1
|
Kodali M, Madhu LN, Somayaji Y, Attaluri S, Huard C, Panda PK, Shankar G, Rao S, Shuai B, Gonzalez JJ, Oake C, Hering C, Babu RS, Kotian S, Shetty AK. Residual microglia following short-term PLX5622 treatment in 5xFAD mice exhibit diminished NLRP3 inflammasome and mTOR signaling, and enhanced autophagy. Aging Cell 2025; 24:e14398. [PMID: 39571180 PMCID: PMC11822669 DOI: 10.1111/acel.14398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 11/27/2024] Open
Abstract
While moderately activated microglia in Alzheimer's disease (AD) are pivotal in clearing amyloid beta (Aβ), hyperactivated microglia perpetuate neuroinflammation. Prior investigations reported that the elimination of ~80% of microglia through inhibition of the colony-stimulating factor 1 receptor (CSF1R) during the advanced stage of neuroinflammation in 5xFamilial AD (5xFAD) mice mitigates synapse loss and neurodegeneration. Furthermore, prolonged CSF1R inhibition diminished the development of parenchymal plaques. Nonetheless, the effects of short-term CSF1R inhibition during the early stages of neuroinflammation on residual microglia are unknown. Therefore, we investigated the effects of 10-day CSF1R inhibition using PLX5622 in three-month-old female 5xFAD mice, a stage characterized by the onset of neuroinflammation and minimal Aβ plaques. We observed ~65% microglia depletion in the hippocampus and cerebral cortex. The leftover microglia displayed a noninflammatory phenotype with reduced NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complexes. Moreover, plaque-associated microglia were reduced with diminished Clec7a expression. Additionally, phosphorylated S6 ribosomal protein and the protein sequestosome 1 analysis suggested reduced mechanistic targets of rapamycin (mTOR) signaling and autophagy in microglia and neurons within the hippocampus and cerebral cortex. Biochemical assays validated the inhibition of NLRP3 inflammasome activation, decreased mTOR signaling in the hippocampus and cerebral cortex, and enhanced autophagy in the hippocampus. However, short-term CSF1R inhibition did not influence Aβ plaques, soluble Aβ-42 levels, astrocyte hypertrophy, or hippocampal neurogenesis. Thus, short-term CSF1R inhibition during the early stages of neuroinflammation in 5xFAD mice promotes the retention of homeostatic microglia with diminished inflammasome activation and mTOR signaling, alongside increased autophagy.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Charles Huard
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Prashanta Kumar Panda
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Goutham Shankar
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Jenny J. Gonzalez
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Chris Oake
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Catherine Hering
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Roshni Sara Babu
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Sanya Kotian
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University Health Science Center School of MedicineCollege StationTexasUSA
| |
Collapse
|
2
|
Kodali M, Madhu LN, Kolla VSV, Attaluri S, Huard C, Somayaji Y, Shuai B, Jordan C, Rao X, Shetty S, Shetty AK. FDA-approved cannabidiol [Epidiolex ®] alleviates Gulf War Illness-linked cognitive and mood dysfunction, hyperalgesia, neuroinflammatory signaling, and declined neurogenesis. Mil Med Res 2024; 11:61. [PMID: 39169440 PMCID: PMC11340098 DOI: 10.1186/s40779-024-00563-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Chronic Gulf War Illness (GWI) is characterized by cognitive and mood impairments, as well as persistent neuroinflammation and oxidative stress. This study aimed to investigate the efficacy of Epidiolex®, a Food and Drug Administration (FDA)-approved cannabidiol (CBD), in improving brain function in a rat model of chronic GWI. METHODS Six months after exposure to low doses of GWI-related chemicals [pyridostigmine bromide, N,N-diethyl-meta-toluamide (DEET), and permethrin (PER)] along with moderate stress, rats with chronic GWI were administered either vehicle (VEH) or CBD (20 mg/kg, oral) for 16 weeks. Neurobehavioral tests were conducted on 11 weeks after treatment initiation to evaluate the performance of rats in tasks related to associative recognition memory, object location memory, pattern separation, and sucrose preference. The effect of CBD on hyperalgesia was also examined. The brain tissues were processed for immunohistochemical and molecular studies following behavioral tests. RESULTS GWI rats treated with VEH exhibited impairments in all cognitive tasks and anhedonia, whereas CBD-treated GWI rats showed improvements in all cognitive tasks and no anhedonia. Additionally, CBD treatment alleviated hyperalgesia in GWI rats. Analysis of hippocampal tissues from VEH-treated rats revealed astrocyte hypertrophy and increased percentages of activated microglia presenting NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) complexes as well as elevated levels of proteins involved in NLRP3 inflammasome activation and Janus kinase/signal transducers and activators of the transcription (JAK/STAT) signaling. Furthermore, there were increased concentrations of proinflammatory and oxidative stress markers along with decreased neurogenesis. In contrast, the hippocampus from CBD-treated GWI rats displayed reduced levels of proteins mediating the activation of NLRP3 inflammasomes and JAK/STAT signaling, normalized concentrations of proinflammatory cytokines and oxidative stress markers, and improved neurogenesis. Notably, CBD treatment did not alter the concentration of endogenous cannabinoid anandamide in the hippocampus. CONCLUSIONS The use of an FDA-approved CBD (Epidiolex®) has been shown to effectively alleviate cognitive and mood impairments as well as hyperalgesia associated with chronic GWI. Importantly, the improvements observed in rats with chronic GWI in this study were attributed to the ability of CBD to significantly suppress signaling pathways that perpetuate chronic neuroinflammation.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Venkata Sai Vashishta Kolla
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Charles Huard
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Chase Jordan
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Sanath Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA.
| |
Collapse
|
3
|
Kodali M, Madhu LN, Somayaji Y, Attaluri S, Huard C, Panda PK, Shankar G, Rao S, Shuai B, Gonzalez JJ, Oake C, Hering C, Babu RS, Kotian S, Shetty AK. Residual Microglia Following Short-term PLX5622 Treatment in 5xFAD Mice Exhibit Diminished NLRP3 Inflammasome and mTOR Signaling, and Enhanced Autophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603157. [PMID: 39071343 PMCID: PMC11275929 DOI: 10.1101/2024.07.11.603157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Chronic neuroinflammation represents a prominent hallmark of Alzheimer's disease (AD). While moderately activated microglia are pivotal in clearing amyloid beta (Aβ), hyperactivated microglia perpetuate neuroinflammation. Prior investigations have indicated that the elimination of ∼80% of microglia through a month-long inhibition of the colony-stimulating factor 1 receptor (CSF1R) during the advanced stage of neuroinflammation in 5xFamilial AD (5xFAD) mice mitigates synapse loss and neurodegeneration without impacting Aβ levels. Furthermore, prolonged CSF1R inhibition diminished the development of parenchymal plaques. Nonetheless, the immediate effects of short-term CSF1R inhibition during the early stages of neuroinflammation on residual microglial phenotype or metabolic fitness are unknown. Therefore, we investigated the effects of 10-day CSF1R inhibition in three-month-old female 5xFAD mice, a stage characterized by the onset of neuroinflammation and minimal Aβ plaques. We observed ∼65% microglia depletion in the hippocampus and cerebral cortex. The leftover microglia demonstrated a noninflammatory phenotype, with highly branched and ramified processes and reduced NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complexes. Moreover, plaque-associated microglia were reduced in number with diminished Clec7a (dectin-1) expression. Additionally, both microglia and neurons displayed reduced mechanistic target of rapamycin (mTOR) signaling and autophagy. Biochemical assays validated the inhibition of NLRP3 inflammasome activation, decreased mTOR signaling, and enhanced autophagy. However, short-term CSF1R inhibition did not influence Aβ plaques, soluble Aβ-42 levels, or hippocampal neurogenesis. Thus, short-term CSF1R inhibition during the early stages of neuroinflammation in 5xFAD mice promotes the retention of homeostatic microglia with diminished inflammasome activation and mTOR signaling, alongside increased autophagy.
Collapse
|
4
|
Strohm AO, Majewska AK. Physical exercise regulates microglia in health and disease. Front Neurosci 2024; 18:1420322. [PMID: 38911597 PMCID: PMC11192042 DOI: 10.3389/fnins.2024.1420322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
There is a well-established link between physical activity and brain health. As such, the effectiveness of physical exercise as a therapeutic strategy has been explored in a variety of neurological contexts. To determine the extent to which physical exercise could be most beneficial under different circumstances, studies are needed to uncover the underlying mechanisms behind the benefits of physical activity. Interest has grown in understanding how physical activity can regulate microglia, the resident immune cells of the central nervous system. Microglia are key mediators of neuroinflammatory processes and play a role in maintaining brain homeostasis in healthy and pathological settings. Here, we explore the evidence suggesting that physical activity has the potential to regulate microglia activity in various animal models. We emphasize key areas where future research could contribute to uncovering the therapeutic benefits of engaging in physical exercise.
Collapse
Affiliation(s)
- Alexandra O. Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Ania K. Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
5
|
Li M, Xu J, Li L, Zhang L, Zuo Z, Feng Y, He X, Hu X. Voluntary wheel exercise improves glymphatic clearance and ameliorates colitis-associated cognitive impairment in aged mice by inhibiting TRPV4-induced astrocytic calcium activity. Exp Neurol 2024; 376:114770. [PMID: 38580155 DOI: 10.1016/j.expneurol.2024.114770] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/08/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND AND OBJECTIVES Chronic colitis exacerbates neuroinflammation, contributing to cognitive impairment during aging, but the mechanism remains unclear. The polarity distribution of astrocytic aquaporin 4 (AQP4) is crucial for the glymphatic system, which is responsible for metabolite clearance in the brain. Physical exercise (PE) improves cognition in the aged. This study aims to investigate the protective mechanism of exercise in colitis-associated cognitive impairment. METHODS To establish a chronic colitis model, 18-month-old C57BL/6 J female mice received periodic oral administration of 1% wt/vol dextran sodium sulfate (DSS) in drinking water. The mice in the exercise group received four weeks of voluntary wheel exercise. High-throughput sequencing was conducted to screen for differentially expressed genes. Two-photon imaging was performed to investigate the function of the astrocytic calcium activity and in vivo intervention with TRPV4 inhibitor HC-067047. Further, GSK1016790A (GSK1), a TRPV4 agonist, was daily intraperitoneally injected during the exercise period to study the involvement of TRPV4 in PE protection. Colitis pathology was confirmed by histopathology. The novel object recognition (NOR) test, Morris water maze test (MWM), and open field test were performed to measure colitis-induced cognition and anxiety-like behavior. In vivo two-photon imaging and ex vivo imaging of fluorescent CSF tracers to evaluate the function of the glymphatic system. Immunofluorescence staining was used to detect the Aβ deposition, polarity distribution of astrocytic AQP4, and astrocytic phenotype. Serum and brain levels of the inflammatory cytokines were tested by Enzyme-linked immunosorbent assay (ELISA). The brain TUNEL assay was used to assess DNA damage. Expression of critical molecules was detected using Western blotting. RESULTS Voluntary exercise alleviates cognitive impairment and anxiety-like behavior in aged mice with chronic colitis, providing neuroprotection against neuronal damage and apoptosis. Additionally, voluntary exercise promotes the brain clearance of Aβ via increased glymphatic clearance. Mechanistically, exercise-induced beneficial effects may be attributed, in part, to the inhibition of TRPV4 expression and TRPV4-related calcium hyperactivity, subsequent promotion of AQP4 polarization, and modulation of astrocyte phenotype. CONCLUSION The present study reveals a novel role of voluntary exercise in alleviating colitis-related cognitive impairment and anxiety disorder, which is mediated by the promotion of AQP4 polarization and glymphatic clearance of Aβ via inhibition of TRPV4-induced astrocytic calcium hyperactivity.
Collapse
Affiliation(s)
- Mingyue Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinghui Xu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lili Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zejie Zuo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yifeng Feng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaofei He
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
6
|
Wei ZYD, Liang K, Shetty AK. Role of Microglia, Decreased Neurogenesis and Oligodendrocyte Depletion in Long COVID-Mediated Brain Impairments. Aging Dis 2023; 14:1958-1966. [PMID: 37815903 PMCID: PMC10676788 DOI: 10.14336/ad.2023.10918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 10/12/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of a recent worldwide coronavirus disease-2019 (COVID-19) pandemic. SARS-CoV-2 primarily causes an acute respiratory infection but can progress into significant neurological complications in some. Moreover, patients with severe acute COVID-19 could develop debilitating long-term sequela. Long-COVID is characterized by chronic symptoms that persist months after the initial infection. Common complaints are fatigue, myalgias, depression, anxiety, and "brain fog," or cognitive and memory impairments. A recent study demonstrated that a mild COVID-19 respiratory infection could generate elevated proinflammatory cytokines and chemokines in the cerebral spinal fluid. This commentary discusses findings from this study, demonstrating that even a mild respiratory SARS-CoV-2 infection can cause considerable neuroinflammation with microglial and macrophage reactivity. Such changes could also be gleaned by measuring chemokines and cytokines in the circulating blood. Moreover, neuroinflammation caused by mild SARS-CoV-2 infection can also impair hippocampal neurogenesis, deplete oligodendrocytes, and decrease myelinated axons. All these changes likely contribute to cognitive deficits in long-COVID syndrome. Therefore, strategies capable of restraining neuroinflammation, maintaining better hippocampal neurogenesis, and preserving oligodendrocyte lineage differentiation and maturation may prevent or reduce the incidence of long-COVID after SARS-CoV-2 respiratory infection.
Collapse
Affiliation(s)
- Zhuang-Yao D. Wei
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, USA.
| | - Ketty Liang
- Sam Houston State University College of Osteopathic Medicine, Conroe, TX, USA.
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, USA.
| |
Collapse
|
7
|
Li D, Yang H, Lyu M, Zhou L, Zhang Y, Kang C, Wang J, Wang Y. Association between Behavioural Risks and Alzheimer's Disease: Elucidated with an Integrated Analysis of Gene Expression Patterns and Molecular Mechanisms. Neurosci Biobehav Rev 2023; 150:105207. [PMID: 37146892 DOI: 10.1016/j.neubiorev.2023.105207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/12/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023]
Abstract
Alzheimer's disease (AD) remains a global health challenge. Previous studies have reported linkages between AD and multiple behavioural risk exposures, however, the underlying biological mechanisms and crucial genes of gene expression patterns driven by behavioural risks on the onset or progression of AD remains ambiguous. In this study, we performed an integrated analysis on the influence of behavioural risks including smoking, excessive alcohol consumption, physical inactivity, and non-healthy dietary pattern on AD with a comprehensive strategy. Our results demonstrated that multiple behavioural risk exposures could independently or collectively influence diverse hierarchical levels of gene expression patterns through multiple biological mechanisms such as Wnt, mitogen-activated protein kinase (MAPK), AMP-activated protein kinase (AMPK), nuclear factor (NF)-κB, phosphatidylinositol 3-kinase (PI3K)-Akt, and insulin (INS) signalling pathways-mediated pathological processes, thereby prodromally or intermediately impacting AD. Our study provided insights into understanding the association of behavioural risk exposures with AD and informative support for further studies.
Collapse
Affiliation(s)
- Dun Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hongxi Yang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mingqian Lyu
- Department of Computer Science, RWTH Aachen University, Aachen, 52062, Germany
| | - Lihui Zhou
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Yuan Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chunsheng Kang
- epartment of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, 300070, China
| | - Yaogang Wang
- School of Integrative Medicine, Public Health Science and Engineering College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Public Health, Tianjin Medical University, Tianjin, 300070, China; National Institute of Health Data Science at Peking University, Peking University, Beijing, 100191, China.
| |
Collapse
|
8
|
Kodali M, Jankay T, Shetty AK, Reddy DS. Pathophysiological basis and promise of experimental therapies for Gulf War Illness, a chronic neuropsychiatric syndrome in veterans. Psychopharmacology (Berl) 2023; 240:673-697. [PMID: 36790443 DOI: 10.1007/s00213-023-06319-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/17/2023] [Indexed: 02/16/2023]
Abstract
This article describes the pathophysiology and potential treatments for Gulf War Illness (GWI), which is a chronic neuropsychiatric illness linked to a combination of chemical exposures experienced by service personnel during the first Gulf War in 1991. However, there is currently no effective treatment for veterans with GWI. The article focuses on the current status and efficacy of existing therapeutic interventions in preclinical models of GWI, as well as potential perspectives of promising therapies. GWI stems from changes in brain and peripheral systems in veterans, leading to neurocognitive deficits, as well as physiological and psychological effects resulting from multifaceted changes such as neuroinflammation, oxidative stress, and neuronal damage. Aging not only renders veterans more susceptible to GWI symptoms, but also attenuates their immune capabilities and response to therapies. A variety of experimental models are being used to investigate the pathophysiology and develop therapies that have the ability to alleviate devastating symptoms. Over two dozen therapeutic interventions targeting neuroinflammation, mitochondrial dysfunction, neuronal injury, and neurogenesis are being tested, including agents such as curcumin, curcumin nanoparticles, monosodium luminol, melatonin, resveratrol, fluoxetine, rolipram, oleoylethanolamide, ketamine, levetiracetam, nicotinamide riboside, minocycline, pyridazine derivatives, and neurosteroids. Preclinical outcomes show that some agents have promise, including curcumin, resveratrol, and ketamine, which are being tested in clinical trials in GWI veterans. Neuroprotectants and other compounds such as monosodium luminol, melatonin, levetiracetam, oleoylethanolamide, and nicotinamide riboside appear promising for future clinical trials. Neurosteroids have been shown to have neuroprotective and disease-modifying properties, which makes them a promising medicine for GWI. Therefore, accelerated clinical studies are urgently needed to evaluate and launch an effective therapy for veterans displaying GWI.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University School of Medicine, College Station, TX, USA
| | - Tanvi Jankay
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University School of Medicine, College Station, TX, USA.,Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA. .,Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
| |
Collapse
|
9
|
Hu X, Ou Y, Li J, Sun M, Ge Q, Pan Y, Cai Z, Tan R, Wang W, An J, Lu H. Voluntary Exercise to Reduce Anxiety Behaviour in Traumatic Brain Injury Shown to Alleviate Inflammatory Brain Response in Mice. Int J Mol Sci 2023; 24:ijms24076365. [PMID: 37047351 PMCID: PMC10093932 DOI: 10.3390/ijms24076365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
Traumatic brain injury is a leading cause of neuroinflammation and anxiety disorders in young adults. Immune-targeted therapies have garnered attention for the amelioration of TBI-induced anxiety. A previous study has indicated that voluntary exercise intervention following TBI could reduce neuroinflammation. It is essential to determine the effects of voluntary exercise after TBI on anxiety via inhibiting neuroinflammatory response. Mice were randomly divided into four groups (sham, TBI, sham + voluntary wheel running (VWR), and TBI + VWR). One-week VWR was carried out on the 2nd day after trauma. The neurofunction of TBI mice was assessed. Following VWR, anxiety behavior was evaluated, and neuroinflammatory responses in the perilesional cortex were investigated. Results showed that after one week of VWR, neurofunctional recovery was enhanced, while the anxiety behavior of TBI mice was significantly alleviated. The level of pro-inflammatory factors decreased, and the level of anti-inflammatory factors elevated. Activation of nucleotide oligomerization domain-like thermal receptor protein domain associated protein 3 (NLRP3) inflammasome was inhibited significantly. All these alterations were consistent with reduced microglial activation at the perilesional site and positively correlated with the amelioration of anxiety behavior. This suggested that timely rehabilitative exercise could be a useful therapeutic strategy for anxiety resulting from TBI by targeting neuroinflammation.
Collapse
Affiliation(s)
- Xiaoxuan Hu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Yuhang Ou
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Jiashuo Li
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Meiqi Sun
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Qian Ge
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Yongqi Pan
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Zhenlu Cai
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Ruolan Tan
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Wenyu Wang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Jing An
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Correspondence: (J.A.); (H.L.)
| | - Haixia Lu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Correspondence: (J.A.); (H.L.)
| |
Collapse
|
10
|
Li C, Hu J, Liu W, Ke C, Huang C, Bai Y, Pan B, Wang J, Wan C. Exercise Intervention Modulates Synaptic Plasticity by Inhibiting Excessive Microglial Activation via Exosomes. Front Cell Neurosci 2022; 16:953640. [PMID: 35928570 PMCID: PMC9345504 DOI: 10.3389/fncel.2022.953640] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background Exosomes can activate microglia to modulate neural activity and synaptic plasticity by phagocytosis of neural spines or synapses. Our previous research found that an early 4-week exercise intervention in middle cerebral artery occlusion (MCAO) rats can promote the release of exosomes and protect the brain. This study intended to further explore the intrinsic mechanism of neuroprotection by exosome release after exercise. Methods Rats were randomly divided into four groups: the sham operation (SHAM), middle cerebral artery occlusion (MCAO) with sedentary intervention (SED-MCAO), MCAO with exercise intervention (EX-MCAO), and MCAO with exercise intervention and exosome injection (EX-MCAO-EXO). Modified neurological severity score (mNSS), cerebral infarction volume ratio, microglial activation, dendritic complexity, and expression of synaptophysin (Syn) and postsynaptic density protein 95 (PSD-95) were detected after 28 days of intervention. Results (1) The exercise improved body weight and mNSS score, and the survival state of the rats after exosome infusion was better. (2) Compared with the SED-MCAO group, the EX-MCAO (P = 0.039) and EX-MCAO-EXO groups (P = 0.002) had significantly lower cerebral infarct volume ratios (P < 0.05), among which the EX-MCAO-EXO group had the lowest (P = 0.031). (3) Compared with the SED-MCAO group, the EX-MCAO and EX-MCAO-EXO groups had a significantly decreased number of microglia (P < 0.001) and significantly increased process length/cell (P < 0.01) and end point/cell (P < 0.01) values, with the EX-MCAO-EXO group having the lowest number of microglia (P = 0.036) and most significantly increased end point/cell value (P = 0.027). (4) Compared with the SED-MCAO group, the total number of intersections and branches of the apical and basal dendrites in the EX-MCAO and EX-MCAO-EXO groups was increased significantly (P < 0.05), and the increase was more significant in the EX-MCAO-EXO group (P < 0.05). (5) The expression levels of Syn and PSD-95 in the EX-MCAO (PSyn = 0.043, PPSD−95 = 0.047) and EX-MCAO-EXO groups were significantly higher than those in the SED-MCAO group (P < 0.05), and the expression levels in the EX-MCAO-EXO group were significantly higher than those in the EX-MCAO group (P < 0.05). Conclusion Early exercise intervention after stroke can inhibit the excessive activation of microglia and regulate synaptic plasticity by exosome release.
Collapse
Affiliation(s)
- Chen Li
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiayi Hu
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wenhong Liu
- Tianjin Rehabilitation Center, Tianjin, China
| | - Changkai Ke
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuan Huang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Yifan Bai
- Department of Rehabilitation Medicine, School of Medicine Technology, Tianjin Medical University, Tianjin, China
| | - Bingchen Pan
- Department of Rehabilitation Medicine, School of Medicine Technology, Tianjin Medical University, Tianjin, China
| | - Junyi Wang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Chunxiao Wan
| |
Collapse
|
11
|
Promise of irisin to attenuate cognitive dysfunction in aging and Alzheimer's disease. Ageing Res Rev 2022; 78:101637. [PMID: 35504553 PMCID: PMC9844023 DOI: 10.1016/j.arr.2022.101637] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/19/2023]
Abstract
Strategies proficient for relieving cognitive impairments in aging and Alzheimer's disease (AD) have an enormous impact. Regular physical exercise (PE) can prevent age-related dementia and slow down AD progression. However, such a lifestyle change is likely not achievable for individuals displaying age-related frailty. Hence, drugs or biologics that could simulate the benefits of PE have received much attention. Previous studies suggested that the fibronectin-domain III containing 5 (FNDC5) underlies the PE-mediated improved cognitive function. A recent study reports that PE-related cognitive benefits in aging and AD are mediated by irisin, the cleaved form of FNDC5 released into the blood after PE. Such a conclusion was apparent from the deletion of irisin through a global knockout of FNDC5, leading to the loss of PE-induced cognitive benefits or inducing memory impairments in adult or aged models. Furthermore, in AD models, peripherally administered irisin mimicked the cognitive benefits of PE by modulating neuroinflammation. This short review discusses the promise of irisin to simulate the cognitive benefits of PE in age- and AD-related dementia. In addition, critical issues such as how blood-borne irisin acts on neural cells, the role of the brain-derived neurotrophic factor in irisin-mediated cognitive benefits, and irisin's ability to inhibit neuroinflammatory cascades in aging and AD are discussed.
Collapse
|
12
|
Attaluri S, Upadhya R, Kodali M, Madhu LN, Upadhya D, Shuai B, Shetty AK. Brain-Specific Increase in Leukotriene Signaling Accompanies Chronic Neuroinflammation and Cognitive Impairment in a Model of Gulf War Illness. Front Immunol 2022; 13:853000. [PMID: 35572589 PMCID: PMC9099214 DOI: 10.3389/fimmu.2022.853000] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Persistent cognitive impairment is a primary central nervous system-related symptom in veterans afflicted with chronic Gulf War Illness (GWI). Previous studies in a rat model have revealed that cognitive dysfunction in chronic GWI is associated with neuroinflammation, typified by astrocyte hypertrophy, activated microglia, and enhanced proinflammatory cytokine levels. Studies in a mouse model of GWI have also shown upregulation of several phospholipids that serve as reservoirs of arachidonic acid, a precursor of leukotrienes (LTs). However, it is unknown whether altered LT signaling is a component of chronic neuroinflammatory conditions in GWI. Therefore, this study investigated changes in LT signaling in the brain of rats displaying significant cognitive impairments six months after exposure to GWI-related chemicals and moderate stress. The concentration of cysteinyl LTs (CysLTs), LTB4, and 5-Lipoxygenase (5-LOX), the synthesizing enzyme of LTs, were evaluated. CysLT and LTB4 concentrations were elevated in the hippocampus and the cerebral cortex, along with enhanced 5-LOX expression in neurons and microglia. Such changes were also associated with increased proinflammatory cytokine levels in the hippocampus and the cerebral cortex. Enhanced CysLT and LTB4 levels in the brain could also be gleaned from their concentrations in brain-derived extracellular vesicles in the circulating blood. The circulating blood in GWI rats displayed elevated proinflammatory cytokines with no alterations in CysLT and LTB4 concentrations. The results provide new evidence that a brain-specific increase in LT signaling is another adverse alteration that potentially contributes to the maintenance of chronic neuroinflammation in GWI. Therefore, drugs capable of modulating LT signaling may reduce neuroinflammation and improve cognitive function in GWI. Additional findings demonstrate that altered LT levels in the brain could be tracked efficiently by analyzing brain-derived EVs in the circulating blood.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX, United States
| |
Collapse
|
13
|
Iannucci J, Nizamutdinov D, Shapiro LA. Neurogenesis and chronic neurobehavioral outcomes are partially improved by vagus nerve stimulation in a mouse model of Gulf War Illness. Neurotoxicology 2022; 90:205-215. [DOI: 10.1016/j.neuro.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/22/2022]
|
14
|
Ebrahimnejad M, Azizi P, Alipour V, Zarrindast MR, Vaseghi S. Complicated Role of Exercise in Modulating Memory: A Discussion of the Mechanisms Involved. Neurochem Res 2022; 47:1477-1490. [PMID: 35195832 DOI: 10.1007/s11064-022-03552-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/15/2022]
Abstract
Evidence has shown the beneficial effects of exercise on learning and memory. However, many studies have reported controversial results, indicating that exercise can impair learning and memory. In this article, we aimed to review basic studies reporting inconsistent complicated effects of exercise on memory in rodents. Also, we discussed the mechanisms involved in the effects of exercise on memory processes. In addition, we tried to find scientific answers to justify the inconsistent results. In this article, the role of brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (involved in synaptic plasticity and neurogenesis), and vascular endothelial growth factor, nerve growth factor, insulin-like growth factor 1, inflammatory markers, apoptotic factors, and antioxidant system was discussed in the modulation of exercise effects on memory. The role of intensity and duration of exercise, and type of memory task was also investigated. We also mentioned to the interaction of exercise with the function of neurotransmitter systems, which complicates the prediction of exercise effect via altering the level of BDNF. Eventually, we suggested that changes in the function of neurotransmitter systems following different types of exercise (depending on exercise intensity or age of onset) should be investigated in further studies. It seems that exercise-induced changes in the function of neurotransmitter systems may have a stronger role than age, type of memory task, or exercise intensity in modulating memory. Importantly, high levels of interactions between neurotransmitter systems and BDNF play a critical role in the modulation of exercise effects on memory performance.
Collapse
Affiliation(s)
- Mahshid Ebrahimnejad
- Department of Physiology, Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Paniz Azizi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Vahide Alipour
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Salar Vaseghi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, P.O. Box: 1419815477, Karaj, Iran.
| |
Collapse
|
15
|
Tang S, Zhang G, Ran Q, Nie L, Liu X, Pan Z, He L. Quantitative susceptibility mapping shows lower brain iron content in children with attention-deficit hyperactivity disorder. Hum Brain Mapp 2022; 43:2495-2502. [PMID: 35107194 PMCID: PMC9057088 DOI: 10.1002/hbm.25798] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/06/2022] [Accepted: 01/19/2022] [Indexed: 11/27/2022] Open
Abstract
To investigate the feasibility of quantitative susceptibility mapping in children with attention‐deficit hyperactivity disorder (ADHD), 53 children with ADHD aged 5–16 years were prospectively selected as the study group and 49 healthy children matched with age and gender were selected as the control group. All children underwent magnetic resonance imaging conventional sequence, 3D‐T1, and enhanced T2*‐weighted magnetic resonance angiography (ESWAN) sequence scanning. The iron content of brain regions was obtained through software postprocessing, and the iron content of brain regions of children with ADHD and healthy children was compared and analyzed to find out the characteristics of the iron content of brain regions of children with ADHD. The iron content in frontal lobe, globus pallidus, caudate nucleus, substantia nigra, putamen, and hippocampus of children with ADHD was lower than that of healthy children (p < .05). There was no significant difference in the content of iron in the left and right brain regions of children with ADHD (p > .05). The volume of frontal lobe and hippocampus of children with ADHD was lower than that of healthy children (p < .05). Iron content in brain areas such as globus pallidus, caudate nucleus, hippocampus, and putamen could distinguish children with ADHD (Area under curve [AUC] > 0.5, p < .05). Quantitative susceptibility mapping showed decreased iron content in some brain regions of children with ADHD.
Collapse
Affiliation(s)
- Shilong Tang
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, Chongqing, China
| | - Guanping Zhang
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, Chongqing, China
| | - Qiying Ran
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, Chongqing, China
| | - Lisha Nie
- GE Healthcare, MR Research China, Beijing, China
| | - Xianfan Liu
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, Chongqing, China
| | - Zhengxia Pan
- Department of Cardiovascular and Thoracic Surgery, Children's Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - Ling He
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, Chongqing, China
| |
Collapse
|
16
|
Michalovicz LT, Kelly KA, Miller DB, Sullivan K, O'Callaghan JP. The β-adrenergic receptor blocker and anti-inflammatory drug propranolol mitigates brain cytokine expression in a long-term model of Gulf War Illness. Life Sci 2021; 285:119962. [PMID: 34563566 PMCID: PMC9047058 DOI: 10.1016/j.lfs.2021.119962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 12/03/2022]
Abstract
Aims: Growing evidence suggests that Gulf War Illness (GWI) is the result of underlying neuroimmune dysfunction. For example, previously we found that several GWI-relevant organophosphate acetylcholinesterase inhibitors produce heightened neuroinflammatory responses following subchronic exposure to stress hormone as a mimic of high physiological stress. The goal of the current study was to evaluate the potential for the β-adrenergic receptor inhibitor and anti-inflammatory drug, propranolol, to treat neuroinflammation in a novel long-term mouse model of GWI. Main methods: Adult male C57BL/6J mice received a subchronic exposure to corticosterone (CORT) at levels mimicking high physiological stress followed by exposure to the sarin surrogate, diisopropyl fluorophosphate (DFP). These mice were then re-exposed to CORT every other week for a total of five weeks, followed by a systemic immune challenge with lipopolysaccharide (LPS). Animals receiving the propranolol treatment were given a single dose (20 mg/kg, i.p.) either four or 11 days prior to the LPS challenge. The potential anti-neuroinflammatory effects of propranolol were interrogated by analysis of cytokine mRNA expression. Key findings: We found that our long-term GWI model produces a primed neuroinflammatory response to subsequent immune challenge that is dependent upon GWI-relevant organophosphate exposure. Propranolol treatment abrogated the elaboration of inflammatory cytokine mRNA expression in the brain instigated in our model, having no treatment effects in non-DFP exposed groups. Significance: Our results indicate that propranolol may be a promising therapy for GWI with the potential to treat the underlying neuroinflammation associated with the illness.
Collapse
Affiliation(s)
- Lindsay T Michalovicz
- Health Effects Laboratory Division, Centers for Disease Control and Prevention-National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Kimberly A Kelly
- Health Effects Laboratory Division, Centers for Disease Control and Prevention-National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Diane B Miller
- Health Effects Laboratory Division, Centers for Disease Control and Prevention-National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | | | - James P O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention-National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|