1
|
Schmidt L, Garscha U. Species-specific optimization of oxylipin ionization in LC-MS: a design of experiments approach to improve sensitivity. Anal Bioanal Chem 2025; 417:1807-1818. [PMID: 39891659 PMCID: PMC11913994 DOI: 10.1007/s00216-025-05759-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Oxylipins are diverse bioactive signaling molecules, which occur in very low concentrations in complex matrices, posing challenges in achieving consistent and sensitive analysis. UHPLC-MS/MS is the preferred technique to separate and quantify these molecules, often optimized using a time-consuming trial-and-error approach. In this study, we applied the design of experiments (DoE) approach to systematically investigate the ionization properties of multiple oxylipin species. Fractional factorial and central composite designs were employed to detect relevant instrument parameters and optimize signal intensity in ESI-MS/MS analysis. Response surface modeling revealed distinct ionization and fragmentation behaviors between polar and apolar oxylipins, driven by their responses to interface temperature and collision-induced dissociation (CID) gas pressure. Particularly, prostaglandins and lipoxins benefit from higher CID gas pressure and lower temperatures compared to the lipophilic HODEs and HETEs to achieve optimal intensity in multiple reaction monitoring analysis. While global source parameters were optimized, analyte-specific entrance/exit potentials and collision energies required individual adjustments. The final method was applied to analyze seven oxylipin classes including leukotrienes, prostaglandins, lipoxins, resolvins, HETEs, HODES, and HoTrEs. Although improvements in lower limits of quantification were modest (< 1 pg on-column), signal-to-noise ratios increased two-fold for lipoxins and resolvins and three- to four-fold for leukotrienes and HETEs, enhancing detection at trace levels. This DoE-guided strategy provides a powerful tool to improve UHPLC-MS/MS analysis of oxylipins across various instrument vendors, guiding the way towards inter-laboratory comparability.
Collapse
Affiliation(s)
- Louis Schmidt
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Greifswald University, 17489, Greifswald, Germany
| | - Ulrike Garscha
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Greifswald University, 17489, Greifswald, Germany.
| |
Collapse
|
2
|
Fang M, Xia F, Teng B, Xia W, Yang Y, Wang J, Tao C, Hu X. RvE1/ChemR23 facilitates hematoma clearance and promotes M2 polarization of macrophages/microglia in intracerebral hemorrhage. Exp Neurol 2025; 386:115173. [PMID: 39892453 DOI: 10.1016/j.expneurol.2025.115173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
INTRODUCTION Previous studies have demonstrated the potent anti-inflammatory effects of RvE1 in various diseases, and recent research has shown that it can also promote macrophage phagocytosis. Given that hematoma clearance is crucial for intracerebral hemorrhage (ICH) treatment, while neuroinflammation significantly influences secondary injury, we hypothesize that RvE1/ChemR23 activation, by modulating the polarization of macrophages/microglia, promotes hematoma resolution and alleviates neuroinflammatory responses after ICH. METHOD A total of 125 WT C57BL/6 and 67 ChemR23-/- male mice were used. Western blot and immunofluorescence staining assessed the temporal and spatial expression of ChemR23 after ICH. T2WI, T2*WI and behavioral tests were obtained to assess the protective effect of the RvE1/ChemR23 pathway in ICH. Additionally, co-staining of M1 (iNOS) or M2 polarization (Arg-1) markers with Iba-1 was used to explore the polarization status of macrophages/microglia in the perihematomal region. Finally, Akt phosphorylation was validated as a downstream mediator of the RvE1/ChemR23 pathway using an Akt inhibitor. RESULTS ChemR23 is mainly expressed in activated microglia and infiltrating macrophages, with expression peaking 5-7 days post-ICH. Activation of the RvE1/ChemR23 pathway promotes hematoma resolution, reduces brain edema, and improves neurological deficits in ICH. These effects are likely mediated by promoting M2 polarization of macrophages/microglia after ICH. Furthermore, the use of an Akt inhibitor can counteract the protective effects of RvE1 in ICH. CONCLUSIONS Our study provides the first evidence of the protective role of RvE1/ChemR23 signaling in ICH. This pathway might offer novel therapeutic targets for the clinical management of ICH.
Collapse
Affiliation(s)
- Mei Fang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bang Teng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanting Xia
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Yunfei Yang
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Jiayan Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chuanyuan Tao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xin Hu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Wang T, Chen S, Zhou D, Hong Z. Exploring receptors for pro-resolving and non-pro-resolving mediators as therapeutic targets for sarcopenia. Metabolism 2025; 165:156148. [PMID: 39892864 DOI: 10.1016/j.metabol.2025.156148] [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: 09/26/2024] [Revised: 01/01/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Sarcopenia is defined by a reduction in both muscle strength and mass. Sarcopenia may be an inevitable component of the aging process, but it may also be accelerated by comorbidities and metabolic derangements. The underlying mechanisms contributing to these pathological changes remain poorly understood. We propose that chronic inflammation-mediated networks and metabolic defects that exacerbate muscle dysfunction are critical factors in sarcopenia and related diseases. Consequently, utilizing specialized pro-resolving mediators (SPMs) that function through specific G-protein coupled receptors (GPCRs) may offer effective therapeutic options for these disorders. However, challenges such as a limited understanding of SPM/receptor signaling pathways, rapid inactivation of SPMs, and the complexities of SPM synthesis impede their practical application. In this context, stable small-molecule SPM mimetics and receptor agonists present promising alternatives. Moreover, the aged adipose-skeletal axis may contribute to this process. Activating non-SPM GPCRs on adipocytes, immune cells, and muscle cells under conditions of systemic, chronic, low-grade inflammation (SCLGI) could help alleviate inflammation and metabolic dysfunction. Recent preclinical studies indicate that both SPM GPCRs and non-SPM GPCRs can mitigate symptoms of aging-related diseases such as obesity and diabetes, which are driven by chronic inflammation and metabolic disturbances. These findings suggest that targeting these receptors could provide a novel strategy for addressing various chronic inflammatory conditions, including sarcopenia.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Neurology, Institute of Neurology and Disease, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Sihan Chen
- West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Dong Zhou
- Department of Neurology, Institute of Neurology and Disease, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, Institute of Neurology and Disease, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Jomova K, Alomar SY, Valko R, Liska J, Nepovimova E, Kuca K, Valko M. Flavonoids and their role in oxidative stress, inflammation, and human diseases. Chem Biol Interact 2025:111489. [PMID: 40147618 DOI: 10.1016/j.cbi.2025.111489] [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: 06/02/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Oxidative stress and chronic inflammation are important drivers in the pathogenesis and progression of many chronic diseases, such as cancers of the breast, kidney, lung, and others, autoimmune diseases (rheumatoid arthritis), cardiovascular diseases (hypertension, atherosclerosis, arrhythmia), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease), mental disorders (depression, schizophrenia, bipolar disorder), gastrointestinal disorders (inflammatory bowel disease, colorectal cancer), and other disorders. With the increasing demand for less toxic and more tolerable therapies, flavonoids have the potential to effectively modulate the responsiveness to conventional therapy and radiotherapy. Flavonoids are polyphenolic compounds found in fruits, vegetables, grains, and plant-derived beverages. Six of the twelve structurally different flavonoid subgroups are of dietary significance and include anthocyanidins (e.g. pelargonidin, cyanidin), flavan-3-ols (e.g. epicatechin, epigallocatechin), flavonols (e.g. quercetin, kaempferol), flavones (e.g. luteolin, baicalein), flavanones (e.g. hesperetin, naringenin), and isoflavones (daidzein, genistein). The health benefits of flavonoids are related to their structural characteristics, such as the number and position of hydroxyl groups and the presence of C2=C3 double bonds, which predetermine their ability to chelate metal ions, terminate ROS (e.g. hydroxyl radicals formed by the Fenton reaction), and interact with biological targets to trigger a biological response. Based on these structural characteristics, flavonoids can exert both antioxidant or prooxidant properties, modulate the activity of ROS-scavenging enzymes and the expression and activation of proinflammatory cytokines (e.g., interleukin-1beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α)), induce apoptosis and autophagy, and target key signaling pathways, such as the nuclear factor erythroid 2-related factor 2 (Nrf2) and Bcl-2 family of proteins. This review aims to briefly discuss the mutually interconnected aspects of oxidative and inflammatory mechanisms, such as lipid peroxidation, protein oxidation, DNA damage, and the mechanism and resolution of inflammation. The major part of this article discusses the role of flavonoids in alleviating oxidative stress and inflammation, two common components of many human diseases. The results of epidemiological studies on flavonoids are also presented.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University in Nitra, Nitra, 949 74 Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Richard Valko
- Zoology Department, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Jan Liska
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic; Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, 708 00, Ostrava-Poruba, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, 5005 Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37 Bratislava, Slovakia.
| |
Collapse
|
5
|
Zhang Q, Wang Y, Zhu J, Zou M, Zhang Y, Wu H, Jin T. Specialized pro-resolving lipid mediators: a key player in resolving inflammation in autoimmune diseases. Sci Bull (Beijing) 2025; 70:778-794. [PMID: 39837719 DOI: 10.1016/j.scib.2024.07.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/21/2024] [Accepted: 07/16/2024] [Indexed: 01/23/2025]
Abstract
Uncontrolled hyperactivation of the immune system is the central mechanism underlying the pathogenesis of autoimmune diseases. Timely control of the inflammatory response is essential to prevent inflammation progression and organ damage. Specialized pro-resolving lipid mediators (SPMs) are autacoid molecules derived from essential polyunsaturated fatty acids during acute inflammatory responses. They promote the resolution of inflammation and orchestrate endogenous reparative responses. The SPM superfamily includes lipoxins, resolvins, protectins, and maresins, as well as novel conjugates involved in tissue regeneration. Much work has been done focusing on the actions of SPMs in autoimmunity and has identified their deficiencies and therapeutic effects in autoimmune diseases. In this review, we provide a brief introduction of SPMs, summarize their effects on key cells involved in innate and adaptive immunity, and highlight their role and therapeutic potential in autoimmune diseases.
Collapse
Affiliation(s)
- Qingxiang Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Ying Wang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm 17176, Sweden
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
6
|
Nessel I, Whiley L, Dyall SC, Michael-Titus AT. A plasma lipid signature in acute human traumatic brain injury: Link with neuronal injury and inflammation markers. J Cereb Blood Flow Metab 2025; 45:443-458. [PMID: 39188133 PMCID: PMC11572080 DOI: 10.1177/0271678x241276951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 08/28/2024]
Abstract
Traumatic brain injury (TBI) leads to major membrane lipid breakdown. We investigated plasma lipids over 3 days post-TBI, to identify a signature of acute human TBI and assess its correlation with neuronal injury and inflammation. Plasma from patients with TBI (Abbreviated Injury Scale (AIS)3 - serious injury, n = 5; AIS4 - severe injury, n = 8), and controls (n = 13) was analysed for lipidomic profile, neurofilament light (NFL) and cytokines, and the omega-3 index was measured in red blood cells. A lipid signature separated TBI from controls, at 24 and 72 h. Major species driving the separation were: lysophosphatidylcholine (LPC), phosphatidylcholine (PC) and hexosylceramide (HexCer). Docosahexaenoic acid (DHA, 22:6) and LPC (0:0/22:6) decreased post-injury. NFL levels were increased at 24 and 72 h post-injury in AIS4 TBI vs. controls. Interleukin (IL-)6, IL-2 and IL-13 were elevated at 24 h in AIS4 patients vs. controls. NFL and IL-6 were negatively correlated with several lipids. The omega-3 index at admission was low in all patients (controls: 4.3 ± 1.1% and TBI: 4.0 ± 1.1%) and did not change significantly over 3 days post-injury. We have identified specific lipid changes, correlated with markers of injury and inflammation in acute TBI. These observations could inform future lipid-based therapeutic approaches.
Collapse
Affiliation(s)
- Isabell Nessel
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Luke Whiley
- Health Futures Institute, Murdoch University, Murdoch, Australia
| | - Simon C Dyall
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
7
|
Shih YRV, Tao H, Gilpin A, Lee YW, Perikamana SM, Varghese S. Specialized pro-resolving mediator Maresin 1 attenuates pain in a mouse model of osteoarthritis. Osteoarthritis Cartilage 2025; 33:341-350. [PMID: 39617202 PMCID: PMC11842212 DOI: 10.1016/j.joca.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVE We test whether the specialized pro-resolving molecule Maresin 1 (MaR1) attenuates nociceptive behaviors in mice with osteoarthritis-like pain. DESIGN Osteoarthritis (OA)-like pain behavior was induced by intra-articular injection of monosodium iodoacetate (MIA) and treated with MaR1 (N=6) or vehicle (N=5) by intraperitoneal injection 8 weeks after injury. Mice without MIA injection were used as control (N=6). Nociceptive behaviors were examined by von Frey and dynamic weight bearing measurements. Calcitonin gene-related peptide (CGRP) expression and activated macrophages in the dorsal root ganglion (DRG) were examined by immunofluorescence staining. The inflammatory profile in circulation was assessed by cytokine array. Calcium imaging was performed to assess the in vitro functional response of DRG neurons from animals with OA-like pain behavior to MaR1 with or without RAR Related Orphan Receptor A (RORA) inverse agonist SR3335. RESULTS MaR1 attenuated knee pain behavior in treated mice (N=6) compared to non-treated mice (N=5) as shown by increased paw withdrawal threshold with a mean difference of 112.2% (95% CI [49.79, 174.6], p=0.0784) at 4 h and 150.9% (95% CI [104.2, 197.5], p=0.0001) at 4 days post-MaR1 treatment, and increased weight bearing with a mean difference of 20.08% (95% CI [2.798, 37.37], p=0.0277) at 1 day post-MaR1 treatment. CGRP expression and activated macrophages were decreased in the DRG, and inflammatory cytokine levels in the circulation were attenuated. Calcium imaging showed MaR1 reduced the functional response of DRG neurons through RORA. CONCLUSIONS Our results show that MaR1 reduces OA-like pain behavior in mice and could be a potential treatment for OA pain.
Collapse
Affiliation(s)
- Yu-Ru V Shih
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Huchen Tao
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA.
| | - Anna Gilpin
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| | - Yuan-Wen Lee
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei 11031, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | | | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
8
|
Karakasis P, Theofilis P, Patoulias D, Vlachakis PK, Antoniadis AP, Fragakis N. Diabetes-Driven Atherosclerosis: Updated Mechanistic Insights and Novel Therapeutic Strategies. Int J Mol Sci 2025; 26:2196. [PMID: 40076813 PMCID: PMC11900163 DOI: 10.3390/ijms26052196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The global rise in diabetes prevalence has significantly contributed to the increasing burden of atherosclerotic cardiovascular disease (ASCVD), a leading cause of morbidity and mortality in this population. Diabetes accelerates atherosclerosis through mechanisms such as hyperglycemia, oxidative stress, chronic inflammation, and epigenetic dysregulation, leading to unstable plaques and an elevated risk of cardiovascular events. Despite advancements in controlling traditional risk factors like dyslipidemia and hypertension, a considerable residual cardiovascular risk persists, highlighting the need for innovative therapeutic approaches. Emerging treatments, including sodium-glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, epigenetic modulators, and RNA-based therapies, are showing promise in addressing the unique challenges of diabetes-associated ASCVD. Precision medicine strategies, such as nanoparticle-based drug delivery and cell-specific therapies, offer further potential for mitigating cardiovascular complications. Advances in multiomics and systems biology continue to deepen our understanding of the molecular mechanisms driving diabetes-associated atherosclerosis. This review synthesizes recent advances in understanding the pathophysiology and treatment of diabetes-related atherosclerosis, offering a roadmap for future research and precision medicine approaches to mitigate cardiovascular risk in this growing population.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Panagiotis Theofilis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (P.K.V.)
| | - Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Faculty of Medicine, School of Health Sciences Aristotle, University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Panayotis K. Vlachakis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (P.K.V.)
| | - Antonios P. Antoniadis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Nikolaos Fragakis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| |
Collapse
|
9
|
Adams L, Rasid O, Hulme H, Quon T, Burchmore R, Milling S, Goodwin RJA, Wall DM. Spatial mapping of dextran sodium sulphate-induced intestinal inflammation and its systemic effects. FASEB J 2025; 39:e70415. [PMID: 39989432 PMCID: PMC11848815 DOI: 10.1096/fj.202402780r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/22/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease, and patients frequently experience extraintestinal manifestations affecting multiple sites. Causes of systemic inflammation remain poorly understood, but molecules originating from the intestine likely play a role, with microbial and host small molecules polarizing host immune cells towards a pro- or anti-inflammatory phenotype. Using the dextran sodium sulfate (DSS) mouse model, which mimics the disrupted barrier function, microbial dysbiosis, and immune cell dysregulation of IBD, we investigated metabolomic and phenotypic changes at intestinal and systemic sites. Using spatial biology approaches, we mapped the distribution and relative abundance of molecules and cell types across a range of tissues, revealing significant changes in DSS-treated mice. Molecules identified as contributing to the statistical separation of treated from control mice were spatially localized within organs to determine their effects on cellular phenotypes through imaging mass cytometry. This spatial approach identified both intestinal and systemic molecular drivers of inflammation, including several not previously implicated in inflammation linked to IBD or the systemic effects of intestinal inflammation. Metabolic and inflammatory pathway interplay underpins systemic disease, and determining drivers at the molecular level may aid the development of new targeted therapies.
Collapse
Affiliation(s)
- Lauren Adams
- School of Infection and Immunology, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Orhan Rasid
- School of Infection and Immunology, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Heather Hulme
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&DAstraZenecaCambridgeUK
| | - Tezz Quon
- School of Infection and Immunology, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Richard Burchmore
- School of Infection and Immunology, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Simon Milling
- School of Infection and Immunology, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Richard J. A. Goodwin
- School of Infection and Immunology, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&DAstraZenecaCambridgeUK
| | - Daniel M. Wall
- School of Infection and Immunology, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
10
|
Vara-Messler M, Trevisi L, Zulato E, Ramaschi GE, Risé P, Pinna C, Indraccolo S, Sala A, Bolego C. Aspirin-triggered DHA metabolites inhibit angiogenesis. Front Pharmacol 2025; 16:1524980. [PMID: 40070577 PMCID: PMC11893558 DOI: 10.3389/fphar.2025.1524980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Background and aim Blood vessels supply oxygen, nutrients and provide gateways for immune surveillance. Since this network nourishes all tissues, vessel abnormalities contribute to many diseases, such as cancer. One of the potential targets for Docosahexaenoic Acid (DHA) in cancer is suppressing angiogenesis, a process of new blood vessel formation within tumors. In addition, aspirin (ASA) has antineoplastic effects that may be mediated, at least in part, by metabolites derived from acetylated COX-2. We aimed at determining the effect of DHA as well as its metabolites in angiogenesis, using in vitro as well as in vivo models. Methods Endothelial cell (EC) proliferation, motility and capillary-like tube formation were determined by MTT, wound healing, Boyden and Matrigel assays, respectively. In vivo angiogenesis was measured by the Matrigel sponge model in mice. The biosynthesis of proresolving lipid mediators by ECs was determined by LC-MS-MS. Results and conclusion DHA, but not arachidonic acid (AA), at concentrations consistent with those reached in blood after fish oil supplementation, decreased EC migration in a time- and concentration-dependent manner. Pretreatment with ASA modulated cell migration already after 24 h, while both DHA and ASA decreased migration at longer incubation times without affecting viability. 17-hydroxy-DHA was detected upon incubation with DHA, and increased amounts were observed upon combined treatment with DHA and ASA, an increase that was associated to a synergic effect on EC migration. 17(R)-hydroxy-DHA (17R-HDHA), the metabolite resulting from acetylated COX-2 activity of DHA, reduced EC migration in a concentration-dependent manner. DHA in the presence of ASA, as well as 17R-HDHA, also reduced EC tube formation. These results were confirmed in vivo where both 17R-HDHA or its downstream metabolite 17RResolvinD1 were able to decrease microvessels density in a Matrigel sponge model. Overall, we demonstrated that DHA in the presence of ASA-dependent acetylation of COX-2 showed increased antiangiogenic effects, possibly resulting from its conversion to its hydroxylated derivatives.
Collapse
Affiliation(s)
- M. Vara-Messler
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UC Louvain), Brussels, Belgium
| | - L. Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - E. Zulato
- Basic and Translational Oncology Unit, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - G. E. Ramaschi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - P. Risé
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - C. Pinna
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - S. Indraccolo
- Basic and Translational Oncology Unit, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - A. Sala
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - C. Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
11
|
Olkowicz M, Yu F, Alvarez JS, Ribeiro RVP, Rosales R, Xin L, Yu M, Jaroch K, Adamson MB, Bissoondath V, Billia F, Badiwala MV, Pawliszyn J. Spatiotemporal metabolic mapping of ex-situ preserved hearts subjected to dialysis by integration of bio-SPME sampling with non-targeted metabolipidomic profiling. Anal Chim Acta 2025; 1340:343581. [PMID: 39863306 DOI: 10.1016/j.aca.2024.343581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Normothermic ex situ heart perfusion (ESHP) has emerged as a valid modality for advanced cardiac allograft preservation and conditioning prior to transplantation though myocardial function declines gradually during ESHP thus limiting its potential for expanding the donor pool. Recently, the utilization of dialysis has been shown to preserve myocardial and coronary vasomotor function. Herein, we sought to determine the changes in myocardial metabolism that could support this improvement. RESULTS Male Yorkshire porcine hearts were subjected to ESHP for 8 h with or without dialysis. Alterations in metabolism were studied with an innovative in vivo solid-phase microextraction (SPME) technology coupled with global metabolite profiling at 15 min, 1.5, 4, and 8 h of perfusion. Bio-SPME sampling was performed by inserting SPME fibres coated with a PAN-based extraction phase containing mixed-mode (C8+benzenesulfonic acid) functionalities into the myocardium to a depth of their entire 8 mm coating or immersing them in the perfusate, followed by a 20-min extraction period for the analytes of interest. Dialyzed hearts demonstrated improved bioenergetics as evidenced by accelerated purine metabolism and less pronounced accumulation of intermediates of fatty acid β/ω-oxidation. Metabolic waste accumulation such as pro-inflammatory lipid mediators (e.g., leukotrienes) was mitigated thereby supporting the process of resolution of inflammation through excretion of specialized pro-resolving mediators (resolvins D1/D2, E2, protecin D1). SIGNIFICANCE Through implementing the unique analytical pipeline we demonstrated that the addition of dialysis may preserve cardiac metabolism allowing for prolonged ESHP. This strategy has the potential to facilitate high-risk donor organs' reconditioning prior to transplantation.
Collapse
Affiliation(s)
- Mariola Olkowicz
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada; Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Frank Yu
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Juglans Souto Alvarez
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada
| | - Roberto Vanin Pinto Ribeiro
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Surgery, Dalhousie University, Halifax, NS, Canada
| | - Roizar Rosales
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada
| | - Liming Xin
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada
| | - Miao Yu
- The Jackson Laboratory, JAX Genomic Medicine, Farmington, CT, USA
| | - Karol Jaroch
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada
| | - Mitchell Brady Adamson
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada
| | - Ved Bissoondath
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute (TGHRI), University Health Network, ON, Canada; Ted Roger's Center for Heart Research, University Health Network, ON, Canada; Division of Cardiology, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada
| | - Mitesh Vallabh Badiwala
- Division of Cardiovascular Surgery, Peter Munk Cardiac Center, University Health Network, Toronto, ON, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Ted Roger's Center for Heart Research, University Health Network, ON, Canada
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
12
|
Gowler PR, Arendt-Tranholm A, Turnbull J, Jha RR, Onion D, Kelly T, Kouraki A, Millns P, Gohir S, Franks S, Barrett DA, Valdes AM, Chapman V. Monocyte eukaryotic initiation factor 2 signaling differentiates 17-hydroxy-docosahexaenoic acid levels and pain. iScience 2025; 28:111862. [PMID: 39995860 PMCID: PMC11848799 DOI: 10.1016/j.isci.2025.111862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/04/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Our goal was to probe the potential transcriptomic basis for the relationship between plasma levels of the specialized pro-resolving precursor, 17-hydroxy-docosahexaenoic acid (17-HDHA) and chronic pain. Participants with osteoarthritis (average age of 62.3, 60% were female, n = 30) were stratified by levels of 17-HDHA and self-reported pain scores. RNAs from CD14++/CD16-/CD66b-/HLA-DR+ (classical) monocytes were sequenced and differentially expressed mRNAs were identified with DESeq2. QIAGEN ingenuity pathway analysis identified the top ranked canonical biological pathway to be eukaryotic initiation factor 2 (EIF2) signaling (lower activation level in the low 17-HDHA-high pain group compared to the high 17-HDHA-low pain group (Z score -3)), followed by EIF4 and P70S6K signaling pathways and mTOR signaling. Our approach provides insight into the biological pathways contributing to the association between 17-HDHA and chronic osteoarthritis (OA) pain, identifying EIF2 signaling, with known roles in osteoclast differentiation, OA pathology, and pain, as a potential downstream target.
Collapse
Affiliation(s)
- Peter R.W. Gowler
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Asta Arendt-Tranholm
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - James Turnbull
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division and NIHR Nottingham Biomedical Research Centre, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Rakesh R. Jha
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division and NIHR Nottingham Biomedical Research Centre, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David Onion
- Flow Cytometry Facility, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Tony Kelly
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Afroditi Kouraki
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Paul Millns
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Sameer Gohir
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Susan Franks
- School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - David A. Barrett
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division and NIHR Nottingham Biomedical Research Centre, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Ana M. Valdes
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Victoria Chapman
- Pain Centre Versus Arthritis and NIHR Nottingham Biomedical Research Centre, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
13
|
Tavares LP, Libreros S, Bitounis D, Nshimiyimana R, Demokritou P, Serhan CN, Levy BD. SiO 2 nanoparticles as disruptors of endogenous resolution mechanisms of inflammatory responses that exacerbate pneumonia. Sci Rep 2025; 15:6398. [PMID: 39984537 PMCID: PMC11845501 DOI: 10.1038/s41598-025-89700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
Occupational exposure to engineered nanomaterials (ENMs) is increasing in the workplace and can impact human health. Amorphous silicon dioxide nanoparticles (SiO2 NPs) are widely produced respirable ENMs used in commercial products. We have investigated their impact on lung inflammation resolution and bacterial defense. Mice exposed to SiO2 NPs, followed by bacteria, exhibited increased lung inflammation, bacterial proliferation, and lung damage compared to mice not exposed to NPs. SiO2 NPs increased human macrophage production of pro-inflammatory mediators and disrupted phagocytosis of bacteria and efferocytosis of apoptotic neutrophils - pivotal responses for host defense and inflammation resolution. A pro-resolving mediator, resolvin D5 (RvD5), restored macrophage phagocytosis of bacteria and partially controlled excess lung inflammation after SiO2 NPs. These findings demonstrate that SiO2 NPs disrupt endogenous resolution processes to give rise to heightened lung inflammation and infection. RvD5 reduced inflammation and partially restored endogenous resolution cellular processes, suggesting that RvD5 can reduce ENP disruption of resolution.
Collapse
Affiliation(s)
- Luciana Pádua Tavares
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephania Libreros
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology and Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, USA
| | - Dimitrios Bitounis
- Nanoscience and Advanced Materials Center, Environmental and Occupational Health Science Institute, Rutgers Biomedical Health Sciences, Piscataway, NJ, 08854, USA
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Sanofi US., Cambridge, USA
| | - Robert Nshimiyimana
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Philip Demokritou
- Nanoscience and Advanced Materials Center, Environmental and Occupational Health Science Institute, Rutgers Biomedical Health Sciences, Piscataway, NJ, 08854, USA
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Tredicine M, Mucci M, Recchiuti A, Mattoscio D. Immunoregulatory mechanisms of the arachidonic acid pathway in cancer. FEBS Lett 2025. [PMID: 39973474 DOI: 10.1002/1873-3468.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
The arachidonic acid (AA) pathway promotes tumor progression by modulating the complex interactions between cancer and immune cells within the microenvironment. In this Review, we summarize the knowledge acquired thus far concerning the intricate mechanisms through which eicosanoids either promote or suppress the antitumor immune response. In addition, we will discuss the impact of eicosanoids on immune cells and how they affect responsiveness to immunotherapy, as well as potential strategies for manipulating the AA pathway to improve anticancer immunotherapy. Understanding the molecular pathways and mechanisms underlying the role played by AA and its metabolites in tumor progression may contribute to the development of more effective anticancer immunotherapies.
Collapse
Affiliation(s)
- Maria Tredicine
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Italy
- Center for Advanced Studies and Technology, University of Chieti-Pescara, Italy
| | - Matteo Mucci
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Italy
- Center for Advanced Studies and Technology, University of Chieti-Pescara, Italy
| | - Antonio Recchiuti
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Italy
- Center for Advanced Studies and Technology, University of Chieti-Pescara, Italy
| | - Domenico Mattoscio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Italy
- Center for Advanced Studies and Technology, University of Chieti-Pescara, Italy
| |
Collapse
|
15
|
Bascuñán KA, Araya M, Rodríguez JM, Roncoroni L, Elli L, Alvarez JDPL, Valenzuela R. Interplay of n-3 Polyunsaturated Fatty Acids, Intestinal Inflammation, and Gut Microbiota in Celiac Disease Pathogenesis. Nutrients 2025; 17:621. [PMID: 40004950 PMCID: PMC11858531 DOI: 10.3390/nu17040621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Celiac disease (CD) is a chronic autoimmune disorder driven by both genetic and environmental factors, with the HLA DQ2/DQ8 genotypes playing a central role in its development. Despite the genetic predisposition, only a small percentage of individuals carrying these genotypes develop the disease. Gluten, a protein found in wheat, rye, and barley, is the primary environmental trigger, but other factors, such as the intestinal microbiota, may also contribute to disease progression. While the gluten-free diet (GFD) remains the cornerstone of treatment, many CD patients experience persistent inflammation and gut dysbiosis, leading to ongoing symptoms and complications. This chronic inflammation, which impairs nutrient absorption, increases the risk of malnutrition, anemia, and other autoimmune disorders. Recent studies have identified an altered gut microbiota in CD patients, both on and off the GFD, highlighting the potential role of the microbiota in disease pathogenesis. An emerging area of interest is the supplementation of n-3 polyunsaturated fatty acids (PUFAs), known for their anti-inflammatory properties, as a potential therapeutic strategy. n-3 PUFAs, found in fish oil and certain plant oils, modulate the immune cell function and cytokine production, making them a promising intervention for controlling chronic inflammation in CD. This review explores the current understanding of n-3 PUFAs' effects on the gut microbiota's composition and inflammation in CD, with the goal of identifying new avenues for complementary treatments to improve disease management.
Collapse
Affiliation(s)
- Karla A. Bascuñán
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (K.A.B.); (J.D.P.L.A.)
| | - Magdalena Araya
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile (J.M.R.)
| | - Juan Manuel Rodríguez
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile (J.M.R.)
| | - Leda Roncoroni
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.R.); (L.E.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.R.); (L.E.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | | | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (K.A.B.); (J.D.P.L.A.)
| |
Collapse
|
16
|
Speckmann B, Jordan PM, Werz O, Hofstetter RK, Ehring E, Vogel ML, Venema K. Bacillusmegaterium DSM 32963 Enhances Specialized Pro-Resolving Mediator Production from an n-3 PUFA Salt in a Dynamic Model of the Human Intestine. Metabolites 2025; 15:105. [PMID: 39997730 PMCID: PMC11857772 DOI: 10.3390/metabo15020105] [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: 01/10/2025] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Omega-3 polyunsaturated fatty acids (n-3 PUFA) have been used in the treatment of inflammatory bowel diseases (IBD) and irritable bowel syndrome (IBS), and their effects are potentiated upon conversion to specialized pro-resolving mediators (SPM). Recent studies indicated that the probiotic bacterial strain Bacillus megaterium DSM 32963 can be used to enhance the production of SPM and its precursors in vivo. METHODS Here, we explored the contribution of Bacillus megaterium DSM 32963 to SPM production in a validated, dynamic model of the upper and lower intestine. The TIM-1 and TIM-2 models were applied, with the TIM-2 model inoculated with the fecal microbiota of healthy individuals and probed with an n-3 PUFA lysine salt with and without Bacillus megaterium DSM 32963 or an SPM-enriched fish oil or placebo. Kinetics of SPM production were assessed by metabololipidomics analysis, and survival and engraftment of the Bacillus megaterium strain was monitored by plate counting and by strain-specific qPCR. RESULTS Bacillus megaterium DSM 32963 poorly survived TIM-1 conditions but propagated in the TIM-2 model, where it enabled the metabolism of n-3 PUFA to SPM (resolvin E2 and protectin DX) and SPM precursors (e.g., 5-hydroxyeicosapentaenoic acid (5-HEPE), 15-HEPE, 18-HEPE, 4-hydroxydocosahexaenoic acid (4-HDHA), 10-HDHA, and 17-HDHA, among other EPA- and DHA-derived metabolites) with significantly higher levels of lipid mediator production compared to the n-3 PUFA lysine salt alone; esterified n-3 PUFA were hardly converted by the microbiota. CONCLUSIONS These findings reinforce that Bacillus megaterium DSM 32963 facilitates SPM production in situ from bioavailable n-3 PUFA in the large intestine, highlighting its use to complement eukaryotic SPM biosynthesis by the host and its possible therapeutic use for, e.g., IBD and IBS.
Collapse
Affiliation(s)
- Bodo Speckmann
- Evonik Operations GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany; (E.E.); (M.-L.V.)
| | - Paul M. Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany; (P.M.J.); (O.W.); (R.K.H.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany; (P.M.J.); (O.W.); (R.K.H.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Robert K. Hofstetter
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany; (P.M.J.); (O.W.); (R.K.H.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Ellen Ehring
- Evonik Operations GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany; (E.E.); (M.-L.V.)
| | - Marie-Luise Vogel
- Evonik Operations GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany; (E.E.); (M.-L.V.)
| | - Koen Venema
- Centre for Healthy Eating & Food Innovation, Campus Venlo, Maastricht University, Villafloraweg 1, 5928 SZ Venlo, The Netherlands;
| |
Collapse
|
17
|
Bose S, Do V, Testini C, Jadhav SS, Sailliet N, Kho AT, Komatsu M, Boneschansker L, Kong SW, Wedel J, Briscoe DM. Immunomodulation by allograft endothelial cells. FRONTIERS IN TRANSPLANTATION 2025; 4:1518772. [PMID: 39967861 PMCID: PMC11832486 DOI: 10.3389/frtra.2025.1518772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
It is increasingly appreciated that the expression of immunoregulatory molecules within tumors have potential to shape a microenvironment that promotes local immunoevasion and immunoregulation. However, little is known about tissue-intrinsic immunomodulatory mechanisms following transplantation. We propose that differences in the phenotype of microvascular endothelial cells impact the alloantigenicity of the graft and its potential to promote immunoregulation following transplantation. We focus this review on the concept that graft-dependent immunoregulation may evolve post-transplantation, and that it is dependent on the phenotype of select subsets of intragraft endothelial cells. We also discuss evidence that long-term graft survival is critically dependent on adaptive interactions among immune cells and endothelial cells within the transplanted tissue microenvironment.
Collapse
Affiliation(s)
- Sayantan Bose
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Vicki Do
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
| | - Chiara Testini
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Suchita S. Jadhav
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Nicolas Sailliet
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Alvin T. Kho
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | - Masaki Komatsu
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Leo Boneschansker
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Sek Won Kong
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | - Johannes Wedel
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | - David M. Briscoe
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Bartold M, Ivanovski S. Biological processes and factors involved in soft and hard tissue healing. Periodontol 2000 2025; 97:16-42. [PMID: 38243683 PMCID: PMC11808446 DOI: 10.1111/prd.12546] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/12/2023] [Accepted: 11/23/2023] [Indexed: 01/21/2024]
Abstract
Wound healing is a complex and iterative process involving myriad cellular and biologic processes that are highly regulated to allow satisfactory repair and regeneration of damaged tissues. This review is intended to be an introductory chapter in a volume focusing on the use of platelet concentrates for tissue regeneration. In order to fully appreciate the clinical utility of these preparations, a sound understanding of the processes and factors involved in soft and hard tissue healing. This encompasses an appreciation of the cellular and biological mediators of both soft and hard tissues in general as well as specific consideration of the periodontal tissues. In light of good advances in this basic knowledge, there have been improvements in clinical strategies and therapeutic management of wound repair and regeneration. The use of platelet concentrates for tissue regeneration offers one such strategy and is based on the principles of cellular and biologic principles of wound repair discussed in this review.
Collapse
Affiliation(s)
- Mark Bartold
- University of QueenslandBrisbaneQueenslandAustralia
| | | |
Collapse
|
19
|
Tyrrell AD, Cisbani G, Smith ME, Chen CT, Chen YT, Chouinard-Watkins R, Hopperton KE, Taha AY, Bazinet RP. Lipid mediators in post-mortem brain samples from patients with Alzheimer's disease: A systematic review. Brain Behav Immun Health 2025; 43:100938. [PMID: 39896840 PMCID: PMC11782888 DOI: 10.1016/j.bbih.2024.100938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 02/04/2025] Open
Abstract
A proposed contributor to Alzheimer's disease (AD) pathology is the induction of neuroinflammation due to tau and beta-amyloid protein accumulation causing neuronal injury and dysfunction. Dysregulation of lipid mediators derived from polyunsaturated fatty acids may contribute to this inflammatory response in the brain of patients with AD, yet the literature has not yet been systematically reviewed. A systematic search was conducted in Medline, Embase and PsychINFO for articles published up to April 22, 2024. Papers were included if they measured levels of lipid mediators and/or enzymes involved in their production in post-mortem brain samples from patients with AD and control without neurological disease. A total of 50 relevant studies were identified. Despite heterogeneity in the results, pro-inflammatory lipid mediators, including 5-, 11-, 12- and 15-hydroxyeicosatetraenoic acid oxylipins and prostaglandin D2, were significantly higher, while anti-inflammatory lipoxin A4 and DHA-derived docosanoids were significantly lower in brains of patients with AD compared to control (16 studies). Thirty-seven articles reported on enzymes, with 32 reporting values for enzyme level changes between AD and controls. Among the 32 articles, the majority reported on levels of cyclooxygenase (COX) (18/32), with fewer studies reporting on phospholipase (8/32), lipoxygenase (LOX) (4/32) and prostaglandin E synthase (4/32). Enzyme levels also exhibited variability in the literature, with a trend towards elevated expression of enzymes involved in the pro-inflammatory response, including COX and LOX enzymes. Overall, these results are consistent with the involvement of neuroinflammation in the pathogenesis of AD measured by lipid mediators. However, the specific contribution of each lipid metabolite and enzymes to either the progression or persistence of AD remains unclear, and more research is required.
Collapse
Affiliation(s)
- Aidan D. Tyrrell
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Giulia Cisbani
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Mackenzie E. Smith
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Chuck T. Chen
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Yue-Tong Chen
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | | | - Kathryn E. Hopperton
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Ameer Y. Taha
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA, USA
| | - Richard P. Bazinet
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| |
Collapse
|
20
|
Lovins HB, Mehta A, Leuenberger LA, Yaeger MJ, Schott E, Hutton G, Manke J, Armstrong M, Reisdorph N, Tighe RM, Cochran SJ, Shaikh SR, Gowdy KM. Dietary Eicosapentaenoic Acid Improves Ozone-Induced Pulmonary Inflammation in C57BL/6 Mice. J Nutr 2025; 155:465-475. [PMID: 39536972 DOI: 10.1016/j.tjnut.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Ambient concentrations of the air pollutant, ozone, are rising with increasing global temperatures. Ozone is known to increase incidence and exacerbation of chronic lung diseases, which will increase as ambient ozone levels rise. Studies have identified diet as a variable that is able to modulate the pulmonary health effects associated with ozone exposure. Eicosapentaenoic acid (EPA) is an n-3 (ω-3) PUFA consumed through diet, which lowers inflammation through conversion to oxylipins including hydroxy-eicosapentaenoic acids (HEPEs). However, the role of dietary EPA in ozone-induced pulmonary inflammation is unknown. OBJECTIVE Therefore, we hypothesized increasing dietary EPA will decrease ozone-induced pulmonary inflammation and injury through the production of HEPEs. METHODS To test this, male C57BL/6J mice were fed a purified control diet or EPA-supplemented diet for 4 wk and then exposed to filtered air or 1 part per million ozone for 3 h. 24 or 48 h after exposure, bronchoalveolar lavage fluid was collected to assess airspace inflammation/injury and lung tissue was collected for targeted liquid chromatography-mass spectrometry lipidomics. RESULTS Following ozone exposure, EPA supplementation did not alter markers of lung injury but decreased ozone-induced airspace neutrophilia. Targeted liquid chromatography-mass spectrometry lipidomics revealed dietary EPA supplementation increased pulmonary EPA-derived metabolites including 5-HEPE and 12-HEPE. Additionally, EPA supplementation decreased pulmonary amounts of proinflammatory arachidonic acid-derived metabolites. To evaluate whether dietary EPA reduces ozone-induced pulmonary inflammation through increased pulmonary HEPEs, C57BL/6J mice were administered 5-HEPEs and 12-HEPEs systemically before filtered air or ozone exposure. Pretreatment with 5-HEPEs and 12-HEPEs reduced ozone-driven increases in airspace macrophages. CONCLUSIONS Together, these data indicate that an EPA-supplemented diet protects against ozone-induced airspace inflammation which is, in part, due to increasing pulmonary amounts of 5-HEPEs and 12-HEPEs. These findings suggest that dietary EPA and/or increasing EPA-derived metabolites in the lung can reduce ozone-driven incidences and exacerbations of chronic pulmonary diseases.
Collapse
Affiliation(s)
- Hannah B Lovins
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Anushka Mehta
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Laura A Leuenberger
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Michael J Yaeger
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Evangeline Schott
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Grace Hutton
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Jonathan Manke
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO, United States
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO, United States
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO, United States
| | - Robert M Tighe
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Samuel J Cochran
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
21
|
Serhan CN, Levy BD. Proresolving Lipid Mediators in the Respiratory System. Annu Rev Physiol 2025; 87:491-512. [PMID: 39303274 PMCID: PMC11810588 DOI: 10.1146/annurev-physiol-020924-033209] [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] [Indexed: 09/22/2024]
Abstract
Lung inflammation, infection, and injury can lead to critical illness and death. The current means to pharmacologically treat excessive uncontrolled lung inflammation needs improvement because many treatments are or will become immunosuppressive. The inflammatory response evolved to protect the host from microbes, injury, and environmental insults. This response brings phagocytes from the bloodstream to the tissue site to phagocytize and neutralize bacterial invaders and enables airway antimicrobial functions. This physiologic response is ideally self-limited with initiation and resolution phases. Polyunsaturated essential fatty acids are precursors to potent molecules that govern both phases. In the initiation phase, arachidonic acid is converted to prostaglandins and leukotrienes that activate leukocytes to transmigrate from postcapillary venules. The omega-3 fatty acids (e.g., DHA and EPA) are precursors to resolvins, protectins, and maresins, which are families of chemically distinct mediators with potent functions in resolution of acute and chronic inflammation in the respiratory system.
Collapse
Affiliation(s)
- Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA;
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
22
|
La Rosa M, Spagnolo A, Gamonal JD, Marín MJ, Figuero E, Sanz M. In Vitro Infection of Human Macrophages with Porphyromonas gingivalis W83. Int J Mol Sci 2025; 26:1054. [PMID: 39940820 PMCID: PMC11817804 DOI: 10.3390/ijms26031054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
This study aimed to investigate the innate immune response of human macrophages to Porphyromonas gingivalis W83 using a novel in vitro infection model. The growth kinetics of P. gingivalis W83 were analyzed, revealing an exponential growth phase at 8 h (optical density = 0.70). To establish a reliable macrophage model, the differentiation of THP-1 monocytes into macrophages was optimized using low concentrations of phorbol 12-myristate 13-acetate (PMA). This approach induced enhanced adherence and morphological changes, with full differentiation achieved after 48 h of PMA treatment followed by 24 h of rest. Polarization towards the pro-inflammatory M1 phenotype was successfully induced with interferon-γ (IFN-γ) and lipopolysaccharide (LPS), as confirmed using cytokine profiling. Cytokine analysis using Luminex® technology demonstrated significant increases in interleukin (IL)-1β, tumor necrosis factor-α (TNF-α), and IL-6, indicating the effective activation of macrophages towards a pro-inflammatory phenotype. Building upon this macrophage model, this study investigated the interactions between macrophages and P. gingivalis W83 during its exponential growth phase. After a one-hour infection period, bacterial DNA quantification in supernatants and lysed macrophages revealed minimal levels of internalized or adherent bacteria, supporting the hypothesis that P. gingivalis effectively evades immune detection. These findings emphasize the utility of this model in uncovering the sophisticated immune evasion strategies employed by P. gingivalis, with significant implications for the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, Faculty of Dentistry, Complutense University, 28040 Madrid, Spain; (M.L.R.); (A.S.); (J.D.G.); (M.J.M.); (E.F.)
| |
Collapse
|
23
|
Parrish CC. Production, Transport, Fate and Effects of Lipids in the Marine Environment. Mar Drugs 2025; 23:52. [PMID: 39997176 PMCID: PMC11857299 DOI: 10.3390/md23020052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/26/2025] Open
Abstract
Lipids form energy storage depots, cellular barriers and signaling molecules. They are generated and metabolized by enzymes under the influence of biotic and abiotic factors, and some-the long-chain polyunsaturated ω3 and ω6 fatty acids and cholesterol-are essential for optimal health in marine organisms. In addition, lipids have direct and indirect roles in the control of buoyancy in marine fauna ranging from copepods to whales. Phytoplankton account for about half of the planet's carbon fixation, and about half of that carbon goes into lipids. Lipids are an important component of the ocean's ability to sequester carbon away from the atmosphere through sinking and especially after transfer to zooplankton. Phytoplankton are the main suppliers of ω3 polyunsaturated fatty acids (PUFAs) in the marine environment. They also supply cholesterol and many phytosterols to ocean ecosystems; however, genomics is indicating that members of the Cnidaria, Rotifera, Annelida, and Mollusca phyla also have the endogenous capacity for the de novo synthesis of ω3 PUFAs as well as phytosterols. It has been predicted that ω3 long-chain PUFAs will decrease in marine organisms with climate change, with implications for human consumption and for carbon sequestration; however, the responses of ω3 PUFA supply to future conditions are likely to be quite diverse.
Collapse
|
24
|
Yildiz O, Hunt GP, Schroth J, Dhillon G, Spargo TP, Al-Chalabi A, Koks S, Turner MR, Shaw PJ, Henson SM, Iacoangeli A, Malaspina A. Lipid-mediated resolution of inflammation and survival in amyotrophic lateral sclerosis. Brain Commun 2025; 7:fcae402. [PMID: 39816195 PMCID: PMC11733686 DOI: 10.1093/braincomms/fcae402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/26/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025] Open
Abstract
Neuroinflammation impacts on the progression of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. Specialized pro-resolving mediators trigger the resolution of inflammation. We investigate the specialized pro-resolving mediator blood profile and their receptors' expression in peripheral blood mononuclear cells in relation to survival in ALS. People living with ALS (pwALS) were stratified based on bulbar versus limb onset and on key progression metrics using a latent class model, to separate faster progressing from slower progressing ALS. Specialized pro-resolving mediator blood concentrations were measured at baseline and in one additional visit in 20 pwALS and 10 non-neurological controls (Cohort 1). Flow cytometry was used to study the GPR32 and GPR18 resolvin receptors' expression in peripheral blood mononuclear cells from 40 pwALS and 20 non-neurological controls (Cohort 2) at baseline and in two additional visits in 17 pwALS. Survival analysis was performed using Cox proportional hazards models, including known clinical predictors and GPR32 and GPR18 mononuclear cell expression. Differential expression and linear discriminant analyses showed that plasma resolvins were able to distinguish phenotypic variants of ALS from non-neurological controls. RvE3 was elevated in blood from pwALS, whilst RvD1, RvE3, RvT4 and RvD1n-3 DPA were upregulated in A-S and RvD2 in A-F. Compared to non-neurological controls, GPR32 was upregulated in monocytes expressing the active inflammation-suppressing CD11b+ integrin from fast-progressing pwALS, including those with bulbar onset disease (P < 0.0024), whilst GPR32 and GPR18 were downregulated in most B and T cell subtypes. Only GPR18 was upregulated in naïve double positive Tregs, memory cytotoxic Tregs, senescent late memory B cells and late senescent CD8+ T cells from pwALS compared to non-neurological controls (P < 0.0431). Higher GPR32 and GPR18 median expression in blood mononuclear cells was associated with longer survival, with GPR32 expression in classical monocytes (hazard ratio: 0.11, P = 0.003) and unswitched memory B cells (hazard ratio: 0.44, P = 0.008) showing the most significant association, along with known clinical predictors. Low levels of resolvins and downregulation of their membrane receptors in blood mononuclear cells are linked to a faster progression of ALS. Higher mononuclear cell expression of resolvin receptors is a predictor of longer survival. These findings suggest a lipid-mediated neuroprotective response that could be harnessed to develop novel therapeutic strategies and biomarkers for ALS.
Collapse
Affiliation(s)
- Ozlem Yildiz
- Neuromuscular Department, Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Guy P Hunt
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- Perron Institute for Neurological and Translational Science, Research Institute in Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Johannes Schroth
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Gurleen Dhillon
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Thomas P Spargo
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- Maurice Wohl Clinical Neuroscience Institute, King’s College Hospital, London SE5 9RS, UK
| | - Sulev Koks
- Perron Institute for Neurological and Translational Science, Research Institute in Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 7JX, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Sian M Henson
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King’s College London, London SE5 8AF, UK
| | - Andrea Malaspina
- Neuromuscular Department, Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
25
|
Puccini SJ, Healy CL, Harsch BA, Ahmed AR, Shearer GC, O’Connell TD. A Cell Autonomous Free fatty acid receptor 4 - ChemR23 Signaling Cascade Protects Cardiac Myocytes from Ischemic Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625260. [PMID: 39829927 PMCID: PMC11741238 DOI: 10.1101/2024.11.26.625260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Acute myocardial infarction (AMI) causes ischemic damage and cardiac remodeling that ultimately progresses into ischemic cardiomyopathy (ICM). Coronary revascularization reduces morbidity and mortality from an MI, however, reperfusion also induces oxidative stress that drives cardiac myocyte (CM) dysfunction and ICM. Oxidative stress in CMs leads to reactive oxygen species (ROS) production and mitochondrial damage. Free fatty acid receptor 4 (Ffar4) is a GPCR for long chain fatty acids (FA) that is expressed in multiple cell types including CMs. We have recently shown that CM-specific overexpression of Ffar4 protects the heart from systolic dysfunction in the context of ischemic injury. Mechanistically, in CMs, Ffar4 increases the levels of 18-hydroxyeicosapentaenoic acid (18-HEPE), an eicosapentaenoic acid (EPA)-derived, cardioprotective oxylipin (oxidatively modified FA). 18-HEPE is the precursor for resolvin E1 (RvE1), a cardioprotective, specialized pro-resolving mediator (SPM) that activates the GPCR ChemR23. We hypothesize Ffar4 in CMs protects the heart from oxidative stress and ischemic injury through activation of a CM-autonomous, Ffar4-ChemR23 cardioprotective signaling pathway. Here, we developed an in vitro hypoxia reoxygenation (H/R) model (3 hours of hypoxia, 17 hours of reoxygenation) in adult CMs as a model for ischemic injury. In adult CMs subjected to H/R, TUG-891, an Ffar4 agonist, attenuated ROS generation and TUG-891, 18-HEPE, and RvE1 protected CMs from H/R-induced cell death. More importantly, we found that the ChemR23 antagonist α-NETA prevented TUG-891 cytoprotection in adult CMs subjected to H/R, demonstrating that ChemR23 is required for Ffar4 cardioprotection. In summary, our data demonstrate co-expression of Ffar4 and ChemR23 in the same CM, that Ffar4, 18-HEPE, and RvE1 attenuate H/R-induced CM death, and that ChemR23 is required for Ffar4 cardioprotection in H/R support a CM-autonomous Ffar4-ChemR23 cardioprotective signaling pathway.
Collapse
Affiliation(s)
- Sara J. Puccini
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Chastity L. Healy
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Brian A. Harsch
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA
| | - Ahmed R. Ahmed
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
| | - Gregory C. Shearer
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA
| | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
26
|
Ratajczak W, Walczakiewicz K, Laszczyńska M, Chmielowiec K, Palma J, Drozd A, Lubkowska A, Sipak O. The profile of oxidative stress markers (arachidonic and linoleic acid derivatives) in patients with benign prostatic hyperplasia in relation to metabolic syndrome. Aging (Albany NY) 2025; 17:116-130. [PMID: 39773533 PMCID: PMC11810064 DOI: 10.18632/aging.206187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
So far, it has been proven that benign prostatic hyperplasia (BPH) is strongly associated with inflammation resulting from, i.a. the presence of infectious agent, autoimmune disease, aging process and lipid disorders associated with metabolic syndrome (MetS). We analyzed the association between serum eicosanoides (HETE, HODE, lipoxins, prostaglandin, and leucotrien) in aging man with benign prostatic hyperplasia (BPH) and healthy controls. The study involved 219 men (with BPH, n = 144; healthy controls, n = 75). We assessed the content arachidonic and linoleic acid derivatives in the serum samples of the study participants using liquid chromatography (HPLC). The levels of: RvE1 (p < 0.001); LXA4 5S,6R,15R (p = 0.001); 10S,17R-DiDHA (p < 0.001); MaR1 (p = 0.002); 9S-HODE (p < 0.05); 15S-HETE (p < 0.05); 12S-HETE (p < 0.001); 5-oxoETE (p < 0.05) and 5-HETE (p < 0.001) were significantly higher in patients with BPH than in the control group. PGE2 (p = 0.007), LTB4 (p < 0.001), and 18RS-HEPE (p < 0.001) were significantly higher in control group. We also analyzed the relationship between LXA4 5S,6R,15R serum levels of oxidative stress markers and concomitance of MetS. We noticed a relationship between levels and MetS (F1216 = 6.114965, p = 0.01). Our research results suggest that pro-inflammatory mediators and suppressors of inflammation are involved in the development of BPH, but their exact contribution has yet to be investigated.
Collapse
Affiliation(s)
- Weronika Ratajczak
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University, Żołnierska, Szczecin 71-210, Poland
| | | | - Maria Laszczyńska
- Department of Nursing, State University of Applied Sciences, Leśna, Koszalin 75-582, Poland
| | - Krzysztof Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, Zyty St., Zielona Góra 65-046, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University, Broniewskiego, Szczecin 71-460, Poland
| | - Arleta Drozd
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Broniewskiego, Szczecin 71-460, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University, Żołnierska, Szczecin 71-210, Poland
| | - Olimpia Sipak
- Department of Obstetrics and Pathology of Pregnancy, Pomeranian Medical University, Żołnierska, Szczecin 71-210, Poland
| |
Collapse
|
27
|
Qian Y, Ding J, Zhao R, Song Y, Yoo J, Moon H, Koo S, Kim JS, Shen J. Intrinsic immunomodulatory hydrogels for chronic inflammation. Chem Soc Rev 2025; 54:33-61. [PMID: 39499495 DOI: 10.1039/d4cs00450g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
The immune system plays a pivotal role in maintaining physiological homeostasis and influencing disease processes. Dysregulated immune responses drive chronic inflammation, which in turn results in a range of diseases that are among the leading causes of death globally. Traditional immune interventions, which aim to regulate either insufficient or excessive inflammation, frequently entail lifelong comorbidities and the risk of severe side effects. In this context, intrinsic immunomodulatory hydrogels, designed to precisely control the local immune microenvironment, have recently attracted increasing attention. In particular, these advanced hydrogels not only function as delivery mechanisms but also actively engage in immune modulation, optimizing interactions with the immune system for enhanced tissue repair, thereby providing a sophisticated strategy for managing chronic inflammation. In this tutorial review, we outline key elements of chronic inflammation and subsequently explore the strategic design principles of intrinsic immunomodulatory hydrogels based on these elements. Finally, we examine the challenges and prospects of such immunomodulatory hydrogels, which are expected to inspire further preclinical research and clinical translation in addressing chronic inflammation.
Collapse
Affiliation(s)
- Yuna Qian
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| | - Jiayi Ding
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Institute of Imaging Diagnosis and Minimally Invasive Intervention, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Rui Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Yang Song
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610065, China
| | - Jiyoung Yoo
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Huiyeon Moon
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Seyoung Koo
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jong Seung Kim
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| |
Collapse
|
28
|
Peh HY, Chen J. Pro-resolving lipid mediators and therapeutic innovations in resolution of inflammation. Pharmacol Ther 2025; 265:108753. [PMID: 39566561 DOI: 10.1016/j.pharmthera.2024.108753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
This review summarizes findings presented at the 19th World Congress of Basic & Clinical Pharmacology 2023 (Glasgow, Scotland, July 3rd to 7th, 2023) from 8 speakers in the field of resolution of inflammation, resolution pharmacology and resolution biology. It is now accepted that the acute inflammatory response is protective to defend the host against infection or tissue injury. Acute inflammation is self-limited and programmed to be limited in space and time: this is achieved through endogenous resolution processes that ensure return to homeostasis. Resolution is brought about by agonist mediators that include specialized pro-resolving lipid mediators (SPMs) and pro-resolving proteins and peptides such as annexin A1 and angiotensin-(1-7), all acting to initiate anti-inflammatory and pro-resolving processes. If the inflammatory reaction remains unchecked through dysfunctional resolution mechanism, it can become chronic and contribute to a plethora of human diseases, including respiratory, cardiovascular, metabolic, allergic diseases, and arthritis. Herein, we discuss how non-resolving inflammation plays a role in the pathogenesis of these diseases. In addition to SPMs, we highlight the discovery, biosynthesis, biofunctions, and latest research updates on innovative therapeutics (including annexin-A1 peptide-mimetic RTP-026, small molecule FPR2 agonist BM-986235/LAR-1219, biased agonist for FPR1/FPR2 Cmpd17b, lipoxin mimetics AT-01-KG and AT-02-CT, melanocortin receptor agonist AP1189, gold nanoparticles, angiotensin-(1-7), and CD300a) that can promote resolution of inflammation directly or through modulation of SPMs production. Drug development strategies based on the biology of the resolution of inflammation can offer novel therapeutic means and/or add-on therapies for the treatment of chronic diseases.
Collapse
Affiliation(s)
- Hong Yong Peh
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Pharmacology, Singapore; Immunology Programme and Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Jianmin Chen
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
29
|
Salamin O, Wheelock CE. Quantification of Octadecanoids in Human Plasma Using Chiral Supercritical Fluid Chromatography-Tandem Mass Spectrometry. Methods Mol Biol 2025; 2855:315-339. [PMID: 39354316 DOI: 10.1007/978-1-0716-4116-3_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Octadecanoids are a subset of oxylipins derived from 18-carbon fatty acids. These compounds have historically been understudied but have more recently attracted attention to their purported biological activity. One obstacle to the study of octadecanoids has been a lack of specific analytical methods for their measurement. A particular limitation has been the need for chiral-based methods that enable separation and quantification of individual stereoisomers. The use of chirality provides an additional dimension for distinguishing analytes produced enzymatically from those formed through autoxidation. In this chapter, we describe a comprehensive method using chiral supercritical fluid chromatography-tandem mass spectrometry (SFC-MS/MS) for the quantification of octadecanoids in human plasma. This method stands as an effective approach for quantifying octadecanoids and is applicable to diverse research applications including clinical research.
Collapse
Affiliation(s)
- Olivier Salamin
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
30
|
Ye Y, Yang Q, Wei J, Shen C, Wang H, Zhuang R, Cao Y, Ding Y, Xu H, Xiang S, Mei H, Li Z, Ren X, Zhang C, Xiao J, Zheng S, Li T, Zeng R, Liu H, Lin H, Shang-Guan W, Li M, Jin S, Wang Q. RvD1 improves resident alveolar macrophage self-renewal via the ALX/MAPK14/S100A8/A9 pathway in acute respiratory distress syndrome. J Adv Res 2025; 67:289-299. [PMID: 38237770 PMCID: PMC11725153 DOI: 10.1016/j.jare.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/23/2023] [Accepted: 01/12/2024] [Indexed: 02/24/2024] Open
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS) is a pulmonary inflammatory process primarily caused by sepsis. The resolution of inflammation is an active process involving the endogenous biosynthesis of specialized pro-resolving mediators, including resolvin D1 (RvD1). Resident alveolar macrophages (RAMs) maintain pulmonary homeostasis and play a key role in the resolution phase. However, the role of RAMs in promoting the resolution of inflammation by RvD1 is unclear. OBJECTIVES Here, we investigated the mechanisms of RvD1 on regulating RAMs to promote the resolution of ARDS. METHODS Mice were administered lipopolysaccharide and/or Escherichia coli via aerosol inhalation to establish a self-limited ARDS model. Then, RvD1 was administered at the peak inflammatory response. RAMs self-renewal was measured by flow cytometry, RAM phagocytosis was measured by two-photon fluorescence imaging. In addition, plasma was collected from intensive care unit patients on days 0-2, 3-5, and 6-9 to measure RvD1 and S100A8/A9 levels using triple quadrupole/linear ion trap mass spectrometry. RESULTS RAMs were found to play a pivotal role in resolving inflammation during ARDS, and RvD1 enhanced RAM proliferation and phagocytosis, which was abrogated by a lipoxin A4 receptor (ALX, RvD1 receptor) inhibitor. Both primary RAMs transfected with rS100A8/A9 and/or S100A8/A9 siRNA and S100A9-/- mice (also deficient in S100A8 function) showed higher turnover and phagocytic function, indicating that RvD1 exerted its effects on RAMs by inhibiting S100A8/A9 production in the resolution phase. RvD1 reduced S100A8/A9 and its upstream MAPK14 levels in vivo and in vitro. Finally, in the patients, RvD1 levels were lower, but S100A8/A9 levels were higher. CONCLUSIONS We propose that RvD1 improved RAM self-renewal and phagocytosis via the ALX/MAPK14/S100A8/A9 signaling pathway. Plasma RvD1 and S100A8/A9 levels were negatively correlated, and associated with the outcome of sepsis-induced ARDS.
Collapse
Affiliation(s)
- Yang Ye
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Qian Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Jinling Wei
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Chenxi Shen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Haixing Wang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Rong Zhuang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Yuan Cao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Yajun Ding
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Haoran Xu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Shuyang Xiang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Hongxia Mei
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Zhongwang Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Xiya Ren
- Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chen Zhang
- Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Ji Xiao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Shengxing Zheng
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Ting Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Ruifeng Zeng
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Huacheng Liu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Han Lin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Wangning Shang-Guan
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China
| | - Ming Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China.
| | - Qian Wang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China; Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, People's Republic of China.
| |
Collapse
|
31
|
Torrez JP, Otsuki DA, Zeferino SP, Sanchez AF, Auler JOC. Vasoplegic Syndrome Following Bypass: A Comprehensive Review of Pathophysiology and Proposed Treatments. Cureus 2025; 17:e78057. [PMID: 40013224 PMCID: PMC11863290 DOI: 10.7759/cureus.78057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
Following cardiopulmonary bypass (CPB) surgery, patients may experience vasoplegic or vasogenic shock syndrome. This condition has a variable incidence, reaching up to 44% in high-risk patients, with mortality rates ranging from 30% to 50%, primarily due to multiple organ failure. This complex condition is characterized by low arterial pressure, unresponsive vascular collapse to high doses of vasopressors, and biochemical signals of cellular oxygen debt. The cardiac output can either be low or abnormally elevated. A fundamental aspect of the pathophysiology of vasogenic syndrome after CPB is related to the dysfunction of vascular smooth muscle cell contraction. This syndrome is often associated with complex cardiac surgery such as reoperations, long periods of bypass and aorta clamping, and excessive blood transfusion. Some potential triggers that might lead to this condition include the preoperative use of antagonists of the renin-angiotensin system, calcium blockers antagonists, and chronic renal disease. Recent literature has advocated treating vasoplegic syndrome after bypass using oxide nitric synthase inhibitors, such as methylene blue and hydroxocobalamin, along with the progressive escalation of potent vasopressors and intravascular volume adjustment.
Collapse
Affiliation(s)
- Jaime P Torrez
- Anesthesiology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - Denise A Otsuki
- Anesthesiology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - Suely P Zeferino
- Anesthesiology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - Ana F Sanchez
- Anesthesiology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - José Otávio C Auler
- Anesthesiology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| |
Collapse
|
32
|
Yip KL, Zhou C, Anderson LL, Hawkins NA, Kearney JA, Arnold JC. A high seizure burden increases brain concentrations of specialized pro-resolving mediators in the Scn1a +/- mouse model of Dravet syndrome. Prostaglandins Other Lipid Mediat 2025; 176:106943. [PMID: 39701411 DOI: 10.1016/j.prostaglandins.2024.106943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVE Dravet syndrome is a severe, intractable epilepsy in which 80 % of patients have a de novo mutation in the gene SCN1A. We recently reported that a high seizure burden increased hippocampal concentrations of an array of pro-inflammatory prostaglandins in the Scn1a+/- mouse model of Dravet syndrome. This raised the possibility that a high seizure burden might also trigger the accumulation of specialized pro-resolving mediators that facilitate the resolution of neuroinflammation and brain repair. The present study therefore aimed to examine whether a high seizure burden increased hippocampal concentrations of various specialized pro-resolving mediators in the Scn1a+/- mouse model of Dravet syndrome. METHODS Scn1a+/- mice at postnatal day 21 (P21) were primed with a single hyperthermia-induced seizure event to induce a high seizure burden. On P24 primed Scn1a+/- mice with a high seizure burden, unprimed naïve Scn1a+/- mice and wild-type (WT) mice were euthanized and hippocampal tissue was collected for analysis of various specialized pro-resolving mediators using liquid chromatography mass spectrometry. RESULTS Scn1a+/- mice with a high seizure burden showed increased hippocampal concentrations of the pro-inflammatory leukotrienes B4 and E4. Further, a high seizure burden increased hippocampal concentrations of various special pro-resolving mediators, including the maresins (maresin1), D-series resolvins (RVD1 and RVD4), and protectin (PCTR1). To further characterize these changes, we determined the mRNA expression of lipoxygenase genes, as these synthetic enzymes are common across classes of specialized pro-resolving mediators. However, hippocampal expression of Alox5, Alox12 and Alox15 were not influenced by a high seizure burden. SIGNIFICANCE We report for the first time that a high seizure burden increases the hippocampal concentrations of various specialized pro-resolving mediators in Scn1a+/- mice. This provides a platform for future studies to examine whether modulation of these mediators might be exploited to reduce seizures and facilitate brain repair in intractable epilepsy syndromes.
Collapse
Affiliation(s)
- Ka Lai Yip
- Discipline of Pharmacology, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW 2050, Australia; Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, NSW 2050, Australia; Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
| | - Cilla Zhou
- Discipline of Pharmacology, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW 2050, Australia; Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, NSW 2050, Australia; Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
| | - Lyndsey L Anderson
- Discipline of Pharmacology, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW 2050, Australia; Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, NSW 2050, Australia; Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
| | - Nicole A Hawkins
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, IL 60611, USA
| | - Jennifer A Kearney
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, IL 60611, USA
| | - Jonathon C Arnold
- Discipline of Pharmacology, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW 2050, Australia; Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, NSW 2050, Australia; Brain and Mind Centre, The University of Sydney, NSW 2050, Australia.
| |
Collapse
|
33
|
Lamon-Fava S. Associations between omega-3 fatty acid-derived lipid mediators and markers of inflammation in older subjects with low-grade chronic inflammation. Prostaglandins Other Lipid Mediat 2025; 176:106948. [PMID: 39756792 PMCID: PMC11835203 DOI: 10.1016/j.prostaglandins.2025.106948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Cardiovascular disease (CVD), the leading cause of death in the United States and globally, is a chronic inflammatory disease likely caused by an impaired ability to resolve inflammation. Pre-clinical studies have provided strong evidence of the activating role of specialized pro-resolving lipid mediators (SPMs) derived from the omega-3 fatty acids eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA) on the resolution of inflammation. However, there is a dearth of information on the role of SPMs on inflammation in humans. Therefore, the aim of this study was to assess whether plasma concentrations of omega-3 fatty acids and their derived SPMs are associated with inflammatory markers in subjects with low-grade chronic inflammation (C-reactive protein >2 µg/mL). The plasma phospholipid content of omega-3 fatty acids, a marker of dietary intake, plasma concentrations of SPMs, and serum concentrations of inflammatory markers were measured in 21 older men and postmenopausal women (age 53-73 y) at the end of a four-week placebo phase (3 g/day high oleic acid sunflower oil). The phospholipid DHA content was inversely related to interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1) and IL-10 concentrations. Moreover, MCP-1 was inversely associated with the DHA-derived 14-HDHA and 4-HDHA, and IL-10 was inversely associated with EPA-derived 18-HEPE, 12-HEPE and 5-HEPE, DPA-derived Rv5DPA, and DHA-derived 4-HDHA. These findings support the anti-inflammatory effect of dietary omega-3 fatty and suggest that lipid mediators derived from EPA, DPA, and DHA participate in the regulation of inflammation in subjects with chronic inflammation.
Collapse
Affiliation(s)
- Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging and Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, United States.
| |
Collapse
|
34
|
Suber TL, Tabary M, Bain W, Olonisakin T, Lockwood K, Xiong Z, Zhang Y, Kohli N, Furguiele L, Peñaloza H, McVerry BJ, Rose JJ, Shah F, Methé B, Li K, Mallampalli RK, Chen K, Fan L, Morris A, Tyurin VA, Samovich SN, Bayir H, Tyurina YY, Kagan V, Lee JS. Oxidized phospholipid and transcriptomic signatures of THC-related vaping associated lung injury. Sci Rep 2024; 14:31622. [PMID: 39738089 PMCID: PMC11686108 DOI: 10.1038/s41598-024-79585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
E-cigarette/vaping-associated lung injury (EVALI) is strongly associated with vitamin E acetate and often occurs with concomitant tetrahydrocannabinol (THC) use. To uncover pathways associated with EVALI, we examined cytokines, transcriptomic signatures, and lipidomic profiles in bronchoalveolar lavage fluid (BALF) from THC-EVALI patients. At a single center, we prospectively enrolled mechanically ventilated patients with EVALI from THC-containing products (N = 4) and patients with non-vaping acute lung injury and airway controls (N = 5). BALF samples were analyzed by Luminex multiplex assay, RNA sequencing, and mass spectrometry. After treating BEAS-2B lung epithelial cells with vaping and non-vaping BALF, LDH release was quantified. THC-EVALI BALF had significant increases in IFNγ, CCL2, CXCL5, and MMP2 relative to non-vaping patients. RNA sequencing showed enrichment for biological oxidation, glucuronidation, and fatty acid metabolism pathways. Oleic acid and arachidonic acid metabolites were increased in THC-EVALI, as were oxidized phosphatidylethanolamines (PE) such as PE(38:4). THC-EVALI BALF induced more LDH release compared to BALF from non-vaping patients. Thus, THC-EVALI is characterized by altered phospholipid composition, accumulation of lipid oxidation products, and increased pro-inflammatory mediators that may contribute to epithelial cell death. These findings serve as a framework to study novel oxidized phospholipids implicated in the pathogenesis of EVALI.
Collapse
Affiliation(s)
- Tomeka L Suber
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Mohammadreza Tabary
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | | | - Karina Lockwood
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zeyu Xiong
- Division of Pulmonary and Critical Care Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Naina Kohli
- Edward via College of Osteopathic Medicine, Spartanburg, SC, USA
| | | | - Hernán Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Laboratorios Clínicos, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bryan J McVerry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jason J Rose
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Faraaz Shah
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Barbara Methé
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kelvin Li
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Kong Chen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Li Fan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alison Morris
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vladimir A Tyurin
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Svetlana N Samovich
- Division of Critical Care and Hospital Medicine, Redox Health Center, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Hülya Bayir
- Division of Critical Care and Hospital Medicine, Redox Health Center, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Janet S Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary and Critical Care Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
35
|
Quinlivan KM, Howard IV, Southan F, Bayer RL, Torres KL, Serhan CN, Panigrahy D. Exploring the unique role of specialized pro-resolving mediators in cancer therapeutics. Prostaglandins Other Lipid Mediat 2024; 178:106944. [PMID: 39722403 DOI: 10.1016/j.prostaglandins.2024.106944] [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: 07/16/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Unresolved chronic inflammation, a hallmark of cancer, promotes tumor growth and metastasis in various cancer types. In contrast to blocking inflammation, stimulation of resolution of inflammation is an entirely novel approach to "resolve" inflammation. Resolution of inflammation mechanisms in cancer includes clearance of tumor debris, counter-regulation of pro-inflammatory eicosanoids and cytokines, and suppression of leukocyte infiltration. Conventional cytotoxic chemotherapy, radiation, anti-angiogenic therapy, and immune checkpoint inhibitors directly or indirectly can lead to the generation of pro-tumorigenic cellular debris. Over the past two decades, a potential paradigm shift has emerged in the inflammation field with the discovery of specialized pro-resolving mediators (SPMs), including resolvins, lipoxins, maresins, and protectins. SPMs are structurally distinct families of mediators grouped together by their pro-resolving "debris-clearing" functions. "Pro-resolving" therapies are in clinical development for various inflammation-driven diseases, including cancer. SPMs, as novel cancer therapeutics, have tremendous potential to enhance current cancer therapy. The mechanisms of SPMs as anti-cancer therapeutics are under active investigation by various laboratories worldwide. Here, we explore the current appreciation of the SPMs as innovative potential treatments designed to harness the unique anti-cancer activity of SPMs.
Collapse
Affiliation(s)
- Katherine M Quinlivan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Isabella V Howard
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Franciska Southan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Rachel L Bayer
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Kimberly L Torres
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| |
Collapse
|
36
|
Regidor PA, Eiblwieser J, Steeb T, Rizo JM. Omega-3 long chain fatty acids and their metabolites in pregnancy outcomes for the modulation of maternal inflammatory- associated causes of preterm delivery, chorioamnionitis and preeclampsia. F1000Res 2024; 13:882. [PMID: 39931317 PMCID: PMC11809487 DOI: 10.12688/f1000research.153569.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 02/13/2025] Open
Abstract
Preterm birth is a major cause of perinatal complications and neonatal deaths. Furthermore, in the field of obstetrics many clinical entities like uterine contractions or the occurrence of pre- eclampsia remain to be serious complications during pregnancy and represent a major psychological, financial, and economic burden for society. Several published guidelines, studies and recommendations have highlighted the importance of supplementation of omega-3 long chain polyunsaturated fatty acids (PUFAs) during pregnancy. This narrative review aims at giving an overview on the modern perception of inflammatory processes and the role of specialized pro-resolving mediators (SPMs) in their resolution, especially in obstetrics. Additionally, we highlight the possible role of SPMs in the prevention of obstetric complications through oral supplementation using enriched marine oil nutritional's. The intake of PUFAs may result in an overall improvement of pregnancy outcomes by contributing to fetal brain growth and neurological development but more importantly though modulation of inflammation-associated pathologies. Especially the use of SPMs represents a promising approach for the management of obstetric and perinatal complications. SPMs are monohydroxylates derived from enriched marine oil nutritional's that involve certain pro-resolutive metabolites of omega-3 long chains PUFAs and may contribute to an attenuation of inflammatory diseases. This may be obtained through various mechanisms necessary for a proper resolution of inflammation such as the termination of neutrophil tissue infiltration, initiation of phagocytosis, downregulation of pro-inflammatory cytokines or tissue regeneration. In this way, acute and chronic inflammatory diseases associated with serious obstetrical complications can be modulated, which might contribute to an improved pregnancy outcome.
Collapse
Affiliation(s)
| | - Johanna Eiblwieser
- Medical Department, Exeltis Germany, Ismaning, Adalperostr. 84, 85737, Germany
| | - Theresa Steeb
- Medical Department, Exeltis Germany, Ismaning, Adalperostr. 84, 85737, Germany
| | | |
Collapse
|
37
|
Cavaillon JM, Chaudry IH. Facing stress and inflammation: From the cell to the planet. World J Exp Med 2024; 14:96422. [PMID: 39713080 PMCID: PMC11551703 DOI: 10.5493/wjem.v14.i4.96422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 10/31/2024] Open
Abstract
As identified in 1936 by Hans Selye, stress is shaping diseases through the induction of inflammation. But inflammation display some yin yang properties. On one hand inflammation is merging with the innate immune response aimed to fight infectious or sterile insults, on the other hand inflammation favors chronic physical or psychological disorders. Nature has equipped the cells, the organs, and the individuals with mediators and mechanisms that allow them to deal with stress, and even a good stress (eustress) has been associated with homeostasis. Likewise, societies and the planet are exposed to stressful settings, but wars and global warming suggest that the regulatory mechanisms are poorly efficient. In this review we list some inducers of the physiological stress, psychologic stress, societal stress, and planetary stress, and mention some of the great number of parameters which affect and modulate the response to stress and render it different from an individual to another, from the cellular level to the societal one. The cell, the organ, the individual, the society, and the planet share many stressors of which the consequences are extremely interconnected ending in the domino effect and the butterfly effect.
Collapse
Affiliation(s)
| | - Irshad H Chaudry
- Department of Surgery, University of Alabama Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
38
|
Threatt AN, White J, Klepper N, Brier Z, Dean LS, Ibarra A, Harris M, Jones K, Wahl MJL, Barahona M, Oyewole EO, Pauly M, Moreno JA, Nordgren TM. Aspirin-triggered resolvin D1 modulates pulmonary and neurological inflammation in an IL-22 knock-out organic dust exposure mouse model. Front Immunol 2024; 15:1495581. [PMID: 39776904 PMCID: PMC11705093 DOI: 10.3389/fimmu.2024.1495581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Agriculture dust contains many organic immunogenic compounds, and organic dust exposure is strongly associated with the development of immune-mediated chronic pulmonary diseases such as chronic obstructive pulmonary disease (COPD). Chronic organic dust exposure from agriculture sources induces chronic lung inflammatory diseases and organic dust exposure has recently been linked to an increased risk of developing dementia. The cytokine interleukin-22 (IL-22) has been established as an important mediator in the resolution and repair of lung tissues. The omega-3 fatty acid metabolite aspirin-triggered Resolvin D1 (AT-RvD1) has shown efficacy in modulating the immune response in both pulmonary and neurological inflammation but has not been explored as a therapeutic in organic dust exposure-induced neuroinflammation. Investigating the link between IL-22 and AT-RvD1 may help in developing effective therapies for these immune-mediated diseases. We aimed to investigate the link between organic dust exposure and neuroinflammation, the role of IL-22 in the pulmonary and neurological immune response to organic dust exposure, and the immune-modulating therapeutic applications of AT-RvD1 in an IL-22 knock-out mouse model of organic dust exposure. C57BL/6J (WT) and IL-22 knock-out (KO) mice were repetitively exposed to aqueous agriculture organic dust extract (DE) 5 days per week for 3 weeks (15 total instillations) and treated with AT-RvD1 either once per week (3 total injections) or 5 times per week (15 total injections) for 3 weeks and allowed to recover for 3 days. We observed a significant pulmonary and neurological immune response to DE characterized by the development of inducible bronchus associated lymphoid tissue in the lung and gliosis in the frontal areas of the brain. We also observed that IL-22 knock-out increased pulmonary and neurological inflammation severity. Animals exposed to DE and treated with AT-RvD1 displayed reduced lung pathology severity and gliosis. Our data demonstrate that DE exposure contributes to neurological inflammation and that IL-22 is crucial to effective tissue repair processes. Our data further suggest that AT-RvD1 may have potential as a novel therapeutic for organic dust exposure-induced, immune-mediated pulmonary and neurological inflammation, improving outcomes of those with these diseases.
Collapse
Affiliation(s)
- Alissa N. Threatt
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Jade White
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Nathan Klepper
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Animal Sciences, College of Agricultural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Zachary Brier
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Logan S. Dean
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
| | - Ash Ibarra
- Department of Chemistry, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Macallister Harris
- Experimental Pathology Facility, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Kaylee Jones
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Maëlis J. L. Wahl
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Melea Barahona
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
| | - Emmanuel O. Oyewole
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Morgan Pauly
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Julie A. Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Brain Research Center, Colorado State University, Fort Collins, CO, United States
| | - Tara M. Nordgren
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
39
|
Osete JM, García-Candel F, Fernández-Gómez FJ, Blanquer M, Atucha NM, García-Estañ J, Iyú D. TRAP-Induced Platelet Reactivity Is Inhibited by Omega-3 Fatty Acid-Derived Prostaglandin E3 (PGE3). Biomedicines 2024; 12:2855. [PMID: 39767761 PMCID: PMC11673155 DOI: 10.3390/biomedicines12122855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Prostaglandins are naturally occurring local mediators that can participate in the modulation of the cardiovascular system through their interaction with Gs/Gi-coupled receptors in different tissues and cells, including platelets. Thrombin is one of the most important factors that regulates platelet reactivity and coagulation. Clinical trials have consistently shown that omega-3 fatty acid supplementation lowers the risk for cardiovascular mortality and morbidity. Since omega-3 fatty acids are the main precursors of PGE3 in vivo, it would be relevant to investigate the effects of PGE3 on Thrombin Receptor Activating Peptide (TRAP-6)-induced platelet reactivity to determine the receptors and possible mechanisms of action of these compounds. Methods: We have measured platelet aggregation, P-selectin expression, and vasodilator-stimulated phosphoprotein (VASP) phosphorylation to evaluate platelet reactivity induced by TRAP-6 to determine the effects of PGE3 on platelet function. Results: We assessed the ability of DG-041, a selective prostanoid EP3 receptor antagonist, and of ONO-AE3-208, a selective prostanoid EP4 receptor antagonist, to modify the effects of PGE3. PGE3 inhibited TRAP-6-induced platelet aggregation and activation. This inhibition was enhanced in the presence of a Gi-coupled EP3 receptor antagonist and abolished in the presence of a Gs-coupled EP4 receptor antagonist. The effects of PGE3 were directly related to changes in cAMP, assessed by VASP phosphorylation. Conclusions: The general effects of PGE3 on human platelet reactivity are the consequence of a balance between activatory and inhibitory effects at receptors that have contrary effects on adenylate cyclase. These results indicate a potential mechanism by which omega-3 fatty acids underlie cardioprotective effects.
Collapse
Affiliation(s)
- José-Miguel Osete
- Department of Physiology, University of Murcia, 30120 Murcia, Spain; (J.-M.O.); (N.M.A.); (J.G.-E.)
| | - Faustino García-Candel
- Servicio de Hematología, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)—Arrixaca, Unidad de Trasplante Hematopoyético y Terapia Celular, 30120 Murcia, Spain;
| | | | - Miguel Blanquer
- Instituto Murciano de Investigación Biosanitaria (IMIB)—Arrixaca, Unidad de Trasplante Hematopoyético y Terapia Celular, 30120 Murcia, Spain;
- Department of Medicine, University of Murcia, 30120 Murcia, Spain
| | - Noemí M. Atucha
- Department of Physiology, University of Murcia, 30120 Murcia, Spain; (J.-M.O.); (N.M.A.); (J.G.-E.)
- Instituto Murciano de Investigación Biosanitaria (IMIB)—Arrixaca, Unidad de Trasplante Hematopoyético y Terapia Celular, 30120 Murcia, Spain;
| | - Joaquín García-Estañ
- Department of Physiology, University of Murcia, 30120 Murcia, Spain; (J.-M.O.); (N.M.A.); (J.G.-E.)
- Instituto Murciano de Investigación Biosanitaria (IMIB)—Arrixaca, Unidad de Trasplante Hematopoyético y Terapia Celular, 30120 Murcia, Spain;
| | - David Iyú
- Department of Physiology, University of Murcia, 30120 Murcia, Spain; (J.-M.O.); (N.M.A.); (J.G.-E.)
- Instituto Murciano de Investigación Biosanitaria (IMIB)—Arrixaca, Unidad de Trasplante Hematopoyético y Terapia Celular, 30120 Murcia, Spain;
| |
Collapse
|
40
|
Ervik K, Li YZ, Ji RR, Serhan CN, Hansen TV. Synthesis of the methyl ester of 17( R/ S)-Me-RvD5 n-3 DPA and relief of postoperative pain in male mice. Org Biomol Chem 2024; 22:9266-9270. [PMID: 39513388 PMCID: PMC11563200 DOI: 10.1039/d4ob01534g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
The synthesis and biological evaluation of 17(R/S)-Me-RvD5n-3 DPA, an analog of the specialized pro-resolving mediators RvD5 and RvD5n-3 DPA, are presented. The synthesis was successfully accomplished utilizing Midland Alpine borane reduction, Sonogashira cross-coupling and a one-pot hydrozirconation/iodination protocol. In vivo evaluation of RvD5, RvD5n-3 DPA and 17(R/S)-Me-RvD5n-3 DPA in a mouse model of fracture revealed that all three compounds inhibited postoperative pain in male mice, but not in female mice.
Collapse
Affiliation(s)
- Karina Ervik
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, P.O Box 1068, 0316 Oslo, Norway.
| | - Yi-Ze Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Hale Building for Transformative Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Trond V Hansen
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, P.O Box 1068, 0316 Oslo, Norway.
| |
Collapse
|
41
|
Wang Y, Zhang J, Shao C. Cytological changes in radiation-induced lung injury. Life Sci 2024; 358:123188. [PMID: 39481833 DOI: 10.1016/j.lfs.2024.123188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Radiation-induced lung injury (RILI) is a prevalent complication associated with radiotherapy for thoracic tumors. Based on the pathological progression, it can be categorized into two stages: early radiation pneumonitis and late radiation pulmonary fibrosis. The occurrence of RILI not only constrains the therapeutic dose that can be administered to the tumor target area but also significantly impairs patients' health and quality of life, thereby limiting the efficacy and applicability of radiotherapy. To effectively prevent and mitigate the development of RILI, it is crucial to disclose its underlying mechanisms. This review aims to elucidate the specific mechanisms involved in RILI and to examine the roles of various cell types, including lung parenchymal cells and different immune cells. The functions and interactions of lung epithelial cells, pulmonary vascular endothelial cells, a variety of immune cells, and fibroblasts during different stages of inflammation, tissue repair, and fibrosis following radiation-induced lung injury are analyzed. A comprehensive understanding of the dynamic changes in these cellular components is anticipated to offer new strategies for the prevention of RILI.
Collapse
Affiliation(s)
- Yun Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China.
| |
Collapse
|
42
|
Zheng Z, Zhao M, Xu Y, Zhang J, Peng S, Liu J, Pan W, Yin Z, Wei C, Qin JJ, Wan J, Wang M. Resolvin D2/GPR 18 axis ameliorates pressure overload-induced heart failure by inhibiting pro-inflammatory macrophage polarization. J Lipid Res 2024; 65:100679. [PMID: 39490925 DOI: 10.1016/j.jlr.2024.100679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Accumulating evidence has revealed that chronic unresolved inflammation can cause significant tissue damage and can be a key mediator of advanced heart failure (HF). Resolvin (Rv) D2, a member of specialized pro-resolving lipid mediators (SPMs), plays a protective role in various diseases by facilitating resolution. However, whether RvD2 participates in the pathogenesis of HF is still unclear. Our study demonstrated that RvD2 treatment mitigated cardiac remodeling and improved cardiac function in HF mice induced by pressure overload. The absence of G protein-coupled receptor 18 (GPR18), an endogenous receptor for RvD2, abolished the beneficial effects of RvD2 on HF. Additionally, RvD2 inhibited inflammatory responses and Ly6Chigh macrophage polarization during both early and late inflammatory stages involved in HF. Further investigation revealed that bone marrow transplantation from Gpr18 deficient mice into WT mice blocked the protective effects of RvD2 in HF mice. Moreover, Gpr18 deficiency impeded RvD2's capacity to downregulate inflammatory responses and Ly6Chigh macrophage polarization. Consistent with experiments in vivo, RvD2 treatment in bone marrow-derived macrophages (BMDMs) reduced inflammatory responses through its receptor GPR18. Mechanistically, RvD2 suppressed the phosphorylation of STAT1 and NF-κB p65, and the effects of RvD2 were reversed by the application of STAT1 or NF-κB p65 agonists in BMDMs. In conclusion, RvD2/GPR18 axis improved cardiac remodeling and function in pressure overload-induced HF mice by modulating macrophage phenotype via STAT1 and NF-κB p65 pathways. Our findings underscore the anti-inflammatory potential of RvD2/GPR18 axis, suggesting that RvD2/GPR18 axis may be a potential strategy for the treatment of HF.
Collapse
Affiliation(s)
- Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
43
|
Suárez LJ, Hasturk H, Tubero Euzebio Alves V, Díaz-Baez D, Van Dyke T, Kantarci A. Overexpression of the receptor for resolvin E1 (ERV1) prevents early alveolar bone loss in leptin receptor deficiency-induced diabetes. J Periodontol 2024; 95:1190-1200. [PMID: 39031577 DOI: 10.1002/jper.24-0060] [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: 01/28/2024] [Revised: 04/24/2024] [Accepted: 05/13/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND This study was designed to test the hypothesis that the leptin receptor (LepR) regulates changes in periodontal tissues and that the overexpression of the receptor for resolvin E1 (ERV1) prevents age- and diabetes-associated alveolar bone loss. METHODS LepR-deficient transgenic (TG) mice were cross-bred with those overexpressing ERV1 (TG) to generate double-TG mice. In total, 95 mice were divided into four experimental groups: wild type (WT), TG, LepR deficient (db/db), and double transgenic (db/db TG). The groups were followed from 4 weeks up to 16 weeks of age. The natural progression of periodontal disease without any additional method of periodontitis induction was assessed by macroscopic and histomorphometric analyses. Osteoclastic activity was measured by tartrate-resistant acid phosphatase (TRAP) staining. RESULTS At 4 weeks, ERV1 overexpression prevented weight gain. From Week 8 onward, there was a significant increase in the weight of db/db mice with or without ERV1 overexpression compared to the WT mice, accompanied by an increase in glucose levels. By 8 weeks of age, the percentage of bone loss in the LepR deficiency groups was significantly greater compared to WT mice. ERV1 overexpression in the db/db TG mice prevented early alveolar bone loss; however, it did not impact the development of diabetic bone loss in aging mice after the onset of weight gain and diabetes. CONCLUSIONS The findings suggest that the overexpression of ERV1 prevents LepR-associated alveolar bone loss during the early phases of periodontal disease by delaying weight gain, diabetes onset, and associated inflammation; however, LepR deficiency increases susceptibility to naturally occurring inflammatory alveolar bone loss as the animal ages, associated with excess weight gain, onset of diabetes, and excess inflammation.
Collapse
Affiliation(s)
- Lina J Suárez
- ADA Forsyth Institute, Cambridge, Massachusetts, USA
- Universidad Nacional de Colombia, Bogotá, Colombia
| | - Hatice Hasturk
- ADA Forsyth Institute, Cambridge, Massachusetts, USA
- Harvard University, Boston, Massachusetts, USA
| | | | | | - Thomas Van Dyke
- ADA Forsyth Institute, Cambridge, Massachusetts, USA
- Harvard University, Boston, Massachusetts, USA
| | - Alpdogan Kantarci
- ADA Forsyth Institute, Cambridge, Massachusetts, USA
- Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Nunes VS, Rogério AP, Abrahão O, Serhan CN. Leukotriene B4 receptor 1 (BLT1) activation by leukotriene B4 (LTB 4) and E resolvins (RvE1 and RvE2). Comput Biol Chem 2024; 113:108236. [PMID: 39395248 PMCID: PMC11645204 DOI: 10.1016/j.compbiolchem.2024.108236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024]
Abstract
Leukotriene B4 (LTB4) is a lipid inflammatory mediator derived from arachidonic acid (AA). Leukotriene B4 receptor 1 (BLT1), a G protein-coupled receptor (GPCR), is a receptor of LTB4. Nonetheless, the resolution of inflammation is driven by specialized pro-resolving lipid mediators (SPMs) such as resolvins E1 (RvE1) and E2 (RvE2). Both resolvins are derived from omega-3 fatty acid eicosapentaenoic acid (EPA). Here, long-term molecular dynamics simulations (MD) were performed to investigate the activation of the BLT1 receptor using two pro-resolution agonists (RvE1 and RvE2) and an inflammatory agonist (LTB4). We have analyzed the receptor's activation state, electrostatic interactions, and the binding affinity the Molecular Mechanics Poisson-Boltzmann Surface Area (MMPBSA) approach. The results showed that LTB4 and RvE1 have kept the receptor in an active state by higher simulation time. MD showed that the ligand-receptor interactions occurred mainly through residues H94, R156, and R267. The MMPBSA calculations showed residues R156 and R267 were the two mainly hotspots. Our MMPBSA results were compatible with experimental results from other studies. Overall, the results from this study provide new insights into the activation mechanisms of the BLT1 receptor, reinforcing the role of critical residues and interactions in the binding of pro-resolution and pro-inflammatory agonists.
Collapse
Affiliation(s)
- Vinicius S Nunes
- Programa de Pós-Graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Macaé, Rio de Janeiro, Brazil; Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil.
| | - Alexandre P Rogério
- Laboratório de Imunofarmacologia Experimental, Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Odonírio Abrahão
- Laboratório de Química Computacional Medicinal, Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, MassGeneral Brigham (MGB) and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Deng Y, Wang F, Wang T, Zhang X, Chen D, Wang Y, Chen C, Pan G. Research progress in the mechanisms and functions of specialized pro-resolving mediators in neurological diseases. Prostaglandins Other Lipid Mediat 2024; 175:106905. [PMID: 39265777 DOI: 10.1016/j.prostaglandins.2024.106905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
The nervous system interacts with the immune system through a variety of cellular regulators, signaling pathways, and molecular mechanisms. Disruptions in these interactions lead to the development of multiple neurological diseases. Recent studies have identified that specialized pro-resolving mediators (SPMs) play a regulatory role in the neuroimmune system. This study reviews recent research on the function of SPMs in the inflammatory process and their association with the nervous system. The review aims to provide new perspectives for studying the pathogenesis of neurological diseases and identify novel targets for clinical therapy.
Collapse
Affiliation(s)
- Yu Deng
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Fei Wang
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, China; Yancheng TCM Hospital, Yancheng, Jiangsu 224000, China
| | - Tianle Wang
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Xu Zhang
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Du Chen
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China
| | - Yuhan Wang
- Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Chaojun Chen
- Guangzhou Hospital of Integrated Chinese and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510800, China.
| | - Guangtao Pan
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, China; Yancheng TCM Hospital, Yancheng, Jiangsu 224000, China.
| |
Collapse
|
46
|
Tozatto‐Maio K, Rós FA, Weinlich R, Rocha V. Inflammatory pathways and anti-inflammatory therapies in sickle cell disease. Hemasphere 2024; 8:e70032. [PMID: 39698332 PMCID: PMC11655128 DOI: 10.1002/hem3.70032] [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: 10/16/2023] [Revised: 07/12/2024] [Accepted: 09/17/2024] [Indexed: 12/20/2024] Open
Abstract
Sickle cell disease (SCD) is a monogenic disease, resulting from a single-point mutation, that presents a complex pathophysiology and high clinical heterogeneity. Inflammation stands as a prominent characteristic of SCD. Over the past few decades, the role of different cells and molecules in the regulation of the inflammatory process has been elucidated. In conjunction with the polymerization of hemoglobin S (HbS), intravascular hemolysis, which releases free heme, HbS, and hemoglobin-related damage-associated molecular patterns, initiates multiple inflammatory pathways that are not yet fully comprehended. These complex phenomena lead to a vicious cycle that perpetuates vaso-occlusion, hemolysis, and inflammation. To date, few inflammatory biomarkers can predict disease complications; conversely, there is a plethora of therapies that reduce inflammation in SCD, although clinical outcomes vary widely. Importantly, whether the clinical heterogeneity and complications are related to the degree of inflammation is not known. This review aims to further our understanding of the roles of main immune cells, and other inflammatory factors, as potential prognostic biomarkers for predicting clinical outcomes or identifying novel treatments for SCD.
Collapse
Affiliation(s)
- Karina Tozatto‐Maio
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Felipe A. Rós
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Ricardo Weinlich
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
| | - Vanderson Rocha
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
- Instituto D'Or de Ensino e Pesquisa, Rede D'OrSao PauloBrazil
- Department of Hematology, Churchill HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
47
|
Prouse T, Majumder S, Majumder R. Functions of TAM Receptors and Ligands Protein S and Gas6 in Atherosclerosis and Cardiovascular Disease. Int J Mol Sci 2024; 25:12736. [PMID: 39684449 DOI: 10.3390/ijms252312736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Atherosclerosis and cardiovascular disease are associated with high morbidity and mortality in industrialized nations. The Tyro3, Axl, and Mer (TAM) family of receptor tyrosine kinases is involved in the amplification or resolution of atherosclerosis pathology and other cardiovascular pathology. The ligands of these receptors, Protein S (PS) and growth arrest specific protein 6 (Gas6), are essential for TAM receptor functions in the amplification and resolution of atherosclerosis. The Axl-Gas6 interaction has various effects on cardiovascular disease. Mer and PS dampen inflammation, thereby protecting against atherosclerosis progression. Tyro3, the least studied TAM receptor in cardiovascular disease, appears to protect against fibrosis in post-myocardial infarction injury. Ultimately, PS, Gas6, and TAM receptors present an exciting avenue of potential therapeutic targets against inflammation associated with atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Teagan Prouse
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Rinku Majumder
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
48
|
Simard M, Nshimiyimana R, Chiang N, Rodriguez AR, Spur BW, Serhan CN. A potent proresolving mediator 17R-resolvin D2 from human macrophages, monocytes, and saliva. SCIENCE ADVANCES 2024; 10:eadq4785. [PMID: 39565847 PMCID: PMC11578181 DOI: 10.1126/sciadv.adq4785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Production of specialized proresolving mediators (SPMs) during the resolution phase in the acute inflammatory response is key to orchestrating complete resolution. Here, we uncovered a trihydroxy resolvin in fresh human saliva. We identified and determined its complete stereochemistry as 7S,16R,17R-trihydroxy-4Z,8E,10Z,12E,14E,19Z-docosahexaenoic acid (17R-RvD2) using total organic synthesis and matching of physical properties. The 17R-RvD2 was produced by activated human M2-like macrophages, M1-like macrophages, and human peripheral blood monocytes. 17R-RvD2 displayed potent proresolving functions (picomolar to nanomolar). Topical application of 17R-RvD2 on mouse ear skin reduced neutrophilic infiltration (~50%). 17R-RvD2 increased M2 markers CD206 and CD163 on human monocyte-derived macrophages and enhanced efferocytosis of senescent red blood cells by M2-like macrophages (EC50 ~ 2.6 × 10-14 M). In addition, 17R-RvD2 activated the RvD2 receptor and was equipotent to its epimer RvD2. 17R-RvD2 also significantly increased phagocytosis of Escherichia coli by human neutrophils. Together, these results establish the complete stereochemistry and potent proresolving functions of the previously unknown 17R-RvD2.
Collapse
Affiliation(s)
- Mélissa Simard
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Robert Nshimiyimana
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Nan Chiang
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ana R. Rodriguez
- Department of Cell Biology and Neuroscience, Virtua Health College of Medicine & Life Sciences of Rowan University, Stratford, NJ 08084, USA
| | - Bernd W. Spur
- Department of Cell Biology and Neuroscience, Virtua Health College of Medicine & Life Sciences of Rowan University, Stratford, NJ 08084, USA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
49
|
Lamminpää I, Amedei A, Parolini C. Effects of Marine-Derived Components on Cardiovascular Disease Risk Factors and Gut Microbiota Diversity. Mar Drugs 2024; 22:523. [PMID: 39590803 PMCID: PMC11595733 DOI: 10.3390/md22110523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiovascular diseases (CVDs), which comprise coronary heart disease, hypertension, and stroke, collectively represent the number one cause of death globally. Atherosclerosis is the dominant cause of CVDs, and its risk factors are elevated levels of low-density lipoprotein cholesterol and triglycerides, hypertension, cigarette smoking, obesity, and diabetes mellitus. In addition, diverse evidence highlights the role played by inflammation and clonal haematopoiesis, eventually leading to immunity involvement. The human microbiota project and subsequent studies using next-generation sequencing technology have indicated that thousands of different microbial species are present in the human gut. Disturbances in the gut microbiota (GM) composition, i.e., gut dysbiosis, have been associated with diseases ranging from localised gastrointestinal disorders to metabolic and cardiovascular illnesses. Of note, experimental studies suggested that GM, host immune cells, and marine-derived ingredients work together to ensure intestinal wall integrity. This review discusses current evidence concerning the links among GM, marine-derived ingredients, and human inflammatory disease. In detail, we summarise the impact of fish-derived proteins/peptides and algae components on CVD risk factors and gut microbiome. Furthermore, we describe the interplay among these dietary components, probiotics/prebiotics, and CVDs.
Collapse
Affiliation(s)
- Ingrid Lamminpää
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy;
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), 50134 Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy;
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), 50134 Florence, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50134 Florence, Italy
| | - Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, ‘Rodolfo Paoletti’, Via Balzaretti 9, Università degli Studi di Milano, 20133 Milano, Italy
| |
Collapse
|
50
|
Livshits G, Kalinkovich A. Resolution of Chronic Inflammation, Restoration of Epigenetic Disturbances and Correction of Dysbiosis as an Adjunctive Approach to the Treatment of Atopic Dermatitis. Cells 2024; 13:1899. [PMID: 39594647 PMCID: PMC11593003 DOI: 10.3390/cells13221899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease with multifactorial and unclear pathogenesis. Its development is characterized by two key elements: epigenetic dysregulation of molecular pathways involved in AD pathogenesis and disrupted skin and gut microbiota (dysbiosis) that jointly trigger and maintain chronic inflammation, a core AD characteristic. Current data suggest that failed inflammation resolution is the main pathogenic mechanism underlying AD development. Inflammation resolution is provided by specialized pro-resolving mediators (SPMs) derived from dietary polyunsaturated fatty acids acting through cognate receptors. SPM levels are reduced in AD patients. Administration of SPMs or their stable, small-molecule mimetics and receptor agonists, as well as supplementation with probiotics/prebiotics, demonstrate beneficial effects in AD animal models. Epidrugs, compounds capable of restoring disrupted epigenetic mechanisms associated with the disease, improve impaired skin barrier function in AD models. Based on these findings, we propose a novel, multilevel AD treatment strategy aimed at resolving chronic inflammation by application of SPM mimetics and receptor agonists, probiotics/prebiotics, and epi-drugs. This approach can be used in conjunction with current AD therapy, resulting in AD alleviation.
Collapse
Affiliation(s)
- Gregory Livshits
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv 6927846, Israel;
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv 6927846, Israel;
| |
Collapse
|