1
|
Wang Z, Sun S, Liu Q, Kranfli AA, Nemes J, Sullan M, Hoisington A, Brenner LA, Skotak M, LaValle CR, Ge Y, Carr W, Haghighi F. Impact of prior exposures on biomarkers of blast during military tactical training. Front Neurol 2025; 16:1589742. [PMID: 40433620 PMCID: PMC12106048 DOI: 10.3389/fneur.2025.1589742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction Blast injuries and subclinical effects are of significant concern among those Service Members (SMs) participating in military operations and tactical trainings. Studies of SMs repeatedly exposed during training find concussion-like symptomology with transient decrements in neurocognitive performance, and alterations in blood biomarkers. How prior mild TBI (mTBI) history interacts with low-level blast (LLB) exposure, however, remains unexplored, which we investigate in the present study, to identify interindividual biomarker changes from LLB exposures influenced by prior history of mTBI. Methods Gene transcript and amyloid-beta (Aβ40 and Aβ42) protein levels were assayed using timeseries blood specimens collected at pre-blast, post-blast (within ~1 h), and follow-up-blast (~16 h) after LLB exposure for 30 SMs (age 30.3 ± 7.5) via RNA-seq and Single Molecule Array (SIMOA). Statistical models with timepoint and mTBI status interaction adjusted for age were used, and p-values adjusted for multiple testing. Results We found enrichment of genes involved in blood brain barrier, inflammatory, and immune responses associated with blast exposure, with significant elevated expression of target genes among SMs with mTBI history. Levels of Aβ40 and Aβ42 did not differ pre-blast vs. post/follow-up-blast LLB exposure when comparing SMs by prior mTBI history. Aβ40 and Aβ42 levels were significantly decreased in response to blast at the follow-up (~16 h) LLB exposure timepoint, concomitant with elevated expression of genes involved in amyloid-beta regulation and clearance in SMs with mTBI. Conclusion Findings show inter-individual differences in biomarker levels following exposures to blast that may be attributed to prior mTBI history.
Collapse
Affiliation(s)
- Zhaoyu Wang
- James J. Peters VA Medical Center, Bronx, NY, United States
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Shengnan Sun
- James J. Peters VA Medical Center, Bronx, NY, United States
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Qingkun Liu
- James J. Peters VA Medical Center, Bronx, NY, United States
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alis Askar Kranfli
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Jeffrey Nemes
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Molly Sullan
- Department of Physical Medicine and Rehabilitation, University of Colorado – Anschutz Medical Campus, Aurora, CO, United States
- Rocky Mountain Regional VA Medical Center, Aurora, CO, United States
| | - Andrew Hoisington
- Department of Physical Medicine and Rehabilitation, University of Colorado – Anschutz Medical Campus, Aurora, CO, United States
- Rocky Mountain Regional VA Medical Center, Aurora, CO, United States
- Air Force Institute of Technology, Wright-Patterson Airforce Base, OH, United States
| | - Lisa A. Brenner
- Department of Physical Medicine and Rehabilitation, University of Colorado – Anschutz Medical Campus, Aurora, CO, United States
- Rocky Mountain Regional VA Medical Center, Aurora, CO, United States
| | - Maciej Skotak
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | | | - Yongchao Ge
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Walter Carr
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Fatemeh Haghighi
- James J. Peters VA Medical Center, Bronx, NY, United States
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
2
|
He L, Ye J, Zhuang X, Shi J, Wu W. Omega-3 polyunsaturated fatty acids alleviate endoplasmic reticulum stress-induced neuroinflammation by protecting against traumatic spinal cord injury through the histone deacetylase 3/ peroxisome proliferator-activated receptor-γ coactivator pathway. J Neuropathol Exp Neurol 2024; 83:939-950. [PMID: 39190872 DOI: 10.1093/jnen/nlae094] [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: 08/29/2024] Open
Abstract
Omega-3 polyunsaturated fatty acids (ω-3 PUFAs) attenuate inflammatory responses in the central nervous system, leading to neuroprotective effects. Inhibition of histone deacetylase 3 (HDAC3) has neuroprotective effects after spinal cord injury (SCI) through the SIRT1 pathway, but the pathophysiological mechanisms of SCI are complex and the interactions between ω-3 PUFAs and organelles remain largely unknown. This study aimed to investigate the effect of ω-3 PUFAs on endoplasmic reticulum (ER) stress-induced neuroinflammation through the HDAC3/peroxisome proliferator-activated receptor-γ coactivator (PGC)-1ɑ pathway after SCI. To this end, a contusion-induced SCI rat model was established to evaluate the effects of ω-3 PUFAs on ER stress-mediated inflammation in SCI. ER stress was rapidly induced in spinal cord lesions after SCI and was significantly reduced after ω-3 PUFA treatment. Consistent with reduced ER stress, HDAC3 expression levels and inflammatory responses were decreased, and PGC-1ɑ expression levels were increased after SCI. We found that ω-3 PUFA treatment attenuated ER stress through HDAC3 inhibition, thereby reducing SCI-induced inflammation. Taken together, these results suggest a role for ω-3 PUFA in protecting against SCI-induced neuroinflammation and promoting neurological functional recovery by regulating the histone deacetylase 3/ peroxisome proliferator-activated receptor-γ coactivator pathway.
Collapse
Affiliation(s)
- Lijiang He
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jingfang Ye
- Department of Nursing Faculty, Quanzhou Medical College, Quanzhou, Fujian Province, China
| | - Xunrong Zhuang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jinnan Shi
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Wenhua Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
3
|
Wright B, King S, Suphioglu C. The Importance of Phosphoinositide 3-Kinase in Neuroinflammation. Int J Mol Sci 2024; 25:11638. [PMID: 39519189 PMCID: PMC11546674 DOI: 10.3390/ijms252111638] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroinflammation, characterised by the activation of immune cells in the central nervous system (CNS), plays a dual role in both protecting against and contributing to the progression of neurodegenerative diseases, such as Alzheimer's disease (AD) and multiple sclerosis (MS). This review explores the role of phosphoinositide 3-kinase (PI3K), a key enzyme involved in cellular survival, proliferation, and inflammatory responses, within the context of neuroinflammation. Two PI3K isoforms of interest, PI3Kγ and PI3Kδ, are specific to the regulation of CNS cells, such as microglia, astrocytes, neurons, and oligodendrocytes, influencing pathways, such as Akt, mTOR, and NF-κB, that control cytokine production, immune cell activation, and neuroprotection. The dysregulation of PI3K signalling is implicated in chronic neuroinflammation, contributing to the exacerbation of neurodegenerative diseases. Preclinical studies show promise in targeting neuronal disorders using PI3K inhibitors, such as AS605240 (PI3Kγ) and idelalisib (PI3Kδ), which have reduced inflammation, microglial activation, and neuronal death in in vivo models of AD. However, the clinical translation of these inhibitors faces challenges, including blood-brain barrier (BBB) permeability, isoform specificity, and long-term safety concerns. This review highlights the therapeutic potential of PI3K modulation in neuroinflammatory diseases, identifying key gaps in the current research, particularly in the need for brain-penetrating and isoform-specific inhibitors. These findings underscore the importance of future research to develop targeted therapies that can effectively modulate PI3K activity and provide neuroprotection in chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- Brock Wright
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| | - Samuel King
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| | - Cenk Suphioglu
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| |
Collapse
|
4
|
Armstrong RC, Sullivan GM, Perl DP, Rosarda JD, Radomski KL. White matter damage and degeneration in traumatic brain injury. Trends Neurosci 2024; 47:677-692. [PMID: 39127568 DOI: 10.1016/j.tins.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
Traumatic brain injury (TBI) is a complex condition that can resolve over time but all too often leads to persistent symptoms, and the risk of poor patient outcomes increases with aging. TBI damages neurons and long axons within white matter tracts that are critical for communication between brain regions; this causes slowed information processing and neuronal circuit dysfunction. This review focuses on white matter injury after TBI and the multifactorial processes that underlie white matter damage, potential for recovery, and progression of degeneration. A multiscale perspective across clinical and preclinical advances is presented to encourage interdisciplinary insights from whole-brain neuroimaging of white matter tracts down to cellular and molecular responses of axons, myelin, and glial cells within white matter tissue.
Collapse
Affiliation(s)
- Regina C Armstrong
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Military Traumatic Brain Injury Initiative (MTBI(2)), Bethesda, MD, USA.
| | - Genevieve M Sullivan
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Military Traumatic Brain Injury Initiative (MTBI(2)), Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Daniel P Perl
- Pathology, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Department of Defense - Uniformed Services University Brain Tissue Repository, Bethesda, MD, USA
| | - Jessica D Rosarda
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kryslaine L Radomski
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
5
|
Zhou Y, Zhang Y, Botchway BOA, Huang M, Liu X. Sestrin2 can alleviate endoplasmic reticulum stress to improve traumatic brain injury by activating AMPK/mTORC1 signaling pathway. Metab Brain Dis 2024; 39:439-452. [PMID: 38047978 DOI: 10.1007/s11011-023-01323-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/08/2023] [Indexed: 12/05/2023]
Abstract
Traumatic brain injury (TBI), as a serious central nervous system disease, can result in severe neurological dysfunction or even disability and death of patients. The early and effective intervention of secondary brain injury can improve the prognosis of TBI. Endoplasmic reticulum (ER) stress is one of the main reasons to recover TBI. ER stress inhibition may be beneficial in treating TBI. Sestrin2 is a crucial regulator of ER stress, and its activation can significantly improve TBI. In this paper, we analyze the biological function of sestrin2, the latest findings on ER stress, and the relationship between ER stress and TBI. We elucidate the relationship of sestrin2 inhibiting ER stress via activating the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin complex 1 (MTORC1) signaling. Finally, we elaborate on the possible role of sestrin2 in TBI and explain how its activation potentially improves TBI.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, 312000, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, 312000, China
| | | | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, 312000, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, 312000, China.
| |
Collapse
|
6
|
Yang Y, Lu D, Wang M, Liu G, Feng Y, Ren Y, Sun X, Chen Z, Wang Z. Endoplasmic reticulum stress and the unfolded protein response: emerging regulators in progression of traumatic brain injury. Cell Death Dis 2024; 15:156. [PMID: 38378666 PMCID: PMC10879178 DOI: 10.1038/s41419-024-06515-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/22/2024]
Abstract
Traumatic brain injury (TBI) is a common trauma with high mortality and disability rates worldwide. However, the current management of this disease is still unsatisfactory. Therefore, it is necessary to investigate the pathophysiological mechanisms of TBI in depth to improve the treatment options. In recent decades, abundant evidence has highlighted the significance of endoplasmic reticulum stress (ERS) in advancing central nervous system (CNS) disorders, including TBI. ERS following TBI leads to the accumulation of unfolded proteins, initiating the unfolded protein response (UPR). Protein kinase RNA-like ER kinase (PERK), inositol-requiring protein 1 (IRE1), and activating transcription factor 6 (ATF6) are the three major pathways of UPR initiation that determine whether a cell survives or dies. This review focuses on the dual effects of ERS on TBI and discusses the underlying mechanisms. It is suggested that ERS may crosstalk with a series of molecular cascade responses, such as mitochondrial dysfunction, oxidative stress, neuroinflammation, autophagy, and cell death, and is thus involved in the progression of secondary injury after TBI. Hence, ERS is a promising candidate for the management of TBI.
Collapse
Affiliation(s)
- Yayi Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Dengfeng Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Menghan Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Guangjie Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Yun Feng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Yubo Ren
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Xiaoou Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
7
|
Dai Y, Dong J, Wu Y, Zhu M, Xiong W, Li H, Zhao Y, Hammock BD, Zhu X. Enhancement of the liver's neuroprotective role ameliorates traumatic brain injury pathology. Proc Natl Acad Sci U S A 2023; 120:e2301360120. [PMID: 37339206 PMCID: PMC10293829 DOI: 10.1073/pnas.2301360120] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/19/2023] [Indexed: 06/22/2023] Open
Abstract
Traumatic brain injury (TBI) is a pervasive problem worldwide for which no effective treatment is currently available. Although most studies have focused on the pathology of the injured brain, we have noted that the liver plays an important role in TBI. Using two mouse models of TBI, we found that the enzymatic activity of hepatic soluble epoxide hydrolase (sEH) was rapidly decreased and then returned to normal levels following TBI, whereas such changes were not observed in the kidney, heart, spleen, or lung. Interestingly, genetic downregulation of hepatic Ephx2 (which encodes sEH) ameliorates TBI-induced neurological deficits and promotes neurological function recovery, whereas overexpression of hepatic sEH exacerbates TBI-associated neurological impairments. Furthermore, hepatic sEH ablation was found to promote the generation of A2 phenotype astrocytes and facilitate the production of various neuroprotective factors associated with astrocytes following TBI. We also observed an inverted V-shaped alteration in the plasma levels of four EET (epoxyeicosatrienoic acid) isoforms (5,6-, 8,9-,11,12-, and 14,15-EET) following TBI which were negatively correlated with hepatic sEH activity. However, hepatic sEH manipulation bidirectionally regulates the plasma levels of 14,15-EET, which rapidly crosses the blood-brain barrier. Additionally, we found that the application of 14,15-EET mimicked the neuroprotective effect of hepatic sEH ablation, while 14,15-epoxyeicosa-5(Z)-enoic acid blocked this effect, indicating that the increased plasma levels of 14,15-EET mediated the neuroprotective effect observed after hepatic sEH ablation. These results highlight the neuroprotective role of the liver in TBI and suggest that targeting hepatic EET signaling could represent a promising therapeutic strategy for treating TBI.
Collapse
Affiliation(s)
- Yongfeng Dai
- School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou510330, China
| | - Jinghua Dong
- School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou510330, China
| | - Yu Wu
- Research Center for Brain Health, Pazhou Lab, Guangzhou510330, China
- School of Psychology, Shenzhen University, Shenzhen518060, China
| | - Minzhen Zhu
- Research Center for Brain Health, Pazhou Lab, Guangzhou510330, China
| | - Wenchao Xiong
- School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
| | - Huanyu Li
- Research Center for Brain Health, Pazhou Lab, Guangzhou510330, China
| | - Yulu Zhao
- School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou510330, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology, University of California, Davis, CA95616
- University of California Davis Comprehensive Cancer Center, University of California, Davis, CA95616
| | - Xinhong Zhu
- School of Basic Medical Science, Southern Medical University, Guangzhou510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou510330, China
- School of Psychology, Shenzhen University, Shenzhen518060, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou510006, China
| |
Collapse
|
8
|
Feng Y, Lang J, Sun B, Yan Z, Zhao Z, Sun G. Atorvastatin prevents endoplasmic reticulum stress-mediated apoptosis via the Nrf2/HO-1 signaling pathway in TBI mice. Neurol Res 2023; 45:590-602. [PMID: 36681943 DOI: 10.1080/01616412.2023.2170905] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Our present study evaluated the neuroprotection effects of atorvastatin by inhibiting TBI-induced ER stress, as well as the potential role of the Nrf2/HO-1 pathway in experimental TBI. METHODS First, the mice were divided into four groups:sham, TBI, TBI+Vehicle and TBI+atorvastatin groups. The mice received atorvastatin (10 mg/kg/day) through intragastric gavage once a day for 3 days before TBI. In addition, Nrf2 WT and Nrf2 knockout mice were randomly divided into four groups: Nrf2+/+ TBI, Nrf2+/+ TBI+atorvastatin, Nrf2-/- TBI, and Nrf2-/- TBI+atorvastatin groups. Several neurobehavioral parameters were assessed post-TBI using mNSS, brain edema and the rotarod test, and the brain was isolated for molecular and biochemical analysis conducted through TUNEL staining and western blotting. . RESULTS The results showed that atorvastatin treatment significantly improved neurological deficits, alleviated brain edema, and apoptosis caused by TBI. Western blotting analysis showed that atorvastatin significantly suppressed ER stress and its related apoptotic pathway after TBI, which may be associated with the further activation of the Nrf2/HO-1 pathway. However, compared with the Nrf2+/+ TBI+Vehicle group, Nrf2 deficiency further aggravated neurological deficits and promoted ER stress-mediated apoptosis induced by TBI. Interestingly, atorvastatin failed to improve neurological deficits but reversed apoptosis, and the loss of the beneficial properties of anti-ER stress in the Nrf2-/- TBI mice. . CONCLUSIONS The results indicated that atorvastatin improves the neurologic functions and protects the brain from injury in the Nrf2+/+ TBI mice, primarily by counteracting ER stress-mediated apoptosis, which may be achieved through the activation of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Yan Feng
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China
| | - Jiadong Lang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China
| | - Boyu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China
| | - Zhongjie Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China
| |
Collapse
|
9
|
Ming Y, Deng Z, Tian X, Jia Y, Ning M, Cheng S. m6A Methyltransferase METTL3 Reduces Hippocampal Neuron Apoptosis in a Mouse Model of Autism Through the MALAT1/SFRP2/Wnt/β-catenin Axis. Psychiatry Investig 2022; 19:771-787. [PMID: 36327957 PMCID: PMC9633173 DOI: 10.30773/pi.2021.0370] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/05/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Hippocampal neuron apoptosis contributes to autism, while METTL3 has been documented to possess great potentials in neuron apoptosis. Our study probed into the role of METTL3 in neuron apoptosis in autism and to determine the underlying mechanism. METHODS Bioinformatics analysis was used to analyze expressed genes in autism samples. Institute of Cancer Research mice were treated with valproic acid to develop autism models. The function of METTL3 in autism-like symptoms in mice was analyzed with behavioral tests and histological examination of their hippocampal tissues. Primary mouse hippocampal neurons were extracted for in vitro studies. Downstream factors of METTL3 were explored and validated. RESULTS METTL3, MALAT1, and Wnt/β-catenin signaling were downregulated, while SFRP2 was upregulated in the hippocampal tissues of a mouse model of autism. METTL3 stabilized MALAT1 expression by promoting m6A modification of MALAT1. MALAT1 promoted SFRP2 methylation and led to reduced SFRP2 expression by recruiting DNMT1, DNMT3A, and DNMT3B to the promoter region of SFRP2. Furthermore, SFRP2 facilitated activation of the Wnt/β-catenin signaling. By this mechanism, METTL3 suppressed autism-like symptoms and hippocampal neuron apoptosis. CONCLUSION This research suggests that METTL3 can reduce autism-like symptoms and hippocampal neuron apoptosis by regulating the MALAT1/SFRP2/Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Yue Ming
- Department of Applied Psychology, College of Teacher Education, Qiqihar University, Qiqihar, China
| | - Zhihui Deng
- Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Xianhua Tian
- Department of Applied Psychology, College of Teacher Education, Qiqihar University, Qiqihar, China
| | - Yuerong Jia
- Department of Applied Psychology, College of Teacher Education, Qiqihar University, Qiqihar, China
| | - Meng Ning
- Department of Applied Psychology, College of Teacher Education, Qiqihar University, Qiqihar, China
| | - Shuhua Cheng
- Department of Applied Psychology, College of Teacher Education, Qiqihar University, Qiqihar, China
| |
Collapse
|
10
|
Revisiting Excitotoxicity in Traumatic Brain Injury: From Bench to Bedside. Pharmaceutics 2022; 14:pharmaceutics14010152. [PMID: 35057048 PMCID: PMC8781803 DOI: 10.3390/pharmaceutics14010152] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality. Consequences vary from mild cognitive impairment to death and, no matter the severity of subsequent sequelae, it represents a high burden for affected patients and for the health care system. Brain trauma can cause neuronal death through mechanical forces that disrupt cell architecture, and other secondary consequences through mechanisms such as inflammation, oxidative stress, programmed cell death, and, most importantly, excitotoxicity. This review aims to provide a comprehensive understanding of the many classical and novel pathways implicated in tissue damage following TBI. We summarize the preclinical evidence of potential therapeutic interventions and describe the available clinical evaluation of novel drug targets such as vitamin B12 and ifenprodil, among others.
Collapse
|
11
|
Huang TC, Luo L, Jiang SH, Chen C, He HY, Liang CF, Li WS, Wang H, Zhu L, Wang K, Guo Y. Targeting integrated stress response regulates microglial M1/M2 polarization and attenuates neuroinflammation following surgical brain injury in rat. Cell Signal 2021; 85:110048. [PMID: 34015470 DOI: 10.1016/j.cellsig.2021.110048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/01/2021] [Accepted: 05/16/2021] [Indexed: 12/24/2022]
Abstract
Integrated stress response (ISR) contributes to various neuropathological processes and acting as a therapy target in CNS injuries. However, the fundamental role of ISR in regulating microglial polarization remains largely unknown. Currently no proper pharmacological approaches to reverse microglia-driven neuroinflammation in surgical brain injury (SBI) have been reported. Here we found that inhibition of the crucial ISR effector, activating transcription factor 4 (ATF4), using the RNA interference suppressed the lipopolysaccharide (LPS)-stimulated microglial M1 polarization in vitro. Interestingly, counteracting ISR with a small-molecule ISR inhibitor (ISRIB) resulted in a significant microglial M1 towards M2 phenotype switching after LPS treatment. The potential underlying mechanisms may related to downregulate the intracellular NADPH oxidase 4 (NOX4) expression under the neuroinflammatory microenvironment. Notably, ISRIB ameliorated the infiltration of microglia and improved the neurobehavioral outcomes in the SBI rat model. Overall, our findings suggest that targeting ISR exerts a novel anti-inflammatory effect on microglia via regulating M1/M2 phenotype and may represent a potential therapeutic target to overcome neuroinflammation following SBI.
Collapse
Affiliation(s)
- Teng-Chao Huang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China; East China Institute of Digital Medical Engineering, Shangrao 334000, PR China
| | - Lun Luo
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Shi-Hai Jiang
- Department of Joint Replacement and Trauma Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China; Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany
| | - Chuan Chen
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Hai-Yong He
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Chao-Feng Liang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Wen-Sheng Li
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Hui Wang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Lei Zhu
- Department of Burns, Plastic & Reconstructive Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Kun Wang
- Department of Joint Replacement and Trauma Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China.
| | - Ying Guo
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China.
| |
Collapse
|
12
|
Jin R, Zhong W, Liu S, Wang M, Li G. Inhibition of PI3Kγ by AS605240 plus low-dose tissue plasminogen activator (tPA) combination improves thrombolytic therapy in a rat model of embolic stroke. Neurosci Lett 2020; 738:135339. [PMID: 32882317 PMCID: PMC8171661 DOI: 10.1016/j.neulet.2020.135339] [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: 06/29/2020] [Revised: 08/10/2020] [Accepted: 08/27/2020] [Indexed: 11/24/2022]
Abstract
Our previous study showed that PI3Kγ inhibition with AS605240 plus a standard rat-dose tPA (10 mg/kg) combination attenuates delayed tPA-induced brain hemorrhage and ameliorates acute stroke injury 3 days after ischemic stroke in rats. The purpose of this study was to investigate whether combining AS605240 with tPA can enhance thrombolytic efficacy, so that lower doses of tPA can be applied to improve long-term outcome after ischemic stroke. The results showed that AS605240 plus low-dose tPA (5 mg/kg) combination therapy at 4 h after stroke onset significantly reduced infarct volume and neurological deficits at 24 h after stroke compared with saline, AS605240 or low-dose tPA alone group. Importantly, the combination therapy significantly reduced the delayed tPA-associated brain hemorrhage. Moreover, the combination therapy significantly decreased the size of the residual embolus within the middle cerebral artery, which was associated with a decrease in plasma plasminogen activator inhibitor-1 (PAI-1) activity compared with saline and tPA alone. Finally, AS605240 plus low-dose tPA combination improved long-term outcome for at least 35 days after stroke compared with the saline-treated group. Taken together, these findings suggest that PI3Kγ inhibition with AS605240 might act as an adjunct approach for enhancing tPA thrombolytic efficacy in acute ischemic stroke.
Collapse
Affiliation(s)
- Rong Jin
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Wei Zhong
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Shan Liu
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Min Wang
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Guohong Li
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
13
|
Liu S, Jin R, Xiao AY, Zhong W, Li G. Inhibition of CD147 improves oligodendrogenesis and promotes white matter integrity and functional recovery in mice after ischemic stroke. Brain Behav Immun 2019; 82:13-24. [PMID: 31356925 PMCID: PMC6800638 DOI: 10.1016/j.bbi.2019.07.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 01/26/2023] Open
Abstract
White matter damage is an important contributor to long-term neurological deficit after stroke. Our previous study has shown that inhibition of CD147 ameliorates acute ischemic stroke in mice. In this study, we aimed to investigate whether inhibition of CD147 promotes white matter repair and long-term functional recovery after ischemic stroke.Male adult C57BL/6 mice were subjected to transient (1-h) middle cerebral artery occlusion (tMCAO). Anti-CD147 function-blocking antibody (αCD147) was injected intravenously once daily for 3 days beginning 4 h after onset of ischemia. Sensorimotor and cognitive functions were evaluated up to 28 days after stroke. We found that αCD147 treatment not only prevented neuronal and oligodendrocyte cell death in the acute phase, but also profoundly protected white matter integrity and reduced brain atrophy and tissue loss in the late phase, leading to improved sensorimotor and cognitive functions for at least 28 days after stroke. Mechanistically, we found that αCD147 treatment increased the number of proliferating NG2(+)/PDGFRα(+) oligodendrocyte precursor cells (OPCs) and newly generated mature APC(+)/Sox10(+) oligodendrocytes after stroke, possibly through upregulation of SDF-1/CXCR4 axis in OPCs. In conclusion, inhibition of CD147 promotes long-term functional recovery after stroke, at least in part, by enhancing oligodendrogenesis and white matter repair.
Collapse
Affiliation(s)
- Shan Liu
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Rong Jin
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Adam Y Xiao
- The Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA
| | - Wei Zhong
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Guohong Li
- From the Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|