1
|
Gupta S, Gupta AK, Mehan S, Khan Z, Gupta GD, Narula AS. Disruptions in cellular communication: Molecular interplay between glutamate/NMDA signalling and MAPK pathways in neurological disorders. Neuroscience 2025:S0306-4522(25)00023-5. [PMID: 39809360 DOI: 10.1016/j.neuroscience.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Neurological disorders significantly impact the central nervous system, contributing to a growing public health crisis globally. The spectrum of these disorders includes neurodevelopmental and neurodegenerative diseases. This manuscript reviews the crucial roles of cellular signalling pathways in the pathophysiology of these conditions, focusing primarily on glutaminase/glutamate/NMDA receptor signalling, alongside the mitogen-activated protein kinase (MAPK) pathways-ERK1/2, C-JNK, and P38 MAPK. Activation of these pathways is often correlated with neuronal excitotoxicity, apoptosis, and inflammation, leading to many other pathological conditions such as traumatic brain injury, stroke, and brain tumor. The interplay between glutamate overstimulation and MAPK signalling exacerbates neurodegenerative processes, underscoring the complexity of cellular communication in maintaining neuronal health. Dysfunctional signalling alters synaptic plasticity and neuronal survival, contributing to cognitive impairments in various neurological diseases. The manuscript emphasizes the potential of targeting these signalling pathways for therapeutic interventions, promoting neuroprotection and reducing neuroinflammation. Incorporating insights from precision medicine and innovative drug delivery systems could enhance treatment efficacy. Overall, understanding the intricate mechanisms of these pathways is essential for developing effective strategies to mitigate the impact of neurological disorders and improve patient outcomes. This review highlights the necessity for further exploration into these signalling cascades to facilitate advancements in therapeutic approaches, ensuring better prognoses for individuals affected by neurological conditions.
Collapse
Affiliation(s)
- Sumedha Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Abhishek Kumar Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India. https://mehanneuroscience.org
| | - Zuber Khan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
2
|
Cui E, Tang P, Zhu X, Lv M, Wang S, Xue Y, Li C, Zhao S. Network Pharmacology Combined with an Experimental Validation Study to Reveal the Effect and Mechanism of Eucommia ulmoides Leaf Polysaccharide against Immunomodulation. Foods 2023; 12:foods12051062. [PMID: 36900578 PMCID: PMC10001223 DOI: 10.3390/foods12051062] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
In the present study, the immuno-enhancing effect of Eucommia ulmoides leaf polysaccharide (ELP) was investigated in immunosuppressed mice induced by cyclophosphamide (CTX). To evaluate the immune enhancement mechanism of ELP, the immunoregulation effect of ELP was evaluated in vitro and in vivo. ELP is primarily composed of arabinose (26.61%), galacturonic acid (25.1%), galactose (19.35%), rhamnose (16.13%), and a small amount of glucose (12.9%). At 1000~5000 μg·mL-1, ELP could significantly enhance the proliferation and the phagocytosis of macrophages in vitro. Additionally, ELP could protect immune organs, reduce pathological damage, and reverse the decrease in the hematological indices. Moreover, ELP significantly increased the phagocytic index, enhanced the ear swelling response, augmented the production of inflammatory cytokines, and markedly up-regulated the expression of IL-1β, IL-6, and TNF-α mRNA levels. Furthermore, ELP improved phosphorylated p38, ERK1/2, and JNK levels, suggesting that MAPKs might be involved in immunomodulatory effects. The results provide a theoretical foundation for exploring the immune modulation function of ELP as a functional food.
Collapse
|
3
|
Zhang W, Wang YD, Xing YJ, Liu PJ, Yang JH. Silencing of circ-NT5C2 retards the progression of IL-1β-induced osteoarthritis in an in vitro cell model by targeting the miR-142-5p/NAMPT axis. Microbiol Immunol 2023; 67:129-141. [PMID: 36540014 DOI: 10.1111/1348-0421.13046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/15/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Osteoarthritis (OA) is a degenerative disease that occurs mostly in the elderly, and its specific pathogenesis is still unknown, but recent studies have found that circular RNA generally display aberrant expression in OA. Our study explored the expression characteristics and mechanism of action of circ-NT5C2 in OA. Circ-NT5C2, microRNA-142-5p (miR-142-5p), and nicotinamide phosphoribosyltransferase (NAMPT) mRNA levels were measured using RT-qPCR. Western blot was employed to assess the protein level of NAMPT and extracellular matrix (ECM) production-related markers. The viability, proliferation, apoptosis and inflammation were examined using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay, 5-ethynyl-2'-deoxyuridine (EdU) assay, flow cytometry, and enzyme-linked immunosorbent assay, respectively. Relationship between miR-142-5p and circ-NT5C2 or NAMPT was demonstrated by dual-luciferase reporter system and RNA immunoprecipitation assay. We reported that circ-NT5C2 and NAMPT were greatly upregulated, and miR-142-5p level was constrained in OA tissues and in a cell model. Circ-NT5C2 silencing alleviated IL-1β-induced inhibitory effects on chondrocyte proliferation and ECM generation, meanwhile the promotional role of IL-1β on chondrocyte apoptosis and inflammation was also weakened. The targeting relationship of miR-142-5p with either circ-NT5C2 or NAMPT was confirmed. Knockdown of miR-142-5p reversed the suppressive effects of circ-NT5C2 silencing on the OA progression in vitro, and NAMPT overexpression also attenuated the effects of miR-142-5p upregulation in an OA cell model. Collectively, circ-NT5C2 accelerated the OA process by targeting the miR-142-5p/NAMPT axis. This study provides valuable information to find a better treatment for OA.
Collapse
Affiliation(s)
- Wei Zhang
- The Second Department of Bone Engineering, Xingyuan Hospital, Yulin City, China
| | - Yan-Dong Wang
- The Second Department of Bone Engineering, Xingyuan Hospital, Yulin City, China
| | - Yong-Jun Xing
- The Second Department of Bone Engineering, Xingyuan Hospital, Yulin City, China
| | - Peng-Jun Liu
- The Second Department of Bone Engineering, Xingyuan Hospital, Yulin City, China
| | - Jian-Hui Yang
- Department of Pain, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
4
|
Alexandris AS, Ryu J, Rajbhandari L, Harlan R, McKenney J, Wang Y, Aja S, Graham D, Venkatesan A, Koliatsos VE. Protective effects of NAMPT or MAPK inhibitors and NaR on Wallerian degeneration of mammalian axons. Neurobiol Dis 2022; 171:105808. [PMID: 35779777 PMCID: PMC10621467 DOI: 10.1016/j.nbd.2022.105808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/14/2022] [Accepted: 06/25/2022] [Indexed: 01/23/2023] Open
Abstract
Wallerian degeneration (WD) is a conserved axonal self-destruction program implicated in several neurological diseases. WD is driven by the degradation of the NAD+ synthesizing enzyme NMNAT2, the buildup of its substrate NMN, and the activation of the NAD+ degrading SARM1, eventually leading to axonal fragmentation. The regulation and amenability of these events to therapeutic interventions remain unclear. Here we explored pharmacological strategies that modulate NMN and NAD+ metabolism, namely the inhibition of the NMN-synthesizing enzyme NAMPT, activation of the nicotinic acid riboside (NaR) salvage pathway and inhibition of the NMNAT2-degrading DLK MAPK pathway in an axotomy model in vitro. Results show that NAMPT and DLK inhibition cause a significant but time-dependent delay of WD. These time-dependent effects are related to NMNAT2 degradation and changes in NMN and NAD+ levels. Supplementation of NAMPT inhibition with NaR has an enhanced effect that does not depend on timing of intervention and leads to robust protection up to 4 days. Additional DLK inhibition extends this even further to 6 days. Metabolite analyses reveal complex effects indicating that NAMPT and MAPK inhibition act by reducing NMN levels, ameliorating NAD+ loss and suppressing SARM1 activity. Finally, the axonal NAD+/NMN ratio is highly predictive of cADPR levels, extending previous cell-free evidence on the allosteric regulation of SARM1. Our findings establish a window of axon protection extending several hours following injury. Moreover, we show prolonged protection by mixed treatments combining MAPK and NAMPT inhibition that proceed via complex effects on NAD+ metabolism and inhibition of SARM1.
Collapse
Affiliation(s)
| | - Jiwon Ryu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Labchan Rajbhandari
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Harlan
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - James McKenney
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yiqing Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan Aja
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - David Graham
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vassilis E Koliatsos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Aprepitant as plausible inhibitor of MAPK/ERK2 pathway to ameliorate neurological deficits post traumatic brain injury. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
6
|
Exosomes derived from bone marrow mesenchymal stem cells attenuate neurological damage in traumatic brain injury by alleviating glutamate-mediated excitotoxicity. Exp Neurol 2022; 357:114182. [PMID: 35901975 DOI: 10.1016/j.expneurol.2022.114182] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) is one of the major contributors to disability and death worldwide. Glutamate-mediated excitotoxicity, one of the secondary injuries occurring after TBI, leads to extreme neuronal apoptosis, and can be a potential target for intervention. Bone marrow mesenchymal stem cells-derived exosomes (BMSCs-Exos) have demonstrated neuroprotective effects on TBI. However, their precise role and the underlying mechanism by which they regulate glutamate-mediated excitotoxicity have not yet been determined. Therefore, this study aimed to determine whether BMSCs-Exos alleviate glutamate excitotoxicity post-TBI and their associated mechanism. METHODS BMSCs-Exos were extracted from the BMSCs incubation medium and identified by transmission electron microscopy, nanoparticle trafficking analysis, and western blotting. The neuroprotective effects of BMSCs-Exos on glutamate excitotoxicity were investigated in the glutamate-mediated excitotoxicity neuronal cell model and the TBI rat model (TBI induced by controlled cortical impact) using western blotting and TUNEL assay. Cortical lesion samples were collected post-TBI on day-1 and day-14 to study histology. In addition, cortical lesion volume on days 1, 3 and 7 following TBI was determined using T2-weighted magnetic resonance imaging (MRI), and cognitive function was assessed at 4 weeks following TBI using the Morris water maze (MWM) test. RESULTS BMSC-Exos were observed to be spherical with a mean diameter of 109.9 nm, and expressed exosomal markers CD9, CD81 and TSg101. BMSCs-Exos were efficiently endocytosed by astrocytes after co-incubation for 24 h. In vitro studies revealed that 125 μM of glutamate significantly induced neuronal apoptosis, which was attenuated by BMSCs-Exos in astrocyte-neuron co-cultures. This attenuation was mediated by the upregulation of glutamate transporter-1 (GLT-1) level and the downregulation of p-p38 MAPK level in astrocytes. Similar results were obtained in vivo, wherein we verified that PKH67-labeled BMSCs-Exos administered intravenously could reach the perilesional cortex crossing the blood-brain barrier and significantly reduce glutamate levels in the perilesional cortex of the TBI rat, accompanied by increased GLT-1 level and downregulation in p-p38 MAPK level. Additionally, western blotting and TUNEL staining also revealed that BMSCs-Exos significantly downregulated the expression of pro-apoptosis markers, including cleaved caspase-3 and cleaved caspase-9, and attenuated neuronal apoptosis following TBI. Immunohistochemical analysis and Nissl staining showed that BMSCs-Exos significantly increased GLT-1-positive cells, and the number of apoptotic neurons decreased in the perilesional cortex. Moreover, MRI and MWM results revealed that BMSCs-Exos significantly minimized cortical lesion volume and ameliorated cognitive function after TBI. The underlying neuroprotective mechanism of BMSCs-Exos may be due to an increase in GLT-1 level in astrocytes by blocking the p38 MAPK signaling pathway. CONCLUSION Taken together, our findings demonstrate that the implementation of BMSCs-Exos may be an effective prospective therapy for attenuating post-TBI neurological damage.
Collapse
|
7
|
Jiang Y, Chen Y, Huang C, Xia A, Wang G, Liu S. Hyperbaric oxygen therapy improves neurological function via the p38-MAPK/CCL2 signaling pathway following traumatic brain injury. Neuroreport 2021; 32:1255-1262. [PMID: 34494990 PMCID: PMC8432607 DOI: 10.1097/wnr.0000000000001719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/13/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The anti-inflammatory mechanisms of hyperbaric oxygenation (HBO) treatment on traumatic brain injury (TBI)-induced neuroinflammation remain unclear. The aim of this study was expected the effect of HBO on CCL2-related signaling pathway following severe TBI in rats. METHODS The severe TBI model in rats was induced by controlled cortical impact. TBI rats were treated with CCR2 antagonist, p38 inhibitor, or HBO. Modified neurological severity scores and Morris water maze were used to evaluate neurological and cognitive function. The expression levels of CCL2 and CCR2 were measured by ELISA and real-time fluorescence quantitative PCR. Phospho-p38 expression was analyzed by western blotting. RESULTS TBI-induced upregulation of CCL2, CCR2, and p38 in the injured cortex. Application of CCR2 antagonist improved neurological and cognitive function of TBI rats. Application of p38 inhibitor decreased expression of CCL2 and CCR2 in the injured of TBI rats, meanwhile improved neurological and cognitive function. HBO improved neurological and cognitive function by decreasing the expressions of CCL2, CCR2, and phospho-p38. CONCLUSIONS This study indicates that the p38-MAPK-CCL2 signaling pathway could mediate neuroinflammation and HBO therapy can modulate neuroinflammation by modulating the p38-MAPK-CCL2 signaling pathways following TBI. This study may provide theoretical evidence for HBO treatment in the treatment of TBI.
Collapse
Affiliation(s)
- Yingzi Jiang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University
- Department of Clinical Medicine, School of Medicine, Nantong University
| | - Yuwen Chen
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University
- Department of Clinical Medicine, School of Medicine, Nantong University
| | - Chunling Huang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University
- Department of Clinical Medicine, School of Medicine, Nantong University
| | - Anqi Xia
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University
- Department of Clinical Medicine, School of Medicine, Nantong University
| | - Guohua Wang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Su Liu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University
| |
Collapse
|
8
|
Adipokines as Immune Cell Modulators in Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms221910845. [PMID: 34639186 PMCID: PMC8509121 DOI: 10.3390/ijms221910845] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS), a chronic inflammatory and demyelinating disease of the central nervous system (CNS), is a major clinical and societal problem, which has a tremendous impact on the life of patients and their proxies. Current immunomodulatory and anti-inflammatory therapies prove to be relatively effective; however, they fail to concomitantly stop ongoing neurological deterioration and do not reverse acquired disability. The proportion to which genetic and environmental factors contribute to the etiology of MS is still incompletely understood; however, a recent association between MS etiology and obesity was shown, with obesity greatly increasing the risk of developing MS. An altered balance of adipokines, which are white adipose tissue (WAT) hormones, plays an important role in the low-grade chronic inflammation during obesity by their pervasive modification of local and systemic inflammation. Vice versa, inflammatory factors secreted by immune cells affect adipokine function. To explore the role of adipokines in MS pathology, we will here review the reciprocal effects of adipokines and immune cells and summarize alterations in adipokine levels in MS patient cohorts. Finally, we will discuss proof-of-concept studies demonstrating the therapeutic potential of adipokines to target both neuroinflammation and neurodegeneration processes in MS.
Collapse
|
9
|
Song JH, Jia HY, Shao TP, Liu ZB, Zhao YP. Hydrogen gas post-conditioning alleviates cognitive dysfunction and anxiety-like behavior in a rat model of subarachnoid hemorrhage. Exp Ther Med 2021; 22:1121. [PMID: 34504575 PMCID: PMC8383778 DOI: 10.3892/etm.2021.10555] [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: 02/23/2021] [Accepted: 07/19/2021] [Indexed: 01/14/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) results in high rates of mortality and lasting disability. Hydrogen gas (H2) is an antioxidant with demonstrated neuroprotective efficacy. The present study examined the therapeutic efficacy of H2 inhalation on early brain injury following experimental SAH in rats and the potential underlying molecular mechanisms. The rats were randomly separated into three groups (n=36 per group): Sham, SAH and SAH + H2. Endovascular perforation of the right internal carotid artery was used to establish SAH. After perforation, rats in the SAH + H2 group inhaled 2.9% H2 with regular oxygen for 2 h. Then, 24 h post-SAH, TUNEL staining was used to detect apoptotic neurons, and both immunostaining and western blotting were conducted to examine changes in p38 MAPK activity and the expression levels of apoptotic regulators (Bcl-2, Bax and cleaved caspase-3) in the ventromedial prefrontal cortex. Then, 30 day post-SAH, Nissl staining was performed to detect neuronal injury, brain MRI was conducted to detect gross changes in brain structure and metabolism, the open field test was used to assess anxiety and the novel object recognition test was performed to assess memory. H2 inhalation following experimental SAH stabilized brain metabolites, improved recognition memory and reduced anxiety-like behavior, the neuronal apoptosis rate, phosphorylated p38 MAPK expression, cleaved caspase-3 expression and the Bax/Bcl-2 ratio. Collectively, the present results suggested that H2 inhalation can alleviate SAH-induced cognitive impairment, behavioral abnormalities and neuronal apoptosis in rats, possibly via inhibition of the p38 MAPK signal pathway.
Collapse
Affiliation(s)
- Jing-Hua Song
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Hong-Yan Jia
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Tian-Peng Shao
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Zhi-Bao Liu
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yuan-Ping Zhao
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
10
|
Lusardi TA, Sandau US, Sakhanenko NA, Baker SCB, Wiedrick JT, Lapidus JA, Raskind MA, Li G, Peskind ER, Galas DJ, Quinn JF, Saugstad JA. Cerebrospinal Fluid MicroRNA Changes in Cognitively Normal Veterans With a History of Deployment-Associated Mild Traumatic Brain Injury. Front Neurosci 2021; 15:720778. [PMID: 34580583 PMCID: PMC8463659 DOI: 10.3389/fnins.2021.720778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/27/2021] [Indexed: 01/09/2023] Open
Abstract
A history of traumatic brain injury (TBI) increases the odds of developing Alzheimer's disease (AD). The long latent period between injury and dementia makes it difficult to study molecular changes initiated by TBI that may increase the risk of developing AD. MicroRNA (miRNA) levels are altered in TBI at acute times post-injury (<4 weeks), and in AD. We hypothesized that miRNA levels in cerebrospinal fluid (CSF) following TBI in veterans may be indicative of increased risk for developing AD. Our population of interest is cognitively normal veterans with a history of one or more mild TBI (mTBI) at a chronic time following TBI. We measured miRNA levels in CSF from three groups of participants: (1) community controls with no lifetime history of TBI (ComC); (2) deployed Iraq/Afghanistan veterans with no lifetime history of TBI (DepC), and (3) deployed Iraq/Afghanistan veterans with a history of repetitive blast mTBI (DepTBI). CSF samples were collected at the baseline visit in a longitudinal, multimodal assessment of Gulf War veterans, and represent a heterogenous group of male veterans and community controls. The average time since the last blast mTBI experienced was 4.7 ± 2.2 years [1.5 - 11.5]. Statistical analysis of TaqManTM miRNA array data revealed 18 miRNAs with significant differential expression in the group comparisons: 10 between DepTBI and ComC, 7 between DepC and ComC, and 8 between DepTBI and DepC. We also identified 8 miRNAs with significant differential detection in the group comparisons: 5 in DepTBI vs. ComC, 3 in DepC vs. ComC, and 2 in DepTBI vs. DepC. When we applied our previously developed multivariable dependence analysis, we found 13 miRNAs (6 of which are altered in levels or detection) that show dependencies with participant phenotypes, e.g., ApoE. Target prediction and pathway analysis with miRNAs differentially expressed in DepTBI vs. either DepC or ComC identified canonical pathways highly relevant to TBI including senescence and ephrin receptor signaling, respectively. This study shows that both TBI and deployment result in persistent changes in CSF miRNA levels that are relevant to known miRNA-mediated AD pathology, and which may reflect early events in AD.
Collapse
Affiliation(s)
- Theresa A Lusardi
- Knight Cancer Institute, Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Ursula S Sandau
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | | | - Sarah Catherine B Baker
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Jack T Wiedrick
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, United States
| | - Jodi A Lapidus
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, United States
| | - Murray A Raskind
- Northwest Mental Illness, Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - Ge Li
- Northwest Mental Illness, Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States.,Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
| | - Elaine R Peskind
- Northwest Mental Illness, Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - David J Galas
- Pacific Northwest Research Institute, Seattle, WA, United States
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States.,Parkinson Center and Movement Disorders Program, School of Medicine, Oregon Health & Science University, Portland, OR, United States.,Portland VAMC Parkinson's Disease Research, Education, and Clinical Center, Portland, OR, United States
| | - Julie A Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
11
|
Li C, Wu LE. Risks and rewards of targeting NAD + homeostasis in the brain. Mech Ageing Dev 2021; 198:111545. [PMID: 34302821 DOI: 10.1016/j.mad.2021.111545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 01/29/2023]
Abstract
Strategies to correct declining nicotinamide adenine dinucleotide (NAD+) levels in neurological disease and biological ageing are promising therapeutic candidates. These strategies include supplementing with NAD+ precursors, small molecule activation of NAD+ biosynthetic enzymes, and treatment with small molecule inhibitors of NAD+ consuming enzymes such as CD38, SARM1 or members of the PARP family. While these strategies have shown efficacy in animal models of neurological disease, each of these has the mechanistic potential for adverse events that could preclude their preclinical use. Here, we discuss the implications of these strategies for treating neurological diseases, including potential off-target effects that may be unique to the brain.
Collapse
Affiliation(s)
- Catherine Li
- School of Medical Sciences, UNSW Sydney, NSW, 2052, Australia
| | - Lindsay E Wu
- School of Medical Sciences, UNSW Sydney, NSW, 2052, Australia.
| |
Collapse
|
12
|
Jiang H, Li H, Cao Y, Zhang R, Zhou L, Zhou Y, Zeng X, Wu J, Wu D, Wu D, Guo X, Li X, Wu H, Li P. Effects of cannabinoid (CBD) on blood brain barrier permeability after brain injury in rats. Brain Res 2021; 1768:147586. [PMID: 34289379 DOI: 10.1016/j.brainres.2021.147586] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/23/2021] [Accepted: 07/14/2021] [Indexed: 01/30/2023]
Abstract
Cannabidiol is a natural herbal medicine known to protect the brain from traumatic brain injury (TBI). Here, a TBI rat model was established, with cannabidiol administered intraperitoneally at doses of 5, 10, or 20 mg/kg, 30 min before surgery and 6 h after surgery until sacrifice. Brain water content, body weight, and modified neurological severity scores were determined, and enzyme-linked immunosorbent assay, immunofluorescence staining, hematoxylin and eosin staining, Nissl staining, Evans-blue dye extravasation, and western blotting were performed. Results showed that cannabidiol decreased the number of aquaporin-4-positive and glial fibrillary acidic protein-positive cells. Cannabidiol also significantly reduced the protein levels of proinflammatory cytokines (TNF-α and IL-1β) and significantly increased the expression of tight junction proteins (claudin-5 and occludin). Moreover, cannabidiol administration significantly mitigated water content in the brain after TBI and blood-brain barrier disruption and ameliorated the neurological deficit score after TBI. Cannabidiol administration improved the integrity and permeability of the blood-brain barrier and reduced edema in the brain after TBI.
Collapse
Affiliation(s)
- Hongyan Jiang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China; Department of Pathology, Suining Central Hospital, Suining 629000, China
| | - Hengxi Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yan Cao
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Ruilin Zhang
- Department of Forensic Medicine of Kunming Medical University, Kunming 650500, China
| | - Lei Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming 650500, China
| | - Ying Zhou
- Department of Kunming Medical University Electron Microscope Laboratory, Kunming Medical University, Kunming 650500, China
| | - Xiaofeng Zeng
- Department of Forensic Medicine of Kunming Medical University, Kunming 650500, China
| | - Jia Wu
- Department of Morphology Laboratory, Kunming Medical University, Kunming 650500, China
| | - Douwei Wu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Deye Wu
- Department of Human Anatomy and Histology/Embryology, Qilu Medical University, Zibo 255213, Shandong, China
| | - Xiaobing Guo
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xiaowen Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Haiying Wu
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical University, Kunming 650032, China.
| | - Ping Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
13
|
Xu Y, Yu L, Liu Y, Tang X, Wang X. Lipopolysaccharide-Induced Microglial Neuroinflammation: Attenuation by FK866. Neurochem Res 2021; 46:1291-1304. [PMID: 33713324 DOI: 10.1007/s11064-021-03267-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Alleviating microglia-mediated neuroinflammation bears great promise to reduce neurodegeneration. Nicotinamide phosphoribosyltransferase (NAMPT) may exert cytokine-like effect in the brain. However, it remains unclear about role of NAMPT in microglial inflammation. Also, it remains unknown about effect of NAMPT inhibition on microglial inflammation. In the present study, we observed that FK866 (a specific noncompetitive NAMPT inhibitor) dose-dependently inhibited lipopolysaccharide (LPS)-induced proinflammatory mediator (interleukin (IL)-6, IL-1β, inducible nitric oxide synthase, nitric oxide and reactive species) level increase in BV2 microglia cultures. FK866 also significantly inhibited LPS-induced polarization change in microglia. Furthermore, LPS significantly increased NAMPT expression and nuclear factor kappa B (NF-κB) phosphorylation in microglia. FK866 significantly decreased NAMPT expression and NF-κB phosphorylation in LPS-treated microglia. Finally, conditioned medium from microglia cultures co-treated with FK866 and LPS significantly increased SH-SY5Y and PC12 cell viability compared with conditioned medium from microglia cultures treated with LPS alone. Our study strongly indicates that NAMPT may be a promising target for microglia modulation and NAMPT inhibition may attenuate microglial inflammation.
Collapse
Affiliation(s)
- Yaling Xu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Lijia Yu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Ying Liu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Xiaohui Tang
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
14
|
Zou X, Xie L, Wang W, Zhao G, Tian X, Chen M. FK866 alleviates cerebral pyroptosis and inflammation mediated by Drp1 in a rat cardiopulmonary resuscitation model. Int Immunopharmacol 2020; 89:107032. [PMID: 33045576 DOI: 10.1016/j.intimp.2020.107032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Dynamin-related protein 1 (Drp1) mediates mitochondrial fission and triggers NLRP3 inflammasome activation. FK866 (a NAMPT inhibitor) exerts a neuroprotective effect in ischemia/reperfusion injury through the suppression of mitochondrial dysfunction. We explored the effects of FK866 on pyroptosis and inflammation mediated by Drp1 in a cardiac arrest/cardiopulmonary resuscitation (CA/CPR) rat model. METHODS Healthy male Sprague-Dawley rats were subjected to 7 min CA by trans-esophageal electrical stimulation followed by CPR. The surviving rats were treated with FK866 (a selective inhibitor of NAMPT), Mdivi-1 (Drp1 inhibitor), FK866 + Mdivi-1, or vehicle and then underwent 24 h reperfusion. Hematoxylin and eosin staining and immunohistochemistry (to detect NSE) were used to evaluate brain injury. We performed immunofluorescent staining to analyze NLRP3 and GSDMD expression in microglia or astrocytes and western blot to determine expression of NLRP3, IL-1β, GSDMD, Drp1, and Mfn2. Transmission electron microscopy was used to observe mitochondria. RESULTS FK866 significantly decreased pathological damage to brain tissue, inhibited the activation of NLRP3 in microglia or astrocytes, downregulated the expression of NLRP3, IL-1β, GSDMD, p-Drp1 protein, upregulated Mfn2 and improve mitochondrial morphology. CONCLUSIONS Our results demonstrated that FK866 protects the brain against ischemia-reperfusion injury in rats after CA/CPR by inhibiting pyroptosis and inflammation mediated by Drp1.
Collapse
Affiliation(s)
- Xinsen Zou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Lu Xie
- Department of Physiology, Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Wenyan Wang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Gaoyang Zhao
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Xinyue Tian
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Menghua Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China.
| |
Collapse
|