1
|
Livingston DBH, Sweet A, Chowdary M, Demissie MS, Rodrigue A, Gedara KP, Kishore L, Mahmoodianfard S, Power KA. Diet alters the effects of lipopolysaccharide on intestinal health and cecal microbiota composition in C57Bl/6 male mice. J Nutr Biochem 2025:109951. [PMID: 40345399 DOI: 10.1016/j.jnutbio.2025.109951] [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: 12/03/2024] [Revised: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
Lipopolysaccharide (LPS), a component of the gram-negative bacteria cell well, is established to induce an acute and transient inflammatory cascade upon exogenous administration in mice, while negatively impacting aspects of the microbiota gut-brain axis (mGBA). Dietary supplementation with flaxseed (FS) has been shown to partially attenuate these impairments across the mGBA, although the importance of intestinal and hepatic health has not been established in this context. In this study, we furthered our investigation of the impact of dietary supplementation with FS and FS oil (FO) in mitigating LPS-induced perturbations to intestinal health (i.e. cecal microbiota, barrier integrity, inflammation, short-chain fatty acid (SCFA) production), hepatic inflammation, and their relationships to LPS-induced systemic and neuroinflammation. We noted distinct, diet-dependent shifts in the microbiota composition twenty-four hours post-LPS injection, that may be partially driven by the LPS-induced anorexia. BD-fed mice challenged with LPS showed reduced Muribaculaceae, while FS-LPS mice had elevated Akkermansia, and both the FS-LPS and FO-LPS mice had reduced Bacteroides, each with unique correlations to diet intake. These LPS-driven shifts in the microbiota were coupled with elevated ileal mucous content in the FS- and FO-fed mice, and intestinal inflammation which was partially attenuated in the FS-fed mice. Collectively, we highlight that FS modulates aspects of the intestinal microenvironment, which may be related to the anti-inflammatory effects across the mGBA.
Collapse
Affiliation(s)
- Dawson B H Livingston
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Allison Sweet
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Maryam Chowdary
- Faculty of Science, Department of Biology, University of Ottawa, Ottawa, Canada, K1N 9A7
| | - Meron Samuel Demissie
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Alexane Rodrigue
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Kasuni Pillagawa Gedara
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Lalit Kishore
- Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5
| | - Salma Mahmoodianfard
- Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5
| | - Krista A Power
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1; Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5.
| |
Collapse
|
2
|
Rewell SSJ, Shad A, Chen L, Macowan M, Chu E, Gandasasmita N, Casillas-Espinosa PM, Li J, O'Brien TJ, Semple BD. A post-injury immune challenge with lipopolysaccharide following adult traumatic brain injury alters neuroinflammation and the gut microbiome acutely, but has little effect on chronic outcomes. Exp Neurol 2025; 386:115150. [PMID: 39842491 DOI: 10.1016/j.expneurol.2025.115150] [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: 09/29/2024] [Revised: 12/22/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
Patients with a traumatic brain injury (TBI) are susceptible to hospital-acquired infections, presenting a significant challenge to an already-compromised immune system. The consequences and mechanisms by which this dual insult worsens outcomes are poorly understood. This study aimed to explore how a systemic immune stimulus (lipopolysaccharide, LPS) influences outcomes following experimental TBI in young adult mice. Male and female C57Bl/6J mice underwent controlled cortical impact or sham surgery, followed by 1 mg/kg i.p. LPS or saline-vehicle at 4 days post-TBI, before behavioral assessment and tissue collection at 6 h, 24 h, 7 days or 6 months. LPS induced acute sickness behaviors including weight loss, transient hypoactivity, and increased anxiety-like behavior. Early systemic immune activation by LPS was confirmed by increased spleen weight and serum cytokines. In brain tissue, gene expression analysis revealed a time course of inflammatory immune activation in TBI or LPS-treated mice (e.g., IL-1β, IL-6, CCL2, TNFα), which was exacerbated in TBI + LPS mice. This group also presented with fecal microbiome dysbiosis at 24 h post-LPS, with reduced bacterial diversity and changes in the relative abundance of key bacterial genera associated with sub-acute neurobehavioral and immune changes. Chronically, TBI induced hyperactivity and cognitive deficits, brain atrophy, and increased seizure susceptibility, similarly in vehicle and LPS-treated groups. Together, findings suggest that an immune challenge with LPS early after TBI, akin to a hospital-acquired infection, alters the acute neuroinflammatory response to injury, but has no lasting effects. Future studies could consider more clinically-relevant models of infection to build upon these findings.
Collapse
Affiliation(s)
- Sarah S J Rewell
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia
| | - Ali Shad
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia
| | - Lingjun Chen
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Matthew Macowan
- Department of Immunology, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Erskine Chu
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Natasha Gandasasmita
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia
| | - Jian Li
- Department of Microbiology, Monash Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia; Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
3
|
Elzinga SE, Guo K, Turfah A, Henn RE, Webber‐Davis IF, Hayes JM, Pacut CM, Teener SJ, Carter AD, Rigan DM, Allouch AM, Jang D, Parent R, Glass E, Murphy GG, Lentz SI, Chen KS, Zhao L, Hur J, Feldman EL. Metabolic stress and age drive inflammation and cognitive decline in mice and humans. Alzheimers Dement 2025; 21:e70060. [PMID: 40110679 PMCID: PMC11923576 DOI: 10.1002/alz.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/17/2025] [Accepted: 01/31/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Metabolic stressors (obesity, metabolic syndrome, prediabetes, and type 2 diabetes [T2D]) increase the risk of cognitive impairment (CI), including Alzheimer's disease (AD). Immune system dysregulation and inflammation, particularly microglial mediated, may underlie this risk, but mechanisms remain unclear. METHODS Using a high-fat diet-fed (HFD) model, we assessed longitudinal metabolism and cognition, and terminal inflammation and brain spatial transcriptomics. Additionally, we performed hippocampal spatial transcriptomics and single-cell RNA sequencing of post mortem tissue from AD and T2D human subjects versus controls. RESULTS HFD induced progressive metabolic and CI with terminal inflammatory changes, and dysmetabolic, neurodegenerative, and inflammatory gene expression profiles, particularly in microglia. AD and T2D human subjects had similar gene expression changes, including in secreted phosphoprotein 1 (SPP1), a pro-inflammatory gene associated with AD. DISCUSSION These data show that metabolic stressors cause early and progressive CI, with inflammatory changes that promote disease. They also indicate a role for microglia, particularly microglial SPP1, in CI. HIGHLIGHTS Metabolic stress causes persistent metabolic and cognitive impairments in mice. Murine and human brain spatial transcriptomics align and indicate a pro-inflammatory milieu. Transcriptomic data indicate a role for microglial-mediated inflammatory mechanisms. Secreted phosphoprotein 1 emerged as a potential target of interest in metabolically driven cognitive impairment.
Collapse
Affiliation(s)
- Sarah E. Elzinga
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- Department of PhysiologyMichigan State UniversityEast LansingMichiganUSA
| | - Kai Guo
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Ali Turfah
- Department of BiostatisticsSchool of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Rosemary E. Henn
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - John M. Hayes
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Crystal M. Pacut
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Samuel J. Teener
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Andrew D. Carter
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Diana M. Rigan
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Adam M. Allouch
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Dae‐Gyu Jang
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Rachel Parent
- Department of Internal MedicineGeneral MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Emily Glass
- Department of Molecular and Integrative PhysiologyDivision of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Geoffrey G. Murphy
- Department of Molecular and Integrative PhysiologyDivision of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Stephen I. Lentz
- Department of Internal MedicineDivision of MetabolismEndocrinology, and DiabetesUniversity of MichiganAnn ArborMichiganUSA
| | - Kevin S. Chen
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Lili Zhao
- Department of BiostatisticsSchool of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Junguk Hur
- Department of Biomedical SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
4
|
Wang J, An W, Wang Z, Zhao Y, Han B, Tao H, Wang J, Wang X. Vanillin Has Potent Antibacterial, Antioxidant, and Anti-Inflammatory Activities In Vitro and in Mouse Colitis Induced by Multidrug-Resistant Escherichia coli. Antioxidants (Basel) 2024; 13:1544. [PMID: 39765873 PMCID: PMC11673545 DOI: 10.3390/antiox13121544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/08/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
A large number of cases of infectious colitis caused by multidrug-resistant (MDR) bacteria, such as Escherichia coli, can result in colon damage and severe inflammation. Vanilla, a widely utilized flavor and fragrance compound, is extensively used in various food. However, the effect of vanilla on MDR E. coli-induced infectious colitis has received less attention. In this study, the antibacterial activity of vanillin against MDR E. coli and other bacteria was determined by the microtiter broth dilution method. The antioxidant and anti-inflammatory capacity of vanillin was assessed in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells and MDR E. coli-induced mouse colitis. The results demonstrated that vanillin exhibited potent antibacterial activity against various strains of MDR E. coli, Salmonella, and Staphylococcus aureus, with a minimal inhibitory concentration (MIC) of 1.25-2.5 mg/mL and a minimum bactericidal concentration (MBC) of 5-10 mg/mL; it effectively inhibited cell division in E. coli. Vanillin also displayed remarkable antioxidant activity by suppressing the levels of malondialdehyde (MDA), superoxide dismutase (SOD), and reactive oxygen species (ROS) in LPS-stimulated RAW 264.7 cell; it significantly reduced the production of inflammatory mediators including nitroxide (NO), tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and interleukin 1β (IL-1β), while increasing interleukin 10 (IL-10). In an MDR E. coli-induced mouse colitis model, vanillin effectively inhibited inflammation by suppressing inflammatory cytokines, mitogen-activated protein kinase (MAPK), and nuclear factor κ-B (NF-κB) cell signaling pathway activation; it ameliorated changes in intestinal microflora characterized by decreased Firmicutes richness alongside increased Bacteroides richness, rebalancing the dysbiosis caused by E. coli. These findings highlight the potential pharmacological applicability of vanillin as a promising bioactive molecule for treating infectious colitis.
Collapse
Affiliation(s)
- Jiaxue Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
- College of Grassland Agriculture, Northwest A&F University, Yangling 712100, China
| | - Wei An
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ya Zhao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Bing Han
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Hui Tao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
5
|
Young GK, Chernyak D, Naik GA, Song SE, Beery AK. Prairie voles seek social contact with peer companions during immune challenge. Horm Behav 2024; 166:105653. [PMID: 39447313 DOI: 10.1016/j.yhbeh.2024.105653] [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: 07/05/2024] [Revised: 09/21/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024]
Abstract
Selection for group living has occurred across taxa, despite inherent risk of disease transmission. Behavioral and immune responses to sickness affect social interactions and can be altered by social contexts. However, the majority of research on sickness behavior has focused on species that do not form selective social relationships. Prairie voles (Microtus ochrogaster) form selective social relationships with mates and peers and provide a useful study system to examine effects of sickness on social seeking in established relationships. We used peripheral injections of lipopolysaccharide (LPS) of E. coli to stimulate the innate immune system and verified effects on activity, core temperature, and corticosterone concentrations for 6 h following treatment. We demonstrated that male and female same-sex pairs of prairie voles increase social contact when sick and that this increase persists when contact is initiated by the sick vole. Finally, we assessed social motivation following immune challenge using operant choice chambers equipped with two levers and side chambers. Voles worked to gain access to chambers with social and non-social rewards. While overall effort decreased following LPS injection, only immune-challenged voles worked significantly harder for their companion than for a non-social chamber. LPS treatment also increased proportion of rewards earned for the partner versus a stranger and again led to increased huddling behavior. Prior studies in other rodent species have shown decreased social interaction when sick; the present results demonstrate an alternative outcome of sickness in the context of dyadic bonds and lay the foundation for future work in peer companions.
Collapse
Affiliation(s)
- Georgia K Young
- Department of Integrative Biology, UC Berkeley, Berkeley, CA 94720, United States of America.
| | - Diana Chernyak
- Department of Integrative Biology, UC Berkeley, Berkeley, CA 94720, United States of America
| | - Gautam A Naik
- Department of Integrative Biology, UC Berkeley, Berkeley, CA 94720, United States of America
| | - Stephen Eun Song
- Department of Integrative Biology, UC Berkeley, Berkeley, CA 94720, United States of America
| | - Annaliese K Beery
- Department of Integrative Biology, UC Berkeley, Berkeley, CA 94720, United States of America; Department of Neuroscience, UC Berkeley, Berkeley, CA 94720, United States of America
| |
Collapse
|
6
|
Kang A, Ye G, Afkhami S, Aleithan F, Singh K, Dvorkin-Gheva A, Berg T, Miller MS, Jeyanathan M, Xing Z. LPS-induced lung tissue-resident trained innate immunity provides differential protection against pneumococci and SARS-CoV-2. Cell Rep 2024; 43:114849. [PMID: 39383035 DOI: 10.1016/j.celrep.2024.114849] [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: 02/23/2024] [Revised: 06/24/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024] Open
Abstract
Recent evidence indicates that tissue-resident innate immune memory and trained innate immunity (TII) can be induced centrally in myeloid cells within the bone marrow and locally in tissue-resident macrophages in respiratory mucosal tissues. However, it remains unclear whether acute exposure to airborne microbial components like lipopolysaccharide (LPS) induces lasting innate immune memory in airway macrophages and TII capable of protection against heterologous pathogens. Using a murine model, we demonstrate that acute LPS exposure leads to dynamic changes in the immune phenotype of airway macrophages that persist long after the acute inflammatory response has subsided. The original airway-resident alveolar macrophage pool remains stable in size despite these changes and the earlier transient acute inflammatory responses, including monocytic recruitment in the lung. We further demonstrate that the induction of innate immune memory in airway macrophages is accompanied by TII capable of robust protection against acute pneumococcal infection, whereas it provides minimal protection against acute SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Alisha Kang
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Gluke Ye
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Fatemah Aleithan
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Kanwaldeep Singh
- Department of Oncology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anna Dvorkin-Gheva
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Tobias Berg
- Department of Oncology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada; Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Zhou Xing
- McMaster Immunology Research Centre, Department of Medicine, and M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
7
|
Guamán SA, Elhadi A, Salama AAK, Manuelian CL, Caja G, Albanell E. Beta-Glucans Improve the Mammary Innate Immune Response to Endotoxin Challenge in Dairy Ewes. Animals (Basel) 2024; 14:3023. [PMID: 39457952 PMCID: PMC11505092 DOI: 10.3390/ani14203023] [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: 07/29/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
This study evaluated short-term immune responses of dairy ewes supplemented with barley β-glucan (BG) following an intramammary Escherichia coli lipopolysaccharide (LPS) challenge. In the adaptation period, 36 ewes were fed an alfalfa hay diet ad libitum and barley grain cv. Hispanic (3.8% BG). Then, ewes were assigned into three experimental groups: (1) Control (CON), the same previous diet (13.3 g BG/d); (2) high β-glucans barley (HBG), new barley (cv. Annapurna) containing 10% BG (35 g BG/d); (3) intraperitoneally injected (INP) with a 1.4% BG solution dose (2 g BG/ewe). At d 9, all ewes were infused with an E. coli LPS or saline solution in each udder half. After the challenge, rectal temperature (RT), milk yield and composition, somatic cell count (SCC), and plasma interleukins (IL-1α and IL-1β) were monitored daily. The INP treatment revealed a transitory increase in RT and decreased milk yield by 38%. Milk fat, protein, and SCC increased in LPS-treated udders but not by BG treatment. The IL-1α plasma concentration was similar among groups but INP ewes showed a lower IL-1β concentration suggesting a lower inflammatory response. The BG administration appears more effective intraperitoneally than orally, which needs additional study.
Collapse
Affiliation(s)
- Santiago A. Guamán
- Ruminant Research Group (G2R), Department of Animal and Food Sciences, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (S.A.G.); (A.E.); (A.A.K.S.); (C.L.M.)
- Sede Orellana, Escuela Superior Politécnica de Chimborazo (ESPOCH), El Coca 220150, Ecuador
| | - Abdelaali Elhadi
- Ruminant Research Group (G2R), Department of Animal and Food Sciences, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (S.A.G.); (A.E.); (A.A.K.S.); (C.L.M.)
| | - Ahmed A. K. Salama
- Ruminant Research Group (G2R), Department of Animal and Food Sciences, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (S.A.G.); (A.E.); (A.A.K.S.); (C.L.M.)
| | - Carmen L. Manuelian
- Ruminant Research Group (G2R), Department of Animal and Food Sciences, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (S.A.G.); (A.E.); (A.A.K.S.); (C.L.M.)
| | - Gerardo Caja
- Ruminant Research Group (G2R), Department of Animal and Food Sciences, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (S.A.G.); (A.E.); (A.A.K.S.); (C.L.M.)
| | - Elena Albanell
- Ruminant Research Group (G2R), Department of Animal and Food Sciences, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (S.A.G.); (A.E.); (A.A.K.S.); (C.L.M.)
| |
Collapse
|
8
|
Scheffzük C, Biedziak D, Gisch N, Goldmann T, Stamme C. Surfactant protein A modulates neuroinflammation in adult mice upon pulmonary infection. Brain Res 2024; 1840:149108. [PMID: 38964703 DOI: 10.1016/j.brainres.2024.149108] [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: 05/05/2024] [Revised: 06/17/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND One of the most common entry gates for systemic infection is the lung. In humans, pulmonary infections can lead to significant neurological impairment, ranging from acute sickness behavior to long-term disorders. Surfactant proteins (SP), essential parts of the pulmonary innate immune defense, have been detected in the brain of rats and humans. Recent evidence suggests that SP-A, the major protein component of surfactant, also plays a functional role in modulating neuroinflammation. This study aimed to determine whether SP-A deficiency affects the inflammatory response in the brain of adult mice during pulmonary infection. EXPERIMENTAL PROCEDURE Adult male wild-type (WT, n = 72) and SP-A-deficient (SP-A-/-, n = 72) mice were oropharyngeally challenged with lipopolysaccharide (LPS), Pseudomonas aeruginosa (P. aeruginosa), or PBS (control). Both, behavioral assessment and subsequent brain tissue analysis, were performed 24, 48, and 72 h after challenge. The brain concentrations of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β were determined by ELISA. Quantitative rtPCR was used to detect SP-A mRNA expression in brain homogenates and immunohistochemistry was applied for the detection of SP-A protein expression in brain coronal slices. RESULTS SP-A mRNA and histological evidence of protein expression were detected in both the lungs and brains of WT mice, with significantly higher amounts in lung samples. SP-A-/- mice exhibited significantly higher baseline concentrations of brain TNF-α, IL-6, and IL-1β compared to WT mice. Oropharyngeal application of either LPS or P. aeruginosa elicited significantly higher brain levels of TNF-α and IL-1β in SP-A-/- mice compared to WT mice at all time points. In comparison, behavioral impairment as a measure of sickness behavior, was significantly stronger in WT than in SP-A-/- mice, particularly after LPS application. CONCLUSION SP-A is known for its anti-inflammatory role in the pulmonary immune response to bacterial infection. Recent evidence suggests that in an abdominal sepsis model SP-A deficiency can lead to increased cytokine levels in the brain. Our results extend this perception and provide evidence for an anti-inflammatory role of SP-A in the brain of adult WT mice after pulmonary infection.
Collapse
Affiliation(s)
- Claudia Scheffzük
- Division of Cellular Pneumology, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Lübeck, Germany; Department of Anesthesiology and Intensive Care Medicine, University Hospital BG Bergmannsheil Bochum, Germany.
| | - Dominika Biedziak
- Division of Cellular Pneumology, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Torsten Goldmann
- Division of Histology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany
| | - Cordula Stamme
- Division of Cellular Pneumology, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
9
|
Wenderoth T, Feldotto M, Hernandez J, Schäffer J, Leisengang S, Pflieger FJ, Bredehöft J, Mayer K, Kang JX, Bier J, Grimminger F, Paßlack N, Rummel C. Effects of Omega-3 Polyunsaturated Fatty Acids on the Formation of Adipokines, Cytokines, and Oxylipins in Retroperitoneal Adipose Tissue of Mice. Int J Mol Sci 2024; 25:9904. [PMID: 39337391 PMCID: PMC11432517 DOI: 10.3390/ijms25189904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Oxylipins and specialized pro-resolving lipid mediators (SPMs) derived from polyunsaturated fatty acids (PUFAs) are mediators that coordinate an active process of inflammation resolution. While these mediators have potential as circulating biomarkers for several disease states with inflammatory components, the source of plasma oxylipins/SPMs remains a matter of debate but may involve white adipose tissue (WAT). Here, we aimed to investigate to what extent high or low omega (n)-3 PUFA enrichment affects the production of cytokines and adipokines (RT-PCR), as well as oxylipins/SPMs (liquid chromatography-tandem mass spectrometry) in the WAT of mice during lipopolysaccharide (LPS)-induced systemic inflammation (intraperitoneal injection, 2.5 mg/kg, 24 h). For this purpose, n-3 PUFA genetically enriched mice (FAT-1), which endogenously synthesize n-3 PUFAs, were compared to wild-type mice (WT) and combined with n-3 PUFA-sufficient or deficient diets. LPS-induced systemic inflammation resulted in the decreased expression of most adipokines and interleukin-6 in WAT, whereas the n-3-sufficient diet increased them compared to the deficient diet. The n-6 PUFA arachidonic acid was decreased in WAT of FAT-1 mice, while n-3 derived PUFAs (eicosapentaenoic acid, docosahexaenoic acid) and their metabolites (oxylipins/SPMs) were increased in WAT by genetic and nutritional n-3 enrichment. Several oxylipins/SPMs were increased by LPS treatment in WAT compared to PBS-treated controls in genetically n-3 enriched FAT-1 mice. Overall, we show that WAT may significantly contribute to circulating oxylipin production. Moreover, n-3-sufficient or n-3-deficient diets alter adipokine production. The precise interplay between cytokines, adipokines, and oxylipins remains to be further investigated.
Collapse
Affiliation(s)
- Tatjana Wenderoth
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
| | - Martin Feldotto
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
| | - Jessica Hernandez
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
| | - Julia Schäffer
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
| | - Stephan Leisengang
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
- Center for Mind Brain and Behavior (CMMB), Universities Giessen and Marburg, 34032 Marburg, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University, 35392 Giessen, Germany
| | - Fabian Johannes Pflieger
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
| | - Janne Bredehöft
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
| | - Konstantin Mayer
- Department of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany;
| | - Jing X. Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical, Charlestown, MA 02129, USA;
| | - Jens Bier
- Cardio-Pulmonary Institute, Justus Liebig University, 35392 Giessen, Germany; (J.B.); (F.G.)
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany
| | - Friedrich Grimminger
- Cardio-Pulmonary Institute, Justus Liebig University, 35392 Giessen, Germany; (J.B.); (F.G.)
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany
| | - Nadine Paßlack
- Small Animal Clinic, Internal Medicine and Department of Veterinary Clinical Sciences, Justus Liebig University, 35392 Giessen, Germany;
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, 35392 Giessen, Germany; (T.W.); (M.F.); (J.H.); (J.S.); (S.L.); (F.J.P.); (J.B.)
- Center for Mind Brain and Behavior (CMMB), Universities Giessen and Marburg, 34032 Marburg, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University, 35392 Giessen, Germany
| |
Collapse
|
10
|
Megagiannis P, Mei Y, Yan RE, Yuan L, Wilde JJ, Eckersberg H, Suresh R, Tan X, Chen H, Farmer WT, Cha K, Le PU, Catoire H, Rochefort D, Kwan T, Yee BA, Dion P, Krishnaswamy A, Cloutier JF, Stifani S, Petrecca K, Yeo GW, Murai KK, Feng G, Rouleau GA, Ideker T, Sanjana NE, Zhou Y. Autism-associated CHD8 controls reactive gliosis and neuroinflammation via remodeling chromatin in astrocytes. Cell Rep 2024; 43:114637. [PMID: 39154337 DOI: 10.1016/j.celrep.2024.114637] [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: 09/25/2023] [Revised: 06/11/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
Reactive changes of glial cells during neuroinflammation impact brain disorders and disease progression. Elucidating the mechanisms that control reactive gliosis may help us to understand brain pathophysiology and improve outcomes. Here, we report that adult ablation of autism spectrum disorder (ASD)-associated CHD8 in astrocytes attenuates reactive gliosis via remodeling chromatin accessibility, changing gene expression. Conditional Chd8 deletion in astrocytes, but not microglia, suppresses reactive gliosis by impeding astrocyte proliferation and morphological elaboration. Astrocyte Chd8 ablation alleviates lipopolysaccharide-induced neuroinflammation and septic-associated hypothermia in mice. Astrocytic CHD8 plays an important role in neuroinflammation by altering the chromatin landscape, regulating metabolic and lipid-associated pathways, and astrocyte-microglia crosstalk. Moreover, we show that reactive gliosis can be directly mitigated in vivo using an adeno-associated virus (AAV)-mediated Chd8 gene editing strategy. These findings uncover a role of ASD-associated CHD8 in the adult brain, which may warrant future exploration of targeting chromatin remodelers in reactive gliosis and neuroinflammation in injury and neurological diseases.
Collapse
Affiliation(s)
- Platon Megagiannis
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Yuan Mei
- Division of Genetics, Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Rachel E Yan
- New York Genome Center, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Lin Yuan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Jonathan J Wilde
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailey Eckersberg
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Rahul Suresh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Xinzhu Tan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Hong Chen
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - W Todd Farmer
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Kuwook Cha
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Phuong Uyen Le
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Helene Catoire
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Daniel Rochefort
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tony Kwan
- McGill Genome Center and Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Brian A Yee
- Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patrick Dion
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Jean-Francois Cloutier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Kevin Petrecca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Guoping Feng
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| | - Neville E Sanjana
- New York Genome Center, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Yang Zhou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
11
|
Kutzer MAM, Cornish B, Jamieson M, Zawistowska O, Monteith KM, Vale PF. Mitochondrial background can explain variable costs of immune deployment. J Evol Biol 2024; 37:1125-1133. [PMID: 39145390 DOI: 10.1093/jeb/voae082] [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: 08/16/2024]
Abstract
Organismal health and survival depend on the ability to mount an effective immune response against infection. Yet immune defence may be energy-demanding, resulting in fitness costs if investment in immune function deprives other physiological processes of resources. While evidence of costly immunity resulting in reduced longevity and reproduction is common, the role of energy-producing mitochondria on the magnitude of these costs is unknown. Here, we employed Drosophila melanogaster cybrid lines, where several mitochondrial genotypes (mitotypes) were introgressed onto a single nuclear genetic background, to explicitly test the role of mitochondrial variation on the costs of immune stimulation. We exposed female flies carrying one of nine distinct mitotypes to either a benign, heat-killed bacterial pathogen (stimulating immune deployment while avoiding pathology) or to a sterile control and measured lifespan, fecundity, and locomotor activity. We observed mitotype-specific costs of immune stimulation and identified a positive genetic correlation in immune-stimulated flies between lifespan and the proportion of time cybrids spent moving while alive. Our results suggests that costs of immunity are highly variable depending on the mitochondrial genome, adding to a growing body of work highlighting the important role of mitochondrial variation in host-pathogen interactions.
Collapse
Affiliation(s)
- Megan A M Kutzer
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Beth Cornish
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Jamieson
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Olga Zawistowska
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Katy M Monteith
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Pedro F Vale
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
12
|
Mishra K, Khatib N, Barasch D, Kumar P, Garti S, Garti N, Kakhlon O. A New Tailored Nanodroplet Carrier of Astaxanthin Can Improve Its Pharmacokinetic Profile and Antioxidant and Anti-Inflammatory Efficacies. Int J Mol Sci 2024; 25:7861. [PMID: 39063101 PMCID: PMC11276774 DOI: 10.3390/ijms25147861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Astaxanthin (ATX) is a carotenoid nutraceutical with poor bioavailability due to its high lipophilicity. We tested a new tailored nanodroplet capable of solubilizing ATX in an oil-in-water micro-environment (LDS-ATX) for its capacity to improve the ATX pharmacokinetic profile and therapeutic efficacy. We used liquid chromatography tandem mass spectrometry (LC-MS/MS) to profile the pharmacokinetics of ATX and LDS-ATX, superoxide mutase (SOD) activity to determine their antioxidant capacity, protein carbonylation and lipid peroxidation to compare their basal and lipopolysaccharide (LPS)-induced oxidative damage, and ELISA-based detection of IL-2 and IFN-γ to determine their anti-inflammatory capacity. ATX and LDS-ATX corrected only LPS-induced SOD inhibition and oxidative damage. SOD activity was restored only by LDS-ATX in the liver and brain and by both ATX and LDS-ATX in muscle. While in the liver and muscle, LDS-ATX attenuated oxidative damage to proteins and lipids better than ATX; only oxidative damage to lipids was preferably corrected by LDS-ATX in the brain. IL-2 and IFN-γ pro-inflammatory response was corrected by LDS-ATX and not ATX in the liver and brain, but in muscle, the IL-2 response was not corrected and the IFN-γ response was mitigated by both. These results strongly suggest an organ-dependent improvement of ATX bioavailability and efficacy by the LDS-ATX nanoformulation.
Collapse
Affiliation(s)
- Kumudesh Mishra
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (K.M.); (P.K.)
| | - Nadin Khatib
- Lyotropic Delivery Systems Ltd., Hi-Tech Park, Row 5(1), Edmond J. Safra Campus, Jerusalem 9139002, Israel; (N.K.); (S.G.)
| | - Dinorah Barasch
- Mass Spectrometry Unit, Institute for Drug Research, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Pradeep Kumar
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (K.M.); (P.K.)
| | - Sharon Garti
- Lyotropic Delivery Systems Ltd., Hi-Tech Park, Row 5(1), Edmond J. Safra Campus, Jerusalem 9139002, Israel; (N.K.); (S.G.)
| | - Nissim Garti
- Lyotropic Delivery Systems Ltd., Hi-Tech Park, Row 5(1), Edmond J. Safra Campus, Jerusalem 9139002, Israel; (N.K.); (S.G.)
| | - Or Kakhlon
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (K.M.); (P.K.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| |
Collapse
|
13
|
Li B, Shi X, Chen E, Wu X. Improvement effects of cyclic peptides from Annona squamosa on cognitive decline in neuroinflammatory mice. Food Sci Biotechnol 2024; 33:1437-1448. [PMID: 38585570 PMCID: PMC10992170 DOI: 10.1007/s10068-023-01441-8] [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: 04/23/2023] [Revised: 08/26/2023] [Accepted: 09/19/2023] [Indexed: 04/09/2024] Open
Abstract
Cyclic peptides can resist enzymatic hydrolysis to pass through the intestine barrier, which may reduce the risk of mild cognition decline. But evidence is lacking on whether they work by alleviating neuroinflammation. A cylic peptide from Annona squamosa, Cylic(PIYAG), was biologically evaluated in vivo and in vitro. Cylic(PIYAG) enhanced the spatial memory ability of LPS-induced mice. And treatment with Cylic(PIYAG) markedly reduced the iNOS, MCP-1, TNF-α, and gp91phox expression induced by LPS. Cylic(PIYAG, 0.01, 0.05 and 0.2 μM) could significantly reduce the protein expression level of COX-2 and iNOS (P < 0.05) in BV2 cells. The concentration of Cylic(PIYAG) in blood reached a peak of 3.64 ± 1.22 μg/ml after intragastric administration in 1 h. And fluorescence microscope shows that Cylic(PIYAG) mainly locates and may play an anti-inflammatory role in the cytoplasm of microglia. This study demonstrates that the peptidic can prevent microglia activation, decrease the inflammatory reaction, improve the cognition of LPS-induced mice. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01441-8.
Collapse
Affiliation(s)
- Bo Li
- Neurology Department, Anqing Medical Center, Anhui Medical University, Anqing, China
| | - Xueying Shi
- Neurology Department, Anqing Medical Center, Anhui Medical University, Anqing, China
| | - Erhua Chen
- Clinical Nutrition Department, Anqing Hospital Affiliated to Anhui Medical University, Anqing, 246000 Anhui China
| | - Xiaocui Wu
- Department of Neurology, Graduate School, Anhui Medical University, Hefei, 230000 Anhui China
| |
Collapse
|
14
|
Şahin S, Şahin E, Esenülkü G, Renda G, Gürgen SG, Alver A, Abidin İ, Cansu A. Oleuropein Has Modulatory Effects on Systemic Lipopolysaccharide-Induced Neuroinflammation in Male Rats. J Nutr 2024; 154:1282-1297. [PMID: 38403251 DOI: 10.1016/j.tjnut.2024.02.017] [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: 10/25/2023] [Revised: 01/20/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Neuroinflammation induced by systemic inflammation is a risk factor for developing chronic neurologic disorders. Oleuropein (OLE) has antioxidant and anti-inflammatory properties; however, its effect on systemic inflammation-related neuroinflammation is unknown. OBJECTIVES This study aimed to determine whether OLE protects against systemic lipopolysaccharide (LPS)-induced neuroinflammation in rats. METHODS Six-wk-old Wistar rats were randomly assigned to 1 of the following 5 groups: 1) control, 2) OLE-only, 3) LPS + vehicle, 4) OLE+LPS (O-LPS), and 5) a single-dose OLE + LPS (SO-LPS group). OLE 200 mg/kg or saline as a vehicle was administered via gavage for 7 d. On the seventh day, 2.5 mg/kg LPS was intraperitoneally administered. The rats were decapitated after 24 h of LPS treatment, and serum collection and tissue dissection were performed. The study assessed astrocyte and microglial activation using glial fibrillary acidic protein (GFAP) and CD11b immunohistochemistry, nod-like receptor protein-3, interleukin (IL)-1β, IL-17A, and IL-4 concentrations in prefrontal and hippocampal tissues via enzyme-linked immunosorbent assay, and total antioxidant/oxidant status (TAS/TOS) in serum and tissues via spectrophotometry. RESULTS In both the O-LPS and SO-LPS groups, LPS-related activation of microglia and astrocytes was suppressed in the cortex and hippocampus (P < 0.001), excluding cortical astrocyte activation, which was suppressed only in the SO-LPS group (P < 0.001). Hippocampal GFAP immunoreactivity and IL-17A concentrations in the dentate gyrus were higher in the OLE group than those in the control group, but LPS-related increases in these concentrations were suppressed in the O-LPS group. The O-LPS group had higher cortical TAS and IL-4 concentrations. CONCLUSIONS OLE suppressed LPS-related astrocyte and microglial activation in the hippocampus and cortex. The OLE-induced increase in cortical IL-4 concentrations indicates the induction of an anti-inflammatory phenotype of microglia. OLE may also modulate astrocyte and IL-17A functions, which could explain its opposing effects on hippocampal GFAP immunoreactivity and IL-17A concentrations when administered with or without LPS.
Collapse
Affiliation(s)
- Sevim Şahin
- Department of Pediatric Neurology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| | - Elif Şahin
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Gülnur Esenülkü
- Department of Pediatric Neurology, Trabzon Kanuni Training, and Research Hospital, Trabzon, Turkey
| | - Gülin Renda
- Department of Pharmacognosy, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkey
| | - Seren Gülşen Gürgen
- Department of Histology and Embryology, School of Vocational Health Service, Manisa Celal Bayar University, Manisa, Turkey
| | - Ahmet Alver
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - İsmail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ali Cansu
- Department of Pediatric Neurology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
15
|
Vázquez-Sánchez A, Rodríguez-Ríos D, Colín-Castelán D, Molina-Torres J, Ramírez-Chávez E, Romo-Morales GDC, Zaina S, Lund G. Effects of paternal arachidonic acid supplementation on offspring behavior and hypothalamus inflammation markers in the mouse. PLoS One 2024; 19:e0300141. [PMID: 38512839 PMCID: PMC10956830 DOI: 10.1371/journal.pone.0300141] [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: 12/21/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Arachidonic acid (AA) is involved in inflammation and plays a role in growth and brain development in infants. We previously showed that exposure of mouse sires to AA for three consecutive generations induces a cumulative change in fatty acid (FA) involved in inflammation and an increase in body and liver weight in the offspring. Here, we tested the hypothesis that paternal AA exposure changes the progeny's behavioral response to a proinflammatory insult, and asked whether tissue-specific FA are associated with that response. Male BALB/c mice were supplemented daily with three doses of AA for 10 days and crossed to non-supplemented females (n = 3/dose). Two-month-old unsupplemented male and female offspring (n = 6/paternal AA dose) were exposed to Gram-negative bacteria-derived lipopolysaccharides (LPS) or saline control two hours prior to open field test (OFT) behavioral analysis and subsequent sacrifice. We probed for significant effects of paternal AA exposure on: OFT behaviors; individual FA content of blood, hypothalamus and hypothalamus-free brain; hypothalamic expression profile of genes related to inflammation (Tnfa, Il1b, Cox1, Cox2) and FA synthesis (Scd1, Elovl6). All parameters were affected by paternal AA supplementation in a sex-specific manner. Paternal AA primed the progeny for behavior associated with increased anxiety, with a marked sex dimorphism: high AA doses acted as surrogate of LPS in males, realigning a number of OFT behaviors that in females were differential between saline and LPS groups. Progeny hypothalamic Scd1, a FA metabolism enzyme with documented pro-inflammatory activity, showed a similar pattern of differential expression between saline and LPS groups at high paternal AA dose in females, that was blunted in males. Progeny FA generally were not affected by LPS, but displayed non-linear associations with paternal AA doses. In conclusion, we document that paternal exposure to AA exerts long-term behavioral and biochemical effects in the progeny in a sex-specific manner.
Collapse
Affiliation(s)
| | | | - Dannia Colín-Castelán
- Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, Leon Campus, Leon, Gto., Mexico
| | - Jorge Molina-Torres
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Irapuato, Mexico
| | | | | | - Silvio Zaina
- Division of Health Sciences, Department of Medical Sciences, University of Guanajuato, Leon Campus, Leon, Gto., Mexico
| | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Irapuato, Mexico
| |
Collapse
|
16
|
Dunstan IK, McLeod R, Radford-Smith DE, Xiong W, Pate T, Probert F, Anthony DC. Unique pathways downstream of TLR-4 and TLR-7 activation: sex-dependent behavioural, cytokine, and metabolic consequences. Front Cell Neurosci 2024; 18:1345441. [PMID: 38414751 PMCID: PMC10896997 DOI: 10.3389/fncel.2024.1345441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
Introduction Post-infection syndromes are characterised by fatigue, muscle pain, anhedonia, and cognitive impairment; mechanistic studies exploring these syndromes have focussed on pathways downstream of Toll-like receptor (TLR) 4 activation. Here, we investigated the mechanistic interplay between behaviour, metabolism, and inflammation downstream of TLR-7 activation compared to TLR-4 activation in male and female CD1 mice. Methods Animals received either a TLR-4 (LPS; 0.83 mg/kg) or TLR-7 (R848, 5 mg/kg) agonist, or saline, and behaviour was analysed in an Open Field (OF) at 24 h (n = 20/group). Plasma, liver, and prefrontal cortex (PFC) were collected for gene expression analysis at 24 h and 1H-NMR metabolomics. Results TLR-4 and TLR-7 activation decreased distance travelled and rearing in the OF, but activation of each receptor induced distinct cytokine responses and metabolome profiles. LPS increased IL-1β expression and CXCL1 in the PFC, but TLR7 activation did not and strongly induced PFC CXCL10 expression. Thus, TLR7 induced sickness behaviour is independent of IL-1β expression. In both cases, the behavioural response to TLR activation was sexually dimorphic: females were more resilient. However, dissociation was observed between the resilient female mice behaviour and the levels of gene cytokine expression, which was, in general, higher in the female mice. However, the metabolic shifts induced by immune activation were better correlated with the sex-dependent behavioural dimorphisms; increased levels of antioxidant potential in the female brain are intrinsic male/female metabolome differences. A common feature of both TLR4 and TLR7 activation was an increase in N-acetyl aspartate (NAA) in the PFC, which is likely be an allostatic response to the challenges as sickness behaviour is inversely correlated with NAA levels. Discussion The results highlight how the cytokine profile induced by one PAMP cannot be extrapolated to another, but they do reveal how the manipulation of the conserved metabolome response might afford a more generic approach to the treatment of post-infection syndromes.
Collapse
Affiliation(s)
- Isobel K. Dunstan
- Medical Sciences Division, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Chemistry, Mathematical, Physical and Life Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Ross McLeod
- Medical Sciences Division, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Chemistry, Mathematical, Physical and Life Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Daniel E. Radford-Smith
- Medical Sciences Division, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Wenzheng Xiong
- Medical Sciences Division, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Chemistry, Mathematical, Physical and Life Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Trinity Pate
- Medical Sciences Division, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Fay Probert
- Medical Sciences Division, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Chemistry, Mathematical, Physical and Life Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Daniel C. Anthony
- Medical Sciences Division, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Volk Robertson K, Schleh MW, Harrison FE, Hasty AH. Microglial-specific knockdown of iron import gene, Slc11a2, blunts LPS-induced neuroinflammatory responses in a sex-specific manner. Brain Behav Immun 2024; 116:370-384. [PMID: 38141840 PMCID: PMC10874246 DOI: 10.1016/j.bbi.2023.12.020] [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: 07/17/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023] Open
Abstract
Neuroinflammation and microglial iron load are significant hallmarks found in several neurodegenerative diseases. In in vitro systems, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and it has been shown that iron can augment cellular inflammation, suggesting a feed-forward loop between mechanisms involved in iron import and inflammatory signaling. However, it is not understood how microglial iron import mechanisms contribute to inflammation in vivo, or whether altering a microglial iron-related gene affects the inflammatory response. These studies aimed to determine the effect of knocking down microglial iron import gene Slc11a2 on the inflammatory response in vivo. We generated a novel model of tamoxifen-inducible, microglial-specific Slc11a2 knockdown using Cx3cr1Cre-ERT2 mice. Transgenic male and female mice were administered intraperitoneal saline or lipopolysaccharide (LPS) and assessed for sickness behavior post-injection. Plasma cytokines and microglial bulk RNA sequencing (RNASeq) analyses were performed at 4 h post-LPS, and microglia were collected for gene expression analysis after 24 h. A subset of mice was assessed in a behavioral test battery following LPS-induced sickness recovery. Control male, but not female, mice significantly upregulated microglial Slc11a2 at 4 and 24 h following LPS. In Slc11a2 knockdown mice, we observed an improvement in the acute behavioral sickness response post-LPS in male, but not female, animals. Microglia from male, but not female, knockdown animals exhibited a significant decrease in LPS-provoked pro-inflammatory cytokine expression after 24 h. RNASeq data from male knockdown microglia 4 h post-LPS revealed a robust downregulation in inflammatory genes including Il6, Tnfα, and Il1β, and an increase in anti-inflammatory and homeostatic markers (e.g., Tgfbr1, Cx3cr1, and Trem2). This corresponded with a profound decrease in plasma pro-inflammatory cytokines 4 h post-LPS. At 4 h, male knockdown microglia also upregulated expression of markers of iron export, iron recycling, and iron homeostasis and decreased iron storage and import genes, along with pro-oxidant markers such as Cybb, Nos2, and Hif1α. Overall, this work elucidates how manipulating a specific gene involved in iron import in microglia alters acute inflammatory signaling and overall cell activation state in male mice. These data highlight a sex-specific link between a microglial iron import gene and the pro-inflammatory response to LPS in vivo, providing further insight into the mechanisms driving neuroinflammatory disease.
Collapse
Affiliation(s)
- Katrina Volk Robertson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
18
|
Shamloo S, Defensor E, Ciari P, Ogawa G, Vidano L, Lin JS, Fortkort JA, Shamloo M, Barron AE. The anti-inflammatory effects of photobiomodulation are mediated by cytokines: Evidence from a mouse model of inflammation. Front Neurosci 2023; 17:1150156. [PMID: 37090796 PMCID: PMC10115964 DOI: 10.3389/fnins.2023.1150156] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/13/2023] [Indexed: 04/25/2023] Open
Abstract
There is an urgent need for therapeutic approaches that can prevent or limit neuroinflammatory processes and prevent neuronal degeneration. Photobiomodulation (PBM), the therapeutic use of specific wavelengths of light, is a safe approach shown to have anti-inflammatory effects. The current study was aimed at evaluating the effects of PBM on LPS-induced peripheral and central inflammation in mice to assess its potential as an anti-inflammatory treatment. Daily, 30-min treatment of mice with red/NIR light (RL) or RL with a 40 Hz gamma frequency flicker for 10 days prior to LPS challenge showed anti-inflammatory effects in the brain and systemically. PBM downregulated LPS induction of key proinflammatory cytokines associated with inflammasome activation, IL-1β and IL-18, and upregulated the anti-inflammatory cytokine, IL-10. RL provided robust anti-inflammatory effects, and the addition of gamma flicker potentiated these effects. Overall, these results demonstrate the potential of PBM as an anti-inflammatory treatment that acts through cytokine expression modulation.
Collapse
Affiliation(s)
- Shirin Shamloo
- Department of Bioengineering, Schools of Medicine and Engineering, Stanford University, Stanford, CA, United States
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Erwin Defensor
- Department of Bioengineering, Schools of Medicine and Engineering, Stanford University, Stanford, CA, United States
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Peter Ciari
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Gaku Ogawa
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Laura Vidano
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jennifer S. Lin
- Department of Bioengineering, Schools of Medicine and Engineering, Stanford University, Stanford, CA, United States
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - John A. Fortkort
- Department of Bioengineering, Schools of Medicine and Engineering, Stanford University, Stanford, CA, United States
| | - Mehrdad Shamloo
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Mehrdad Shamloo,
| | - Annelise E. Barron
- Department of Bioengineering, Schools of Medicine and Engineering, Stanford University, Stanford, CA, United States
- Annelise E. Barron,
| |
Collapse
|
19
|
Fruekilde SK, Bailey CJ, Lambertsen KL, Clausen BH, Carlsen J, Xu NL, Drasbek KR, Gutiérrez-Jiménez E. Disturbed microcirculation and hyperaemic response in a murine model of systemic inflammation. J Cereb Blood Flow Metab 2022; 42:2303-2317. [PMID: 35999817 PMCID: PMC9670001 DOI: 10.1177/0271678x221112278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systemic inflammation affects cognitive functions and increases the risk of dementia. This phenomenon is thought to be mediated in part by cytokines that promote neuronal survival, but the continuous exposure to which may lead to neurodegeneration. The effects of systemic inflammation on cerebral blood vessels, and their provision of adequate oxygen to support critical brain parenchymal cell functions, remains unclear. Here, we demonstrate that neurovascular coupling is profoundly disturbed in lipopolysaccharide (LPS) induced systemic inflammation in awake mice. In the 24 hours following LPS injection, the hyperaemic response of pial vessels to functional activation was attenuated and delayed. Concurrently, under steady-state conditions, the capillary network displayed a significant increase in the number of capillaries with blocked blood flow, as well as increased duration of 'capillary stalls'-a phenomenon previously reported in animal models of stroke and Alzheimer's disease pathology. We speculate that vascular changes and impaired oxygen availability may affect brain functions following acute systemic inflammation and contribute to the long-term risk of neurodegenerative changes associated with chronic, systemic inflammation.
Collapse
Affiliation(s)
- Signe Kirk Fruekilde
- Center for Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, 1006Aarhus University, Aarhus C, Denmark.,Sino-Danish Center for Education and Research (SDC), University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Christopher J Bailey
- Center for Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, 1006Aarhus University, Aarhus C, Denmark.,Sino-Danish Center for Education and Research (SDC), University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, 6174University of Southern Denmark, Odense C, Denmark.,BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, 6174University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Odense University Hospital, Odense C, Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, 6174University of Southern Denmark, Odense C, Denmark.,BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, 6174University of Southern Denmark, Odense C, Denmark
| | - Jasper Carlsen
- Research Unit for Molecular Medicine (MMF), Department of Clinical Medicine, 1006Aarhus University, Aarhus N, Denmark
| | - Ning-Long Xu
- Sino-Danish Center for Education and Research (SDC), University of Chinese Academy of Sciences, Beijing, P.R. China.,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Kim Ryun Drasbek
- Center for Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, 1006Aarhus University, Aarhus C, Denmark.,Sino-Danish Center for Education and Research (SDC), University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Eugenio Gutiérrez-Jiménez
- Center for Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, 1006Aarhus University, Aarhus C, Denmark
| |
Collapse
|
20
|
Deng I, Bobrovskaya L. Lipopolysaccharide mouse models for Parkinson's disease research: a critical appraisal. Neural Regen Res 2022; 17:2413-2417. [PMID: 35535880 PMCID: PMC9120679 DOI: 10.4103/1673-5374.331866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/25/2022] Open
Abstract
Parkinson's disease, the most common movement disorder, has a strong neuroinflammatory aspect. This is evident by increased pro-inflammatory cytokines in the serum, and the presence of activated microglial cells, and inflammatory cytokines in the substantia nigra of post-mortem brains as well as cerebrospinal fluid of Parkinson's disease patients. The central and peripheral neuroinflammatory aspects of Parkinson's disease can be investigated in vivo via administration of the inflammagen lipopolysaccharide, a component of the cell wall of gram-negative bacteria. In this mini-review, we will critically evaluate different routes of lipopolysaccharide administration (including intranasal systemic and stereotasic), their relevance to clinical Parkinson's disease as well as the recent findings in lipopolysaccharide mouse models. We will also share our own experiences with systemic and intrastriatal lipopolysaccharide models in C57BL/6 mice and will discuss the usefulness of lipopolysaccharide mouse models for future research in the field.
Collapse
Affiliation(s)
- Isaac Deng
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
21
|
Coales I, Tsartsalis S, Fancy N, Weinert M, Clode D, Owen D, Matthews PM. Alzheimer's disease-related transcriptional sex differences in myeloid cells. J Neuroinflammation 2022; 19:247. [PMID: 36199077 PMCID: PMC9535846 DOI: 10.1186/s12974-022-02604-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
Sex differences have been identified in many diseases associated with dysregulated immune responses, including Alzheimer's disease (AD), for which approximately two-thirds of patients are women. An accumulating body of research indicates that microglia may play a causal role in the pathogenesis of this disease. We hypothesised that sex differences in the transcriptome of human myeloid cells may contribute to the sex difference observed in AD prevalence. To explore this, we assessed bulk and single-nuclear RNA sequencing data sets generated from four human derived myeloid cell populations: post-mortem microglial nuclei, peripheral monocytes, monocyte-derived macrophages (MDMs) and induced pluripotent stem cell derived microglial-like cells (MGLs). We found that expression of AD risk genes, gene signatures associated with the inflammatory response in AD, and genes related to proinflammatory immune responses were enriched in microglial nuclei isolated from aged female donors without ante-mortem neurological disease, relative to those from males. In addition, these inflammation-associated gene sets were found to be enriched in peripheral monocytes isolated from postmenopausal women and in MDMs obtained from premenopausal individuals relative to age-matched males. Expression of these gene sets did not differ in MDMs derived from women whose blood was sampled across the menstrual cycle or in MGLs cultured with 17β-oestradiol. This suggests that the observed gene set enrichments in myeloid cells from women were not being driven by acute hormonal influences. Together, these data support the hypothesis that the increased prevalence of AD in women may be partly explained by a myeloid cell phenotype biased towards expression of biological processes relevant to AD.
Collapse
Affiliation(s)
- Isabelle Coales
- Department of Brain Sciences, Imperial College London, London, UK
- Centre for Host Microbiome Interactions, King's College London, London, SE1 9RT, UK
| | - Stergios Tsartsalis
- Department of Brain Sciences, Imperial College London, London, UK
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Nurun Fancy
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Centre at Imperial College London, London, UK
| | - Maria Weinert
- Department of Brain Sciences, Imperial College London, London, UK
| | - Daniel Clode
- UK Dementia Research Centre at Imperial College London, London, UK
| | - David Owen
- Department of Brain Sciences, Imperial College London, London, UK.
- Clinical Research Facility, Hammersmith Hospital, ICTM Building, DuCane Road, London, W12 0NN, UK.
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Centre at Imperial College London, London, UK.
- Hammersmith Hospital, E502, Burlington Danes Building, DuCane Road, London, W12 0NN, UK.
| |
Collapse
|
22
|
Sosnowski DK, Jamieson KL, Gruzdev A, Li Y, Valencia R, Yousef A, Kassiri Z, Zeldin DC, Seubert JM. Cardiomyocyte-specific disruption of soluble epoxide hydrolase limits inflammation to preserve cardiac function. Am J Physiol Heart Circ Physiol 2022; 323:H670-H687. [PMID: 35985007 PMCID: PMC9512117 DOI: 10.1152/ajpheart.00217.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022]
Abstract
Endotoxemia elicits a multiorgan inflammatory response that results in cardiac dysfunction and often leads to death. Inflammation-induced metabolism of endogenous N-3 and N-6 polyunsaturated fatty acids generates numerous lipid mediators, such as epoxy fatty acids (EpFAs), which protect the heart. However, EpFAs are hydrolyzed by soluble epoxide hydrolase (sEH), which attenuates their cardioprotective actions. Global genetic disruption of sEH preserves EpFA levels and attenuates cardiac dysfunction in mice following acute lipopolysaccharide (LPS)-induced inflammatory injury. In leukocytes, EpFAs modulate the innate immune system through the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. However, the mechanisms by which both EpFAs and sEH inhibition exert their protective effects in the cardiomyocyte are still elusive. This study investigated whether cardiomyocyte-specific sEH disruption attenuates inflammation and cardiac dysfunction in acute LPS inflammatory injury via modulation of the NLRP3 inflammasome. We use tamoxifen-inducible CreER recombinase technology to target sEH genetic disruption to the cardiomyocyte. Primary cardiomyocyte studies provide mechanistic insight into inflammasome signaling. For the first time, we demonstrate that cardiomyocyte-specific sEH disruption preserves cardiac function and attenuates inflammatory responses by limiting local cardiac inflammation and activation of the systemic immune response. Mechanistically, inhibition of cardiomyocyte-specific sEH activity or exogenous EpFA treatment do not prevent upregulation of NLRP3 inflammasome machinery in neonatal rat cardiomyocytes. Rather, they limit downstream activation of the pathway leading to release of fewer chemoattractant factors and recruitment of immune cells to the heart. These data emphasize that cardiomyocyte sEH is vital for mediating detrimental systemic inflammation.NEW & NOTEWORTHY The cardioprotective effects of genetic disruption and pharmacological inhibition of sEH have been demonstrated in a variety of cardiac disease models, including acute LPS inflammatory injury. For the first time, it has been demonstrated that sEH genetic disruption limited to the cardiomyocyte profoundly preserves cardiac function and limits local and systemic inflammation following acute LPS exposure. Hence, cardiomyocytes serve a critical role in the innate immune response that can be modulated to protect the heart.
Collapse
Affiliation(s)
- Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Artiom Gruzdev
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Yingxi Li
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Robert Valencia
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Darryl C Zeldin
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
23
|
Schapovalova O, Gorlova A, de Munter J, Sheveleva E, Eropkin M, Gorbunov N, Sicker M, Umriukhin A, Lyubchyk S, Lesch KP, Strekalova T, Schroeter CA. Immunomodulatory effects of new phytotherapy on human macrophages and TLR4- and TLR7/8-mediated viral-like inflammation in mice. Front Med (Lausanne) 2022; 9:952977. [PMID: 36091684 PMCID: PMC9450044 DOI: 10.3389/fmed.2022.952977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background While all efforts have been undertaken to propagate the vaccination and develop remedies against SARS-CoV-2, no satisfactory management of this infection is available yet. Moreover, poor availability of any preventive and treatment measures of SARS-CoV-2 in economically disadvantageous communities aggravates the course of the pandemic. Here, we studied a new immunomodulatory phytotherapy (IP), an extract of blackberry, chamomile, garlic, cloves, and elderberry as a potential low-cost solution for these problems given the reported efficacy of herbal medicine during the previous SARS virus outbreak. Methods The key feature of SARS-CoV-2 infection, excessive inflammation, was studied in in vitro and in vivo assays under the application of the IP. First, changes in tumor-necrosis factor (TNF) and lnteurleukin-1 beta (IL-1β) concentrations were measured in a culture of human macrophages following the lipopolysaccharide (LPS) challenge and treatment with IP or prednisolone. Second, chronically IP-pre-treated CD-1 mice received an agonist of Toll-like receptors (TLR)-7/8 resiquimod and were examined for lung and spleen expression of pro-inflammatory cytokines and blood formula. Finally, chronically IP-pre-treated mice challenged with LPS injection were studied for "sickness" behavior. Additionally, the IP was analyzed using high-potency-liquid chromatography (HPLC)-high-resolution-mass-spectrometry (HRMS). Results LPS-induced in vitro release of TNF and IL-1β was reduced by both treatments. The IP-treated mice displayed blunted over-expression of SAA-2, ACE-2, CXCL1, and CXCL10 and decreased changes in blood formula in response to an injection with resiquimod. The IP-treated mice injected with LPS showed normalized locomotion, anxiety, and exploration behaviors but not abnormal forced swimming. Isoquercitrin, choline, leucine, chlorogenic acid, and other constituents were identified by HPLC-HRMS and likely underlie the IP immunomodulatory effects. Conclusions Herbal IP-therapy decreases inflammation and, partly, "sickness behavior," suggesting its potency to combat SARS-CoV-2 infection first of all via its preventive effects.
Collapse
Affiliation(s)
- Olesia Schapovalova
- Caparica Faculdade de Ciencias e Tecnologia da Universidade Nova de Lisboa, NOVA Lisbon University, Lisbon, Portugal
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
| | - Anna Gorlova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Johannes de Munter
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
| | - Elisaveta Sheveleva
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Mikhail Eropkin
- Department of Etiology and Epidemiology, Smorodintsev Research Institute of Influenza, St. Petersburg State University, Saint Petersburg, Russia
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Michail Sicker
- Rehabilitation Research Unit of Clinic of Bad Kreuzbach, Bad Kreuzbach, Germany
| | - Aleksei Umriukhin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sergiy Lyubchyk
- Caparica Faculdade de Ciencias e Tecnologia da Universidade Nova de Lisboa, NOVA Lisbon University, Lisbon, Portugal
- EIGES Center, Universidade Lusofona, Lisboa, Portugal
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
24
|
Kisipan ML, Ojoo RO, Kanui TI, Abelson KSP. Bodyweight, locomotion, and behavioral responses of the naked mole rat (Heterocephalus glaber) to lipopolysaccharide administration. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2022; 208:493-504. [PMID: 35731263 PMCID: PMC9250917 DOI: 10.1007/s00359-022-01557-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/10/2022] [Indexed: 11/26/2022]
Abstract
The naked mole rat has unique biologic characteristics that include atypical inflammatory responses. Lipopolysaccharide induces inflammation which triggers brain centers controlling feeding, and behavior to result in “sick animal behavior”. We characterized the bodyweight, locomotor, and other behavioral responses of this rodent to lipopolysaccharide administration. Lipopolysaccharide caused weight losses, which were not prevented by TAK 242. In the open field test, lipopolysaccharide did not depress locomotion, while urination, defecation, and activity freezing were rare. The animals exhibited walling but not rearing and fast backward movements that were unaffected by lipopolysaccharide. Failure to depress locomotion suggests either a unique immunity-brain crosstalk or motor responses/centers that tolerate depressive effects of inflammation. The absence of activity freezing and rarity of urination and defecation suggests that novel environments or lipopolysaccharide do not induce anxiety, or that anxiety is expressed differently in the animal. The absence of rearing could be due to the design of the animal’s locomotor apparatus while fast backward movement could be a mechanism for quick escape from threats in the tunnels of their habitat. Our results elucidate the unique biology of this rodent, which elicits interest in the animal as a model for inflammatory research, although the findings require mechanistic corroborations.
Collapse
Affiliation(s)
- Mosiany Letura Kisipan
- Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark.
- Department of Veterinary Anatomy and Physiology, Egerton University, Njoro, Kenya.
| | - Rodi Omondi Ojoo
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Titus Ikusya Kanui
- Department of Agricultural Sciences, South Eastern Kenya University, Kitui, Kenya
| | - Klas S P Abelson
- Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Sharma R, Casillas-Espinosa PM, Dill LK, Rewell SSJ, Hudson MR, O'Brien TJ, Shultz SR, Semple BD. Pediatric traumatic brain injury and a subsequent transient immune challenge independently influenced chronic outcomes in male mice. Brain Behav Immun 2022; 100:29-47. [PMID: 34808288 DOI: 10.1016/j.bbi.2021.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 01/30/2023] Open
Abstract
Traumatic brain injury (TBI) is a major contributor to death and disability worldwide. Children are at particularly high risk of both sustaining a TBI and experiencing serious long-term consequences, such as cognitive deficits, mental health problems and post-traumatic epilepsy. Severe TBI patients are highly susceptible to nosocomial infections, which are mostly acquired within the first week of hospitalization post-TBI. Yet the potential chronic impact of such acute infections following pediatric TBI remains unclear. In this study, we hypothesized that a peripheral immune challenge, such as lipopolysaccharide (LPS)-mimicking a hospital-acquired infection-would worsen inflammatory, neurobehavioral, and seizure outcomes after experimental pediatric TBI. To test this, three-week old male C57Bl/6J mice received a moderate controlled cortical impact or sham surgery, followed by 1 mg/kg i.p. LPS (or 0.9% saline vehicle) at 4 days TBI. Mice were randomized to four groups; sham-saline, sham-LPS, TBI-saline or TBI-LPS (n = 15/group). Reduced general activity and increased anxiety-like behavior were observed within 24 h in LPS-treated mice, indicating a transient sickness response. LPS-treated mice also exhibited a reduction in body weights, which persisted chronically. From 2 months post-injury, mice underwent a battery of tests for sensorimotor, cognitive, and psychosocial behaviors. TBI resulted in hyperactivity and spatial memory deficits, independent of LPS; whereas LPS resulted in subtle deficits in spatial memory retention. At 5 months post-injury, video-electroencephalographic recordings were obtained to evaluate both spontaneous seizure activity as well as the evoked seizure response to pentylenetetrazol (PTZ). TBI increased susceptibility to PTZ-evoked seizures; whereas LPS appeared to increase the incidence of spontaneous seizures. Post-mortem analyses found that TBI, but not LPS, resulted in robust glial reactivity and loss of cortical volume. A TBI × LPS interaction in hippocampal volume suggested that TBI-LPS mice had a subtle increase in ipsilateral hippocampus tissue loss; however, this was not reflected in neuronal cell counts. Both TBI and LPS independently had modest effects on chronic hippocampal gene expression. Together, contrary to our hypothesis, we observed minimal synergy between TBI and LPS. Instead, pediatric TBI and a subsequent transient immune challenge independently influenced chronic outcomes. These findings have implications for future preclinical modeling as well as acute post-injury patient management.
Collapse
Affiliation(s)
- Rishabh Sharma
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Larissa K Dill
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Sarah S J Rewell
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Matthew R Hudson
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Prahran, VIC, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
26
|
Piergallini TJ, Scordo JM, Pino PA, Schlesinger LS, Torrelles JB, Turner J. Acute Inflammation Confers Enhanced Protection against Mycobacterium tuberculosis Infection in Mice. Microbiol Spectr 2021; 9:e0001621. [PMID: 34232086 PMCID: PMC8552513 DOI: 10.1128/spectrum.00016-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/02/2021] [Indexed: 01/02/2023] Open
Abstract
Inflammation plays a crucial role in the control of Mycobacterium tuberculosis infection. In this study, we demonstrate that an inflammatory pulmonary environment at the time of infection mediated by lipopolysaccharide treatment in mice confers enhanced protection against M. tuberculosis for up to 6 months postinfection. This early and transient inflammatory environment was associated with a neutrophil and CD11b+ cell influx and increased inflammatory cytokines. In vitro infection demonstrated that neutrophils from lipopolysaccharide-treated mice exhibited increased association with M. tuberculosis and had a greater innate capacity for killing M. tuberculosis. Finally, partial depletion of neutrophils in lipopolysaccharide-treated mice showed an increase in M. tuberculosis burden, suggesting neutrophils played a part in the protection observed in lipopolysaccharide-treated mice. These results indicate a positive role for an inflammatory environment in the initial stages of M. tuberculosis infection and suggest that acute inflammation at the time of M. tuberculosis infection can positively alter disease outcome. IMPORTANCE Mycobacterium tuberculosis, the causative agent of tuberculosis disease, is estimated to infect one-fourth of the world's population and is one of the leading causes of death due to an infectious disease worldwide. The high-level variability in tuberculosis disease responses in the human populace may be linked to immune processes related to inflammation. In many cases, inflammation appears to exasperate tuberculosis responses; however, some evidence suggests inflammatory processes improve control of M. tuberculosis infection. Here, we show an acute inflammatory stimulus in mice provides protection against M. tuberculosis for up to 6 months, suggesting acute inflammation can positively affect M. tuberculosis infection outcome.
Collapse
Affiliation(s)
- Tucker J. Piergallini
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Julia M. Scordo
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
- The Barshop Institute, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Paula A. Pino
- Population Health Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Larry S. Schlesinger
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jordi B. Torrelles
- Population Health Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Joanne Turner
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
27
|
Gregus AM, Levine IS, Eddinger KA, Yaksh TL, Buczynski MW. Sex differences in neuroimmune and glial mechanisms of pain. Pain 2021; 162:2186-2200. [PMID: 34256379 PMCID: PMC8277970 DOI: 10.1097/j.pain.0000000000002215] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
ABSTRACT Pain is the primary motivation for seeking medical care. Although pain may subside as inflammation resolves or an injury heals, it is increasingly evident that persistency of the pain state can occur with significant regularity. Chronic pain requires aggressive management to minimize its physiological consequences and diminish its impact on quality of life. Although opioids commonly are prescribed for intractable pain, concerns regarding reduced efficacy, as well as risks of tolerance and dependence, misuse, diversion, and overdose mortality rates limit their utility. Advances in development of nonopioid interventions hinge on our appreciation of underlying mechanisms of pain hypersensitivity. For instance, the contributory role of immunity and the associated presence of autoimmune syndromes has become of particular interest. Males and females exhibit fundamental differences in innate and adaptive immune responses, some of which are present throughout life, whereas others manifest with reproductive maturation. In general, the incidence of chronic pain conditions, particularly those with likely autoimmune covariates, is significantly higher in women. Accordingly, evidence is now accruing in support of neuroimmune interactions driving sex differences in the development and maintenance of pain hypersensitivity and chronicity. This review highlights known sexual dimorphisms of neuroimmune signaling in pain states modeled in rodents, which may yield potential high-value sex-specific targets to inform future analgesic drug discovery efforts.
Collapse
Affiliation(s)
- Ann M. Gregus
- School of Neuroscience, Virginia Polytechnic and State University, 970 Washington Street SW, Blacksburg, VA 24061
| | - Ian S. Levine
- School of Neuroscience, Virginia Polytechnic and State University, 970 Washington Street SW, Blacksburg, VA 24061
| | - Kelly A. Eddinger
- Dept. of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA, USA 92093-0818
| | - Tony L. Yaksh
- Dept. of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA, USA 92093-0818
- Dept. of Pharmacology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA, USA 92093-0601
| | - Matthew W. Buczynski
- School of Neuroscience, Virginia Polytechnic and State University, 970 Washington Street SW, Blacksburg, VA 24061
| |
Collapse
|
28
|
Manouchehrian O, Ramos M, Bachiller S, Lundgaard I, Deierborg T. Acute systemic LPS-exposure impairs perivascular CSF distribution in mice. J Neuroinflammation 2021; 18:34. [PMID: 33514389 PMCID: PMC7844902 DOI: 10.1186/s12974-021-02082-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background The exchange of cerebrospinal (CSF) and interstitial fluid is believed to be vital for waste clearance in the brain. The sleep-dependent glymphatic system, which is comprised of perivascular flow of CSF and is largely dependent on arterial pulsatility and astrocytic aquaporin-4 (AQP4) expression, facilitates much of this brain clearance. During the last decade, several observations have indicated that impaired glymphatic function goes hand in hand with neurodegenerative diseases. Since pathologies of the brain carry inflammatory components, we wanted to know how acute inflammation, e.g., with lipopolysaccharide (LPS) injections, would affect the glymphatic system. In this study, we aim to measure the effect of LPS on perivascular CSF distribution as a measure of glymphatic function. Methods Three hours after injection of LPS (1 mg/kg i.p.), C57bl/6 mice were (1) imaged for two CSF tracers, injected into cisterna magna, (2) transcardially perfused with buffer, or (3) used for physiological readouts. Tracer flow was imaged using a low magnification microscope on fixed brains, as well as using vibratome-cut slices for measuring tracer penetration in the brain. Cytokines, glial, and BBB-permeability markers were measured with ELISAs, Western blots, and immunohistochemistry. Cerebral blood flow was approximated using laser Doppler flowmetry, respiration and heart rate with a surgical monitor, and AQP4-polarization was quantified using confocal microscopy of immunolabeled brain sections. Results LPS-injections significantly lowered perivascular CSF tracer flow and penetration into the parenchyma. No differences in AQP4 polarization, cytokines, astroglial and BBB markers, cerebral blood flow, or respiration were detected in LPS-injected mice, although LPS did elevate cortical Iba1+ area and heart rate. Conclusions This study reports another physiological response after acute exposure to the bacterial endotoxin LPS, namely the statistically significant decrease in perivascular distribution of CSF. These observations may benefit our understanding of the role of systemic inflammation in brain clearance.
Collapse
Affiliation(s)
- Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.
| | - Marta Ramos
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, SE-223 62, Lund, Sweden
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, SE-223 62, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| |
Collapse
|
29
|
Oxidative and/or Inflammatory Thrust Induced by Silver Nanoparticles in Rabbits: Effect of Vitamin E or NSAID Administration on Semen Parameters. Mediators Inflamm 2020; 2020:6664062. [PMID: 33424436 PMCID: PMC7781726 DOI: 10.1155/2020/6664062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 11/18/2022] Open
Abstract
The aim of this research was to evaluate the inflammatory and/or oxidative damage related to silver nanoparticles (AgNPs), which are responsible for negative effects on sperm physiology and metabolism. Thirty New Zealand White rabbit bucks were divided into 5 experimental groups (6 animals/group): Control, treated with 0.9% NaCl; AgNP, treated with a 5 mM AgNP solution; LPS, treated with 50 g/kg b.w. E. coli LPS; AgNPs + NSAID, treated with an anti-inflammatory drug at 0.2 mg/kg b.w. and 5 mM AgNPs; and AgNPs + Vit E, treated with 0.18 mg/kg b.w. vitamin E and 5 mM AgNPs. Sperm quality and oxidative and inflammatory status were assessed at different times (0-60 days). Two statistical models were built: the first evaluated the effects of AgNPs and LPS (vs. Control), whereas the second evaluated the protective effect of an NSAID and vitamin E against AgNP-induced damage. Three principal component analyses were performed: sperm traits (motility, volume), oxidative status (antioxidants, oxidative metabolites, and redox reactions), and cytokines (TNF-α, IL-8, and IL-6). A negative effect on reproductive traits resulted after NP administration. In particular, an inflammatory/oxidative response took place in the reproductive tract during the first 2-3 wks of AgNP administration (cytokine and oxidative metabolite generation); the inflammatory/oxidative thrust impaired the status of rabbit tissues (seminal plasma, sperm, and blood), inducing a response (increased antioxidant enzymes and redox reactions) at 4-7 wks; oxidative stress, if not totally counteracted, likely induced toxicity in the late phases of AgNP administration (8-9 wks). In conclusion, exposure to silver nanoparticles produced a similar but more persistent effect than that of LPS on rabbit reproductive tissues: AgNP administration triggered a proinflammatory response linked to oxidative thrust, worsening many sperm parameters. However, only anti-inflammatory treatment counteracted the negative effects of AgNPs, whereas vitamin E seemed to act as an adjuvant, attenuating the oxidative cascade.
Collapse
|
30
|
Tsuji Y, Kaji K, Kitade M, Kaya D, Kitagawa K, Ozutsumi T, Fujinaga Y, Takaya H, Kawaratani H, Namisaki T, Moriya K, Akahane T, Yoshiji H. Bile Acid Sequestrant, Sevelamer Ameliorates Hepatic Fibrosis with Reduced Overload of Endogenous Lipopolysaccharide in Experimental Nonalcoholic Steatohepatitis. Microorganisms 2020; 8:925. [PMID: 32575352 PMCID: PMC7357162 DOI: 10.3390/microorganisms8060925] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/11/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the use of various pharmacotherapeutic strategies, fibrosis due to nonalcoholic steatohepatitis (NASH) remains an unsatisfied clinical issue. We investigated the effect of sevelamer, a hydrophilic bile acid sequestrant, on hepatic fibrosis in a murine NASH model. Male C57BL/6J mice were fed a choline-deficient, L-amino acid-defined, high-fat (CDHF) diet for 12 weeks with or without orally administered sevelamer hydrochloride (2% per diet weight). Histological and biochemical analyses revealed that sevelamer prevented hepatic steatosis, macrophage infiltration, and pericellular fibrosis in CDHF-fed mice. Sevelamer reduced the portal levels of total bile acid and inhibited both hepatic and intestinal farnesoid X receptor activation. Gut microbiome analysis demonstrated that sevelamer improved a lower α-diversity and prevented decreases in Lactobacillaceae and Clostridiaceae as well as increases in Desulfovibrionaceae and Enterobacteriaceae in the CDHF-fed mice. Additionally, sevelamer bound to lipopolysaccharide (LPS) in the intestinal lumen and promoted its fecal excretion. Consequently, the sevelamer treatment restored the tight intestinal junction proteins and reduced the portal LPS levels, leading to the suppression of hepatic toll-like receptor 4 signaling pathway. Furthermore, sevelamer inhibited the LPS-mediated induction of fibrogenic activity in human hepatic stellate cells in vitro. Collectively, sevelamer inhibited the development of murine steatohepatitis by reducing hepatic LPS overload.
Collapse
Affiliation(s)
| | - Kosuke Kaji
- Department of Gastroenterology, Nara Medical University, Kashihara, Nara 634-8521, Japan; (Y.T.); (M.K.); (D.K.); (K.K.); (T.O.); (Y.F.); (H.T.); (H.K.); (T.N.); (K.M.); (T.A.); (H.Y.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Piirsalu M, Taalberg E, Lilleväli K, Tian L, Zilmer M, Vasar E. Treatment With Lipopolysaccharide Induces Distinct Changes in Metabolite Profile and Body Weight in 129Sv and Bl6 Mouse Strains. Front Pharmacol 2020; 11:371. [PMID: 32292347 PMCID: PMC7118216 DOI: 10.3389/fphar.2020.00371] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/11/2020] [Indexed: 12/19/2022] Open
Abstract
Mouse strains differ significantly in their behaviors and responses to pathogenic and pharmacological agents. This study seeks to characterize behavioral and metabolomic profiles of two widely used mouse lines, 129S6/SvEvTac (129Sv) and C57BL/6NTac (Bl6), to acute administration of lipopolysaccharide (LPS). LPS caused a significant suppression of locomotor activity and a decline in body weight (BW) in both strains within 24 h. However, the BW loss was more pronounced in Bl6 than in 129Sv. Comparison of strains revealed clear differences between their metabolomic profiles. According to the general linear model analysis (GLM), the 1.5 h LPS challenge in Bl6 caused a decrease of propionylcarnitine (C3), glucogenic amino acids, and acetylornithine (Ac-Orn), whereas the response of 129Sv included decreased concentrations of short-chain acylcarnitines (SCACs), citrulline, and elevation of glycerophospholipid (PCaa C42:0) and sphingolipid [SM(OH)C16:1]. 24 h after LPS administration, robust alterations in lipid profile were observed in both strains. LPS treatment caused elevation of sphingolipids, phosphatidylcholine diacyls (PCaa) as well as a decrease in lysophosphatidylcholines (LysoPC). However, the number of elevated PCaa and sphingolipids was considerably higher in 129Sv. In addition to lipids, 24 h LPS challenge in Bl6 mice induced increased levels of kynurenine (KYN), putrescine and decreased levels of citrulline, hexoses, Ac-Orn, and PC acyl-alkyl (PCae 38:2) as well as severe BW loss. In contrast, the 24 h LPS challenge in 129Sv mice induced increased levels of KYN, long-chain acylcarnitines (LCACs) and decreased levels of citrulline as well as moderate BW loss. Altogether, our study revealed both similarities and differences in response to LPS in Bl6 and 129Sv strains. For major differences, Bl6 mice showed stronger reduction of BW 24 h after LPS treatment, accompanied by significantly reduced levels of hexoses, the ratio between LysoPC16:1/LysoPC16:0, and elevated levels of neuroprotective putrescine. In 129Sv mice, the BW loss was milder, accompanied by increased levels of hydroxylated LCACs, probably reflecting shifts in oxidative metabolism of fatty acids. One may suggest that LPS caused stronger hypometabolic state in the Bl6 mice than in the 129Sv strain. Altogether, this study confirms that Bl6 and 129Sv mice display vastly distinct adaptation capacities independent from the nature of stressful challenge.
Collapse
Affiliation(s)
- Maria Piirsalu
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Egon Taalberg
- Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia.,Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Li Tian
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Mihkel Zilmer
- Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia.,Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
32
|
Lee JS, Jeon YJ, Park SY, Son CG. An Adrenalectomy Mouse Model Reflecting Clinical Features for Chronic Fatigue Syndrome. Biomolecules 2020; 10:71. [PMID: 31906307 PMCID: PMC7023174 DOI: 10.3390/biom10010071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 12/24/2019] [Accepted: 12/29/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic fatigue syndrome (CFS) is one of the most intractable diseases and is characterized by severe central fatigue that impairs even daily activity. To date, the pathophysiological mechanisms are uncertain and no therapies exist. Therefore, a proper animal model reflecting the clinical features of CFS is urgently required. We compared two CFS animal models most commonly used, by injection with lipopolysaccharide (LPS from Escherichia coli O111:B4) or polyinosinic: polycytidylic acid (poly I:C), along with bilateral adrenalectomy (ADX) as another possible model. Both LPS- and poly I:C-injected mice dominantly showed depressive behaviors, while ADX led to fatigue-like performances with high pain sensitivity. In brain tissues, LPS injection notably activated microglia and the 5-hydroxytryptamine (HT)1A receptor in the prefrontal cortex and hippocampus. Poly I:C-injection also remarkably activated the 5-HT transporter and 5-HT1A receptor with a reduction in serotonin levels in the brain. ADX particularly activated astrocytes and transforming growth factor beta (TGF-β) 1 in all brain regions. Our results revealed that LPS and poly I:C animal models approximate depressive disorder more closely than CFS. We suggest that ADX is a possible method for establishing a mouse model of CFS reflecting clinical features, especially in neuroendocrine system.
Collapse
Affiliation(s)
- Jin-Seok Lee
- Institute of Traditional Medicine and Bioscience, Dunsan Hospital of Daejeon University, Daejeon 34323, Korea; (J.-S.L.); (Y.-J.J.)
| | - Yoo-Jin Jeon
- Institute of Traditional Medicine and Bioscience, Dunsan Hospital of Daejeon University, Daejeon 34323, Korea; (J.-S.L.); (Y.-J.J.)
| | - Samuel-Young Park
- Korean Medical College of Daejeon University, 62, Daehak-ro, Dong-gu, Daejeon 34323, Korea;
| | - Chang-Gue Son
- Institute of Traditional Medicine and Bioscience, Dunsan Hospital of Daejeon University, Daejeon 34323, Korea; (J.-S.L.); (Y.-J.J.)
| |
Collapse
|
33
|
Gamma Visual Stimulation Induces a Neuroimmune Signaling Profile Distinct from Acute Neuroinflammation. J Neurosci 2019; 40:1211-1225. [PMID: 31871276 PMCID: PMC7002142 DOI: 10.1523/jneurosci.1511-19.2019] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022] Open
Abstract
Many neurodegenerative and neurological diseases are rooted in dysfunction of the neuroimmune system; therefore, manipulating this system has strong therapeutic potential. Prior work has shown that exposing mice to flickering lights at 40 Hz drives gamma frequency (∼40 Hz) neural activity and recruits microglia, the primary immune cells of the brain, revealing a novel method to manipulate the neuroimmune system. Many neurodegenerative and neurological diseases are rooted in dysfunction of the neuroimmune system; therefore, manipulating this system has strong therapeutic potential. Prior work has shown that exposing mice to flickering lights at 40 Hz drives gamma frequency (∼40 Hz) neural activity and recruits microglia, the primary immune cells of the brain, revealing a novel method to manipulate the neuroimmune system. However, the biochemical signaling mechanisms between 40 Hz neural activity and immune recruitment remain unknown. Here, we exposed wild-type male mice to 5–60 min of 40 Hz or control flicker and assessed cytokine and phosphoprotein networks known to play a role in immune function. We found that 40 Hz flicker leads to increases in the expression of cytokines which promote microglial phagocytic states, such as IL-6 and IL-4, and increased expression of microglial chemokines, such as macrophage-colony-stimulating factor and monokine induced by interferon-γ. Interestingly, cytokine effects differed as a function of stimulation frequency, revealing a range of neuroimmune effects of stimulation. To identify possible mechanisms underlying cytokine expression, we quantified the effect of the flicker on intracellular signaling pathways known to regulate cytokine levels. We found that a 40 Hz flicker upregulates phospho-signaling within the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. While cytokine expression increased after 1 h of 40 Hz flicker stimulation, protein phosphorylation in the NF-κB pathway was upregulated within minutes. Importantly, the cytokine expression profile induced by 40 Hz flicker was different from cytokine changes in response to acute neuroinflammation induced by lipopolysaccharides. These results are the first, to our knowledge, to show how visual stimulation rapidly induces critical neuroimmune signaling in healthy animals. SIGNIFICANCE STATEMENT Prior work has shown that exposing mice to lights flickering at 40 Hz induces neural spiking activity at 40 Hz (within the gamma frequency) and recruits microglia, the primary immune cells of the brain. However, the immediate effect of 40 Hz flicker on neuroimmune biochemical signaling was unknown. We found that 40 Hz flicker leads to significant increases in the expression of cytokines, key immune signals known to recruit microglia. Furthermore, we found that 40 Hz flicker rapidly changes the phosphorylation of proteins in the NF-κB and MAPK pathways, both known to regulate cytokine expression. Our findings are the first to delineate a specific rapid immune signaling response following 40 Hz visual stimulation, highlighting both the unique nature and therapeutic potential of this treatment.
Collapse
|
34
|
Kenig A, Ilan Y. A Personalized Signature and Chronotherapy-Based Platform for Improving the Efficacy of Sepsis Treatment. Front Physiol 2019; 10:1542. [PMID: 31920730 PMCID: PMC6930923 DOI: 10.3389/fphys.2019.01542] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/05/2019] [Indexed: 12/27/2022] Open
Abstract
Sepsis remains a major therapeutic challenge and is associated with a high rate of morbidity and mortality. It is a dynamic condition in which multiple parameters change over time, rendering it difficult to overcome the various injurious responses, which worsen the prognosis in these patients. The prognosis of sepsis is associated with a disbalance of compensatory responses to infectious triggers, part of which can be deleterious. Marked inter- and intra-patient variability characterizes the mechanisms that underlie sepsis progression and determine the response to therapy. In this paper, we review some of the data on the use of chronopharmacological approaches for the treatment of patients with sepsis and discuss the role of the autonomic nervous system in the mechanisms associated with immune response and chronotherapy in these patients. We describe the implementation of an individualized platform that is based on the personalized autonomic nervous system, immune, and chronobiology-derived parameters for generating a patient-tailored therapeutic regimen. The notion of overcoming the deleterious compensatory response in a highly dynamic system in sepsis is presented to ensure an improved response to current therapies.
Collapse
Affiliation(s)
- Ariel Kenig
- Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yaron Ilan
- Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
35
|
Le Page LM, Guglielmetti C, Najac CF, Tiret B, Chaumeil MM. Hyperpolarized 13 C magnetic resonance spectroscopy detects toxin-induced neuroinflammation in mice. NMR IN BIOMEDICINE 2019; 32:e4164. [PMID: 31437326 PMCID: PMC6817388 DOI: 10.1002/nbm.4164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/27/2019] [Accepted: 07/15/2019] [Indexed: 05/04/2023]
Abstract
Lipopolysaccharide (LPS) is a commonly used agent for induction of neuroinflammation in preclinical studies. Upon injection, LPS causes activation of microglia and astrocytes, whose metabolism alters to favor glycolysis. Assessing in vivo neuroinflammation and its modulation following therapy remains challenging, and new noninvasive methods allowing for longitudinal monitoring would be highly valuable. Hyperpolarized (HP) 13 C magnetic resonance spectroscopy (MRS) is a promising technique for assessing in vivo metabolism. In addition to applications in oncology, the most commonly used probe of [1-13 C] pyruvate has shown potential in assessing neuroinflammation-linked metabolism in mouse models of multiple sclerosis and traumatic brain injury. Here, we aimed to investigate LPS-induced neuroinflammatory changes using HP [1-13 C] pyruvate and HP 13 C urea. 2D chemical shift imaging following simultaneous intravenous injection of HP [1-13 C] pyruvate and HP 13 C urea was performed at baseline (day 0) and at days 3 and 7 post-intracranial injection of LPS (n = 6) or saline (n = 5). Immunofluorescence (IF) analyses were performed for Iba1 (resting and activated microglia/macrophages), GFAP (resting and reactive astrocytes) and CD68 (activated microglia/macrophages). A significant increase in HP [1-13 C] lactate production was observed at days 3 and 7 following injection, in the injected (ipsilateral) side of the LPS-treated mouse brain, but not in either the contralateral side or saline-injected animals. HP 13 C lactate/pyruvate ratio, without and with normalization to urea, was also significantly increased in the ipsilateral LPS-injected brain at 7 days compared with baseline. IF analyses showed a significant increase in CD68 and GFAP staining at 3 days, followed by increased numbers of Iba1 and GFAP positive cells at 7 days post-LPS injection. In conclusion, we can detect LPS-induced changes in the mouse brain using HP 13 C MRS, in alignment with increased numbers of microglia/macrophages and astrocytes. This study demonstrates that HP 13 C spectroscopy has substantial potential for providing noninvasive information on neuroinflammation.
Collapse
Affiliation(s)
- Lydia M Le Page
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Caroline Guglielmetti
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Chloé F Najac
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Brice Tiret
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Myriam M Chaumeil
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| |
Collapse
|
36
|
Giménez-Llort L, Ramírez-Boix P, de la Fuente M. Mortality of septic old and adult male mice correlates with individual differences in premorbid behavioral phenotype and acute-phase sickness behavior. Exp Gerontol 2019; 127:110717. [PMID: 31479727 DOI: 10.1016/j.exger.2019.110717] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/08/2019] [Accepted: 08/30/2019] [Indexed: 11/18/2022]
Abstract
Individual differences in premorbid behaviors and in those exhibited in the course of an infection disease may be useful to explain the individual susceptibility to infections, the underlying neuroimmunological mechanisms and be helpful to design patient oriented treatments with better prediction of pharmacological reactivity/outcome. Age (old) and gender (male) are also considered vulnerability factors. In the present study, the motor, emotional, anxious-like and social phenotypes of adult (6-month-old) and old (18-month-old) male C57BL/6 × 129Sv mice were determined using both a transversal and longitudinal designs prior to the analysis of LPS (150 mg/kg, i.p.)-induced sickness behavior and mortality. The results show: i) Individual premorbid behavioral phenotype had short- and long-term predictive value of hours of survival; ii) Persistence of behavioral traits from adulthood to old age and predictive value on hours of survival; iii) First signs of sickness behavior were also predicting mortality, mostly in old animals; iv) LPS-sickness behavior was the same at both ages but adult animals were able to show attempts of motor recovery; v) The mortality rate over 96 h was 100% in both ages, but old animals showed shorter survival times. In summary, these results confirm the relevance of age/aging but also individual behavioral differences in the premorbid phenotype and the morbidity response to the LPS-induced-sepsis that correlate with the individual's mortality. Thus, this work supports the translational scenarios to study personalized evaluation of risks factors and psycho-neuro-immunological mechanisms relevant for better interventions and prognosis in the critically ill young but specially aged patient population.
Collapse
Affiliation(s)
- L Giménez-Llort
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neuroscience, School of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| | - P Ramírez-Boix
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neuroscience, School of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - M de la Fuente
- Department of Animal Physiology II, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
37
|
Sex-Related Differences in Immune Response and Symptomatic Manifestations to Infection with Leishmania Species. J Immunol Res 2019; 2019:4103819. [PMID: 30756088 PMCID: PMC6348913 DOI: 10.1155/2019/4103819] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/22/2018] [Accepted: 12/11/2018] [Indexed: 12/19/2022] Open
Abstract
Worldwide, an estimated 12 million people are infected with Leishmania spp. and an additional 350 million are at risk of infection. Leishmania are intracellular parasites that cause disease by suppressing macrophage microbicidal responses. Infection can remain asymptomatic or lead to a spectrum of diseases including cutaneous, mucocutaneous, and visceral leishmaniasis. Ultimately, the combination of both pathogen and host factors determines the outcome of infection. Leishmaniasis, as well as numerous other infectious diseases, exhibits sex-related differences that cannot be explained solely in terms of environmental exposure or healthcare access. Furthermore, transcriptomic evidence is revealing that biological sex is a variable impacting physiology, immune response, drug metabolism, and consequently, the progression of disease. Herein, we review the distribution, morbidity, and mortality among male and female leishmaniasis patients. Additionally, we discuss experimental findings and new avenues of research concerning sex-specific responses in cutaneous and visceral leishmaniasis. The limitations of current therapies and the emergence of drug-resistant parasites underscore the need for new treatments that could harness the host immune response. As such, understanding the mechanisms driving the differential immune response and disease outcome of males versus females is a necessary step in the development of safer and more effective treatments against leishmaniasis.
Collapse
|
38
|
Dando SJ, Kazanis R, Chinnery HR, McMenamin PG. Regional and functional heterogeneity of antigen presenting cells in the mouse brain and meninges. Glia 2018; 67:935-949. [DOI: 10.1002/glia.23581] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 10/11/2018] [Accepted: 11/27/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Samantha J. Dando
- Department of Anatomy and Developmental BiologyMonash Biomedicine Discovery Institute, Monash University Clayton Victoria Australia
| | - Renee Kazanis
- Department of Anatomy and Developmental BiologyMonash Biomedicine Discovery Institute, Monash University Clayton Victoria Australia
| | - Holly R. Chinnery
- Department of Optometry and Vision SciencesThe University of Melbourne Parkville Victoria Australia
| | - Paul G. McMenamin
- Department of Anatomy and Developmental BiologyMonash Biomedicine Discovery Institute, Monash University Clayton Victoria Australia
| |
Collapse
|
39
|
Meneses G, Cárdenas G, Espinosa A, Rassy D, Pérez-Osorio IN, Bárcena B, Fleury A, Besedovsky H, Fragoso G, Sciutto E. Sepsis: developing new alternatives to reduce neuroinflammation and attenuate brain injury. Ann N Y Acad Sci 2018; 1437:43-56. [DOI: 10.1111/nyas.13985] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Gabriela Meneses
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Graciela Cárdenas
- Instituto Nacional de Neurología y Neurocirugía; SSA; Mexico City Mexico
| | - Alejandro Espinosa
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Dunia Rassy
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Ivan Nicolás Pérez-Osorio
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Brandon Bárcena
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Agnes Fleury
- Instituto Nacional de Neurología y Neurocirugía; SSA; Mexico City Mexico
| | - Hugo Besedovsky
- The Institute of Physiology and Pathophysiology, Medical Faculty; Philipps University; Marburg Germany
| | - Gladis Fragoso
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Edda Sciutto
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| |
Collapse
|