or postpone the development of diabetes mellitus and potentially reverse diabetes mellitus. Based on prior knowledge of the above-mentioned causes, promising therapeutic approaches may include direct transplantation of islets, implantation and subsequent induced differentiation of progenitors/stem cells to β-cells, replication of pre-existing β-cells, or activation of endogenous β-cell progenitors. More recently, with regards to cell replacement and regenerative treatment for diabetes patients, the identification of cellular signaling pathways with related genes or corresponding proteins involved in diabetes has become a topic of interest. However, the majority of pathways and molecules associated with β-cells remain unresolved, and the specialized functions of known pathways remain unclear, particularly in humans. The current article has evaluated the progress of research on pivotal cellular signaling pathways involved with β-cell proliferation and survival, and their validity for therapeutic adult β-cell regeneration in diabetes. More efforts are required to elucidate the cellular events involved in human β-cell proliferation in terms of the underlying mechanisms and functions.
Collapse
Affiliation(s)
- Wen-Juan Jiang
- Institute of Anatomy, Basic Medical College of Dali University, Dali, Yunnan 671000, P.R. China
| | - Yun-Chuan Peng
- Institute of Anatomy, Basic Medical College of Dali University, Dali, Yunnan 671000, P.R. China
| | - Kai-Ming Yang
- Institute of Anatomy, Basic Medical College of Dali University, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
34
|
Gao Y, Zheng SC, Zheng CQ, Shi YC, Xie XL, Wang KJ, Liu HP. The immune-related fatty acids are responsive to CO 2 driven seawater acidification in a crustacean brine shrimp Artemia sinica. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 81:342-347. [PMID: 29288063 DOI: 10.1016/j.dci.2017.12.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 06/07/2023]
Abstract
The gradual increase of CO2 concentration in the atmosphere, absorbed by the ocean surface water through air to sea equilibration termed ocean acidification (OA), leads to the decline of pH in seawater. It is not clear so far how the composition of fatty acids, particular the immune-related, in marine crustacean and the subsequent energy supply in marine ecosystem are affected by OA. The brine shrimp Artemia sinica is an open and common feed that provide essential fatty acids for mariculture. In this study, the fatty acids profiles of brine shrimp cultured under different lower pH levels of CO2 driven seawater were investigated. The results showed a significant reduction of the proportion of total saturated fatty acids under the pH7.6 within one week. Meanwhile, the percentage of total monounsaturated fatty acids was significantly decreased at day 14 under pH7.8, and this percentage gave a significant increase of proportion within one week under pH7.6. Furthermore, the relative content of total polyunsaturated fatty acids (PUFAs) was found to be clearly increased with exposure to different seawater acidification at day 1, suggesting that the brine shrimp immune response was likely to be affected by acidified seawater as the PUFAs have been well known to be involved in immunomodulatory effects through alterations on cell membrane fluidity/lipid mediators and gene expression of cell signaling pathways. Notably, eicosapentaenoic acid and docosahexaenoic acid, which have essential effect on various physiological processes such as inflammatory cytokines production and cell structural stability, were strongly increased under two lower pH treatments within one week and with the significant increase at day 1 under pH7.6. These data clearly supported the hypothesis that OA might affect fatty acids composition, likely also the innate immunity, in crustacean and the subsequent energy transfer by food-chain system in the marine ecosystem.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, Fujian, PR China
| | - Shu-Cheng Zheng
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, Fujian, PR China
| | - Chao-Qun Zheng
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, Fujian, PR China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Yue-Chen Shi
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, Fujian, PR China
| | - Xiao-Lu Xie
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, Fujian, PR China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, Fujian, PR China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen 361102, Fujian, PR China
| | - Hai-Peng Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, Fujian, PR China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen 361102, Fujian, PR China.
| |
Collapse
|
35
|
Association between Maternal and Foetal Erythrocyte Fatty Acid Profiles and Birth Weight. Nutrients 2018; 10:nu10040402. [PMID: 29570689 PMCID: PMC5946187 DOI: 10.3390/nu10040402] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/13/2018] [Accepted: 03/20/2018] [Indexed: 11/16/2022] Open
Abstract
Regular foetal development is crucial for assuring good health status in the offspring. The quality and quantity of maternal dietary fatty acids (FAs) can affect growth. The study aimed to: (1) investigate the association of maternal/foetal lipid profiles with birth weight (BW); and (2) compare these profiles in small, appropriate, and large for gestational age (SGA, AGA, and LGA) infants. FAs were measured in erythrocyte membranes using gas chromatography analysis in 607 mother–infant pairs (316 males, 52.1%). In the quantile regression, a significant association between BW and levels of maternal linoleic acid (LA; C18:2, n-6; coefficient: 18.66; p = 0.010), arachidonic acid (AA; C20:4, n-6; coefficient: 11.35; p = 0.007), docosahexaenoic acid (DHA; C22:6, n-3; coefficient: 29.73; p = 0.007), polyunsaturated FAs (coefficient: 8.55; p = 0.001), foetal DHA (coefficient: −22.82; p = 0.037), and saturated FAs (coefficient: −65.41; p = 0.002) was found. Myristic (C14:0) and pentadecanoic acids (C15:0), both maternal (p = 0.000; p = 0.017) and foetal (p = 0.009; p = 0.002), and maternal erucic acid (C22:1, n-9; p = 0.026) were found at higher levels in SGA infants as compared to AGA ones. Conversely, maternal LA, AA, and omega 6 FAs levels were higher in AGA infants (p = 0.037; p = 0.003; p = 0.026, respectively). Maternal and foetal polyunsaturated and omega 6 FAs levels are positively related to BW, while a lipid profile rich in saturated FAs and erucic acid may influence the risk of SGA.
Collapse
|
36
|
Toll-Like Receptors: Regulators of the Immune Response in the Human Gut. Nutrients 2018; 10:nu10020203. [PMID: 29438282 PMCID: PMC5852779 DOI: 10.3390/nu10020203] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/28/2022] Open
Abstract
Toll-like receptors (TLRs) are powerful molecular regulators by which the immune system may "sense" the environment and protect the host from pathogens or endogenous threats. In mammalian cells, several TLRs were identified with a tissue and cell type-specific distribution. Understanding the functions of specific TLRs is crucial for the development and discovery of compounds useful to maintaining or re-establishing homeostasis in the gastrointestinal tract (GIT). Due to their relevance in regulating the inflammatory response in the GIT, we will focus here on TLR2, TLR4, and TLR5. In particular, we describe (a) the molecular pathways activated by the stimulation of these receptors with their known bacterial ligands; (b) the non-bacterial ligands known to interact directly with TLR2 and TLR4 and their soluble forms. The scope of this minireview is to highlight the importance of bacterial and non-bacterial compounds in affecting the gut immune functions via the activation of the TLRs.
Collapse
|
37
|
Yang X, Glazebrook P, Ranasinghe GC, Haghiac M, Calabuig-Navarro V, Minium J, O'Tierney-Ginn P. Fatty acid transporter expression and regulation is impaired in placental macrovascular endothelial cells in obese women. J Matern Fetal Neonatal Med 2017; 32:971-978. [PMID: 29065800 DOI: 10.1080/14767058.2017.1397119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Fetal fatty acid (FA) delivery is ultimately controlled by placental transport. Focus has been the maternal-placental interface, but regulation at the feto-placental interface is unknown. METHODS Placental macrovascular endothelial cells (EC) (n = 4/group) and trophoblasts (TB) (n = 5/group) were isolated from lean (pregravid BMI <25 kg/m2) and obese (body mass index (BMI) > 30) women. Fatty acid transporters FAT/CD36, FABPpm, FATP4, FABP 3, 4 and 5, PLIN2 and PPARα, δ, γ expression, was measured in EC and TB. Transporter response to 24 h palmitate (PA) was assessed. RESULTS mRNA expression of FABP3, 4, 5 and PPARγ was 2- to 3-fold reduced in EC of obese versus lean women (p < .03), but not in TB. Protein level of FABPpm was 20% lower in obese (p < .05). Palmitate (PA) up-regulated CD36, FABP3, FABP4, and PLIN2 gene expression by 3- to 4-fold in lean but not obese EC (p < .05), while PA increased FABP4 and PLIN2 in lean and obese TB, and FABP5 in lean (p < .05) EC. PA exposure up-regulated peroxisome proliferator activated receptors (PPARs) 2-fold in lean and obese EC (p < .05), but not in TB. CONCLUSIONS In obese women, FA transporter expression is lower in placental EC, but not TB, and less sensitive to saturated FA, compared to lean women. FA transport may be regulated at the feto-placental interface.
Collapse
Affiliation(s)
- Xiaohua Yang
- a Center for Reproductive Health, Metrohealth Medical Center , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Patricia Glazebrook
- a Center for Reproductive Health, Metrohealth Medical Center , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Geraldine C Ranasinghe
- a Center for Reproductive Health, Metrohealth Medical Center , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Maricela Haghiac
- a Center for Reproductive Health, Metrohealth Medical Center , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Virtu Calabuig-Navarro
- a Center for Reproductive Health, Metrohealth Medical Center , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Judi Minium
- a Center for Reproductive Health, Metrohealth Medical Center , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| | - Perrie O'Tierney-Ginn
- a Center for Reproductive Health, Metrohealth Medical Center , Case Western Reserve University School of Medicine , Cleveland , OH , USA
| |
Collapse
|
38
|
Statovci D, Aguilera M, MacSharry J, Melgar S. The Impact of Western Diet and Nutrients on the Microbiota and Immune Response at Mucosal Interfaces. Front Immunol 2017; 8:838. [PMID: 28804483 PMCID: PMC5532387 DOI: 10.3389/fimmu.2017.00838] [Citation(s) in RCA: 300] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/03/2017] [Indexed: 12/11/2022] Open
Abstract
Recent findings point toward diet having a major impact on human health. Diets can either affect the gut microbiota resulting in alterations in the host’s physiological responses or by directly targeting the host response. The microbial community in the mammalian gut is a complex and dynamic system crucial for the development and maturation of both systemic and mucosal immune responses. Therefore, the complex interaction between available nutrients, the microbiota, and the immune system are central regulators in maintaining homeostasis and fighting against invading pathogens at mucosal sites. Westernized diet, defined as high dietary intake of saturated fats and sucrose and low intake of fiber, represent a growing health risk contributing to the increased occurrence of metabolic diseases, e.g., diabetes and obesity in countries adapting a westernized lifestyle. Inflammatory bowel diseases (IBD) and asthma are chronic mucosal inflammatory conditions of unknown etiology with increasing prevalence worldwide. These conditions have a multifactorial etiology including genetic factors, environmental factors, and dysregulated immune responses. Their increased prevalence cannot solely be attributed to genetic considerations implying that other factors such as diet can be a major contributor. Recent reports indicate that the gut microbiota and modifications thereof, due to a consumption of a diet high in saturated fats and low in fibers, can trigger factors regulating the development and/or progression of both conditions. While asthma is a disease of the airways, increasing evidence indicates a link between the gut and airways in disease development. Herein, we provide a comprehensive review on the impact of westernized diet and associated nutrients on immune cell responses and the microbiota and how these can influence the pathology of IBD and asthma.
Collapse
Affiliation(s)
- Donjete Statovci
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Mònica Aguilera
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - John MacSharry
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Silvia Melgar
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
39
|
Montserrat-de la Paz S, Rodriguez D, Cardelo MP, Naranjo MC, Bermudez B, Abia R, Muriana FJ, Lopez S. The effects of exogenous fatty acids and niacin on human monocyte-macrophage plasticity. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600824] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/09/2017] [Accepted: 02/15/2017] [Indexed: 12/24/2022]
Affiliation(s)
| | - Dolores Rodriguez
- Laboratory of Cellular and Molecular Nutrition; Instituto de la Grasa; Seville Spain
| | - Magdalena P. Cardelo
- Laboratory of Cellular and Molecular Nutrition; Instituto de la Grasa; Seville Spain
| | - Maria C. Naranjo
- Laboratory of Cellular and Molecular Nutrition; Instituto de la Grasa; Seville Spain
| | - Beatriz Bermudez
- Department of Cell Biology; School of Biology; University of Seville; Seville Spain
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition; Instituto de la Grasa; Seville Spain
| | | | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition; Instituto de la Grasa; Seville Spain
| |
Collapse
|
40
|
Zhang Z, Ma Z. Saturated fatty acids recognition by the CD14-TLR4-MD2 complex may engage in the presurgical anxiety-induced persistent postsurgical pain. Med Hypotheses 2017; 103:105-107. [PMID: 28571793 DOI: 10.1016/j.mehy.2017.03.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 01/07/2017] [Accepted: 03/11/2017] [Indexed: 12/13/2022]
Abstract
It has been proved that presurgical anxiety can induce the development and progression of persistent postsurgical pain through elevating circulating corticosterone levels and activating the glucocorticoids receptor. Under stressful circumstances, the concentration of blood saturated fatty acids (SFAs) increases rapidly to provide enough ATP for individuals' survival owing to stress hormones such as glucocorticoid, catecholamine and glucagon. It is reported that SFAs can trigger an inflammatory response through CD14-TLR4-MD2 complex. The role of TLR4 and its downstream signaling pathway has been confirmed in the pathogenesis of cancer pain and inflammatory pain. Furthermore, a newly clinical research uncovers that the children with recurrent abdominal pain, which is triggered by early psychosomatic stress, have higher levels of SFAs than healthy individuals. We therefore put forward the hypothesis that the elevated level of SFAs induced by stress hormones may be engaged in the presurgical anxiety-induced persistent postsurgical pain. If established, it's of important clinical significance, which will make great contributions to the prevention and treatment of the presurgical anxiety-induced persistent postsurgical pain.
Collapse
Affiliation(s)
- Zuoxia Zhang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Jiangsu, China
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Jiangsu, China.
| |
Collapse
|
41
|
TLR4 knockout attenuated high fat diet-induced cardiac dysfunction via NF-κB/JNK-dependent activation of autophagy. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2001-2011. [PMID: 28108421 DOI: 10.1016/j.bbadis.2017.01.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 12/19/2022]
Abstract
Obesity is commonly associated with a low grade systemic inflammation, which may contribute to the onset and development of myocardial remodeling and contractile dysfunction. Toll-like receptor 4 (TLR4) plays an important role in innate immunity and inflammation although its role in high fat diet-induced obesity cardiac dysfunction remains elusive. This study was designed to examine the effect of TLR4 ablation on high fat diet intake-induced cardiac anomalies, if any, and underlying mechanism(s) involved. Wild-type (WT) and TLR4 knockout mice were fed normal or high fat (60% calorie from fat) diet for 12weeks prior to assessment of mechanical and intracellular Ca2+ properties. The inflammatory signaling proteins (TLR4, NF-κB, and JNK) and autophagic markers (Atg5, Atg12, LC3B and p62) were evaluated. Our results revealed that high fat diet intake promoted obesity, marked decrease in fractional shortening, and cardiomyocyte contractile capacity with dampened intracellular Ca2+ release and clearance, elevated ROS generation and oxidative stress as measured by aconitase activity, the effects of which were significantly attenuated by TLR4 knockout. In addition, high fat intake downregulated levels of Atg5, Atg12 and LC3B, while increasing p62 accumulation. TLR4 knockout itself did not affect Atg5, Atg12, LC3B and p62 levels while it reconciled high fat diet intake-induced changes in autophagy. In addition, TLR4 knockout alleviated high fat diet-induced phosphorylation of IKKβ, JNK and mTOR. In vitro study revealed that palmitic acid suppressed cardiomyocyte contractile function, the effect of which was inhibited the TLR4 inhibitor CLI-095, the JNK inhibitor AS601245 or the NF-κB inhibitor Celastrol. Taken together, these data showed that TLR4 knockout ameliorated high fat diet-induced cardiac contractile and intracellular Ca2+ anomalies through inhibition of inflammation and ROS, possibly through a NF-κB/JNK-dependent activation of autophagy. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
|
42
|
Zuo H, Gao J, Yuan J, Deng H, Yang L, Weng S, He J, Xu X. Fatty acid synthase plays a positive role in shrimp immune responses against Vibrio parahaemolyticus infection. FISH & SHELLFISH IMMUNOLOGY 2017; 60:282-288. [PMID: 27903451 DOI: 10.1016/j.fsi.2016.11.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/08/2016] [Accepted: 11/25/2016] [Indexed: 06/06/2023]
Abstract
Fatty acid synthase (FAS) is an important enzyme that catalyzes the synthesis of fatty acids. In this study, the role of the FAS gene from pacific white shrimp Litopenaeus vannamei (LvFAS) in immune responses against Vibrio parahaemolyticus infection was studied. The expression of LvFAS could be up-regulated upon infection of V. parahaemolyticus and stimulation of lipopolysaccharide and poly (I:C). The promoter of LvFAS was predicted to harbor a NF-κB binding site and dual-luciferase reporter assays demonstrated that the NF-κB family proteins Relish, sRelish and Dorsal could activate the transcription of LvFAS. After knockdown of LvFAS expression using RNAi strategy, both the mortality of V. parahaemolyticus infected shrimps and the bacterial load in shrimp tissues were significantly increased. Meanwhile, the expression of many immune-responsive genes, such as antimicrobial peptides, C-type lectins (CTLs), lysozyme and hemolin, was down-regulated. These suggested that LvFAS could play a positive role in anti-V. parahaemolyticus responses in shrimp. To our knowledge, this is the first study that investigates the role of FAS in antibacterial immunity in animals, which may indicate the relationship between the anabolism of fatty acids and immune responses in crustaceans.
Collapse
Affiliation(s)
- Hongliang Zuo
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou, PR China
| | - Jiefeng Gao
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Jia Yuan
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Hengwei Deng
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Linwei Yang
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Jianguo He
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou, PR China.
| | - Xiaopeng Xu
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou, PR China.
| |
Collapse
|
43
|
Reynés B, Palou M, Palou A. Gene expression modulation of lipid and central energetic metabolism related genes by high-fat diet intake in the main homeostatic tissues. Food Funct 2017; 8:629-650. [DOI: 10.1039/c6fo01473a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
HF diet feeding affects the energy balance by transcriptional metabolic adaptations, based in direct gene expression modulation, perinatal programing and transcriptional factor regulation, which could be affected by the animal model, gender or period of dietary treatment.
Collapse
Affiliation(s)
- Bàrbara Reynés
- Laboratory of Molecular Biology
- Nutrition and Biotechnology
- Universitat de les Illes Balears and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn)
- Palma de Mallorca
- Spain
| | - Mariona Palou
- Alimentómica SL (Spin off no. 001 from UIB)
- Palma Mallorca
- Spain
| | - Andreu Palou
- Laboratory of Molecular Biology
- Nutrition and Biotechnology
- Universitat de les Illes Balears and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn)
- Palma de Mallorca
- Spain
| |
Collapse
|
44
|
Yang X, Li M, Haghiac M, Catalano PM, O'Tierney-Ginn P, Hauguel-de Mouzon S. Causal relationship between obesity-related traits and TLR4-driven responses at the maternal-fetal interface. Diabetologia 2016; 59:2459-2466. [PMID: 27535280 PMCID: PMC5583648 DOI: 10.1007/s00125-016-4073-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/18/2016] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS Obesity triggers complex inflammatory networks within the innate immune system. During pregnancy, the placenta amplifies the low-grade inflammation through activation of Toll-like receptor 4 (TLR4) signalling pathways. The purpose of this study was to investigate the impact of obesity on placental TLR4 expression and inflammatory signals. The secondary aim was to analyse the placental cell type responsible for TLR4 activation. METHODS Thirty-nine women recruited at term-scheduled Caesarean section were grouped according to their pre-gravid BMI (<25 kg/m(2) and >30 kg/m(2)). Placenta, venous maternal and cord blood were obtained at delivery for analysis. Data were analysed with linear regression and Spearman's rank correlation coefficient analysis. RESULTS TLR4, IL6 and IL8 expression was increased three- to ninefold (p < 0.001) in the placenta of obese vs lean women. There was a positive correlation between placental TLR4 and maternal systemic and placental IL6 and IL8 concentrations. Placental TLR4 expression was correlated with maternal pre-gravid BMI, insulin resistance index, plasma insulin and C-reactive protein (r = 0.57, 0.31, 0.35, 0.53, respectively; p < 0.001) but not with plasma glucose, maternal age, gestational age and gestational weight gain (r < 0.2; p > 0.1). TLR4 was located in both trophoblast and macrovascular endothelial cells lining fetal vasculature. Lipopolysaccharide-induced TLR4 activation was more robust in trophoblasts than in endothelial vascular cells (100-fold vs tenfold; p < 0.001). CONCLUSIONS/INTERPRETATION Trophoblastic TLR4 is strongly implicated in the propagation of placental inflammation. Placental inflammation is related to maternal metabolic conditions such as pre-gravid BMI, whilst gestational weight gain or gestational age are not. These results implicate the pre-gravid condition as a significant contributor to metabolic inflammation in late pregnancy.
Collapse
Affiliation(s)
- Xiaohua Yang
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Ming Li
- Epidemiology and Biostatistics Department, School of Medicine, Case Western Reserve University, Cleveland, USA
| | - Maricela Haghiac
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Patrick M Catalano
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Perrie O'Tierney-Ginn
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Sylvie Hauguel-de Mouzon
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA.
| |
Collapse
|
45
|
Maternal obesity modulates intracellular lipid turnover in the human term placenta. Int J Obes (Lond) 2016; 41:317-323. [PMID: 27780978 PMCID: PMC5309341 DOI: 10.1038/ijo.2016.188] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/22/2016] [Accepted: 09/25/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Obesity before pregnancy is associated with impaired metabolic status of the mother and the offspring later in life. These adverse effects have been attributed to epigenetic changes in utero, but little is known about the role of placental metabolism and its contribution to fetal development. OBJECTIVES We examined the impact of maternal pre-pregnancy obesity on the expression of genes involved in placental lipid metabolism in lean and obese women. SUBJECTS/METHODS Seventy-three lean and obese women with healthy pregnancy were recruited at term elective cesarean delivery. Metabolic parameters were measured on maternal venous blood samples. Expression of 88 genes involved in lipid metabolism was measured in whole placenta tissue. Proteins of genes differently expressed in response to maternal obesity were quantified, correlated with maternal parameters and immunolocalized in placenta sections. Isolated primary trophoblasts were used for in vitro assays. RESULTS Triglyceride (TG) content was increased in placental tissue of obese (1.10, CI 1.04-1.24 mg g-1, P<0.05) vs lean (0.84, CI 0.72-1.02 mg g-1) women. Among target genes examined, six showed positive correlation (P<0.05) with maternal pre-pregnancy BMI, namely ATGL (PNPLA2), FATP1 (SLC27A1), FATP3 (SLC27A3), PLIN2, PPARG and CGI-58 (ABHD5). CGI-58 protein abundance was twofold higher (P<0.001) in placentas of obese vs lean women. CGI-58 protein levels correlated positively with maternal insulin levels and pre-pregnancy body mass index (R=0.63, P<0.001 and R=0.64, P<0.001, respectively). CGI-58 and PLIN2 were primarily located in the syncytiotrophoblast and, were upregulated (1.38- and 500-fold, respectively) upon oleic acid and insulin treatment of cultured trophoblast cells. CONCLUSION Pre-gravid obesity significantly modifies the expression of placental genes related to transport and storage of neutral lipids. We propose that the upregulation of CGI-58, a master regulator of TG hydrolysis, contributes to the turnover of intracellular lipids in placenta of obese women, and is tightly regulated by metabolic factors of the mother.
Collapse
|
46
|
Liu B, Sun R, Luo H, Liu X, Jiang M, Yuan C, Yang L, Hu J. Both intrinsic and extrinsic apoptotic pathways are involved in Toll-like receptor 4 (TLR4)-induced cell death in monocytic THP-1 cells. Immunobiology 2016; 222:198-205. [PMID: 27720227 DOI: 10.1016/j.imbio.2016.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 09/02/2016] [Accepted: 10/03/2016] [Indexed: 01/17/2023]
Abstract
Our previous study showed that TLR3 induces apoptosis via both death receptors and mitochondial in human endothelial cells. We report here that the activation of TLR4 induced dose- and time-dependent cell death in moncytic THP-1 cells. LPS treatment of THP-1 cells induced the activation of both caspase 8 and 9, suggesting the involvement of intrinsic and extrinsic apoptosis pathways. TNFα was induced by TLR4 activation at both mRNA and protein levels, but its neutralization did not down-regulated TLR4-induced cell death. TLR4 activation also induced the up-regulation of TRAIL and its receptors DR4 and DR5, and the neutralization of TRAIL ameliorated TLR4 induced apoptosis, suggesting the involvement of TRAIL and its receptors DR4 and DR5 in LPS-induced cell death. Meanwhile, LPS treatment down-regulated the expression of FLICE inhibitory protein (FLIP), a suppressor of death receptor-induced cell death. In addition, TLR4 activation down-regulated the anti-apoptotic protein bcl-2, and up-regulated the pro-apoptotic proteins Noxa and Puma, suggesting that mitochondrial apoptotic pathway was also involved in LPS-induced cell death. Furthermore, we found that TAP63α might confer to the activation of intrinsic and extrinsic apoptotic pathways. The treatment of THP-1 cells with LPS induced the translocation of TAP63α from cytoplasm to nucleus. Taken together, our study suggested that both death receptors and mitochondial were involved in TLR4-induced cell death, and TAP63α may be a target for the prevention of LPS-induced cell death.
Collapse
Affiliation(s)
- Bei Liu
- Medical Research Center, Changsha Central Hospital, Changsha, China; Department of Pathology, Affiliated Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Ruili Sun
- Department of Laboratory Medicine, XinXiang Medical University, XinXiang, China
| | - Hongbo Luo
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Chuang Yuan
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Li Yang
- Tuberculosis Research Center, Changsha Central Hospital, Changsha, China.
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, China.
| |
Collapse
|
47
|
Wang S, Xiang N, Yang L, Zhu C, Zhu X, Wang L, Gao P, Xi Q, Zhang Y, Shu G, Jiang Q. Linoleic acid and stearic acid elicit opposite effects on AgRP expression and secretion via TLR4-dependent signaling pathways in immortalized hypothalamic N38 cells. Biochem Biophys Res Commun 2016; 471:566-71. [PMID: 26879142 DOI: 10.1016/j.bbrc.2016.02.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/09/2016] [Indexed: 12/28/2022]
|