1
|
Toker D, Thum JA, Guang J, Miyamoto H, Yamakawa K, Vespa PM, Schnakers C, Bari AA, Hudson A, Pouratian N, Monti MM. An AI-Driven Model of Consciousness, Its Disorders, and Their Treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618720. [PMID: 39463979 PMCID: PMC11507942 DOI: 10.1101/2024.10.16.618720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Understanding the neural signatures of consciousness and the mechanisms underlying its disorders, such as coma and unresponsive wakefulness syndrome, remains a critical challenge in neuroscience. In this study, we present a novel computational approach for the in silico discovery of neural correlates of consciousness, the mechanisms driving its disorders, and potential treatment strategies. Inspired by generative adversarial networks, which have driven recent advancements in generative artificial intelligence (AI), we trained deep neural networks to detect consciousness across multiple brain areas and species, including humans. These networks were then integrated with a genetic algorithm to optimize a brain-wide mean-field model of neural electrodynamics. The result is a realistic simulation of conscious brain states and disorders of consciousness (DOC), which not only recapitulates known mechanisms of unconsciousness but also predicts novel causes expected to lead to these conditions. Beyond simulating DOC, our model provides a platform for exploring therapeutic interventions, specifically deep brain stimulation (DBS), which has shown promise in improving levels of awareness in DOC in over five decades of study. We systematically applied simulated DBS to various brain regions at a wide range of frequencies to identify an optimal paradigm for reigniting consciousness in this cohort. Our findings suggest that in addition to previously studied thalamic and pallidal stimulation, high-frequency stimulation of the subthalamic nucleus, a relatively underexplored target in DOC, may hold significant promise for restoring consciousness in this set of disorders.
Collapse
|
2
|
Tolstova T, Dotsenko E, Luzgina N, Rusanov A. Preconditioning of Mesenchymal Stem Cells Enhances the Neuroprotective Effects of Their Conditioned Medium in an Alzheimer's Disease In Vitro Model. Biomedicines 2024; 12:2243. [PMID: 39457556 PMCID: PMC11504366 DOI: 10.3390/biomedicines12102243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) develops as a result of oxidative damage to neurons and chronic inflammation of microglia. These processes can be influenced by the use of a conditioned medium (CM) derived from mesenchymal stem cells (MSCs). The CM contains a wide range of factors that have neurotrophic, antioxidant, and anti-inflammatory effects. In addition, the therapeutic potential of the CM can be further enhanced by pretreating the MSCs to increase their paracrine activity. The current study aimed to investigate the neuroprotective effects of CM derived from MSCs, which were either activated by a TLR3 ligand or exposed to CoCl2, a hypoxia mimetic (pCM or hCM, respectively), in an in vitro model of AD. METHODS We have developed a novel in vitro model of AD that allows us to investigate the neuroprotective and anti-inflammatory effects of MSCs on induced neurodegeneration in the PC12 cell line and the activation of microglia using THP-1 cells. RESULTS This study demonstrates for the first time that pCM and hCM exhibit more pronounced immunosuppressive effects on proinflammatory M1 macrophages compared to CM derived from untreated MSCs (cCM). This may help prevent the development of neuroinflammation by balancing the M1 and M2 microglial phenotypes via the decreased secretion of proinflammatory cytokines (IL-1β, IL-6, and TNF-α) and increased secretion of IL-4, as well as the expression of IL-10 and TGF-β by macrophages. Moreover, a previously unknown increase in the neurotrophic properties of hCM was discovered, which led to an increase in the viability of neuron-like PC12 cells under H2O2-induced oxidative-stress conditions. These results are likely associated with an increase in the production of growth factors, including vascular endothelial growth factor (VEGF). In addition, the neuroprotective effects of CM from preconditioned MSCs are also mediated by the activation of the Nrf2/ARE pathway in PC12 cells. CONCLUSIONS TLR3 activation in MSCs leads to more potent immunosuppressive effects of the CM against pro-inflammatory M1 macrophages, while the use of hCM led to increased neurotrophic effects after H2O2-induced damage to neuronal cells. These results are of interest for the potential treatment of AD with CM from preactivated MSCs.
Collapse
Affiliation(s)
- Tatiana Tolstova
- Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| | | | | | - Alexander Rusanov
- Institute of Biomedical Chemistry, Pogodinskaya 10, 119121 Moscow, Russia
| |
Collapse
|
3
|
El-Menyar A, Asim M, Khan N, Rizoli S, Mahmood I, Al-Ani M, Kanbar A, Alaieb A, Hakim S, Younis B, Taha I, Jogol H, Siddiqui T, Hammo AA, Abdurraheim N, Alabdallat M, Bahey AAA, Ahmed K, Atique S, Chaudry IH, Prabhu KS, Uddin S, Al-Thani H. Systemic and cerebro-cardiac biomarkers following traumatic brain injury: an interim analysis of randomized controlled clinical trial of early administration of beta blockers. Sci Rep 2024; 14:19574. [PMID: 39179700 PMCID: PMC11343837 DOI: 10.1038/s41598-024-70470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
This is an interim analysis of the Beta-blocker (Propranolol) use in traumatic brain injury (TBI) based on the high-sensitive troponin status (BBTBBT) study. The BBTBBT is an ongoing double-blind placebo-controlled randomized clinical trial with a target sample size of 771 patients with TBI. We sought, after attaining 50% of the sample size, to explore the impact of early administration of beta-blockers (BBs) on the adrenergic surge, pro-inflammatory cytokines, and the TBI biomarkers linked to the status of high-sensitivity troponin T (HsTnT). Patients were stratified based on the severity of TBI using the Glasgow coma scale (GCS) and HsTnT status (positive vs negative) before randomization. Patients with positive HsTnT (non-randomized) received propranolol (Group-1; n = 110), and those with negative test were randomized to receive propranolol (Group-2; n = 129) or placebo (Group-3; n = 111). Propranolol was administered within 24 h of injury for 6 days, guided by the heart rate (> 60 bpm), systolic blood pressure (≥ 100 mmHg), or mean arterial pressure (> 70 mmHg). Luminex and ELISA-based immunoassays were used to quantify the serum levels of pro-inflammatory cytokines (Interleukin (IL)-1β, IL-6, IL-8, and IL-18), TBI biomarkers [S100B, Neuron-Specific Enolase (NSE), and epinephrine]. Three hundred and fifty patients with comparable age (mean 34.8 ± 9.9 years) and gender were enrolled in the interim analysis. Group 1 had significantly higher baseline levels of IL-6, IL-1B, S100B, lactate, and base deficit than the randomized groups (p = 0.001). Group 1 showed a significant temporal reduction in serum IL-6, IL-1β, epinephrine, and NSE levels from baseline to 48 h post-injury (p = 0.001). Patients with severe head injuries had higher baseline levels of IL-6, IL-1B, S100B, and HsTnT than mild and moderate TBI (p = 0.01). HsTnT levels significantly correlated with the Injury Severity Score (ISS) (r = 0.275, p = 0.001), GCS (r = - 0.125, p = 0.02), and serum S100B (r = 0.205, p = 0.001). Early Propranolol administration showed a significant reduction in cytokine levels and TBI biomarkers from baseline to 48 h post-injury, particularly among patients with positive HsTnT, indicating the potential role in modulating inflammation post-TBI.Trial registration: ClinicalTrials.gov NCT04508244. It was registered first on 11/08/2020. Recruitment started on 29 December 2020 and is ongoing. The study was partly presented at the 23rd European Congress of Trauma and Emergency Surgery (ECTES), April 28-30, 2024, in Estoril, Lisbon, Portugal.
Collapse
Affiliation(s)
- Ayman El-Menyar
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar.
- Department of Clinical Medicine, Weill Cornell Medicine, P.O. Box 24144, Doha, Qatar.
| | - Mohammad Asim
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Naushad Khan
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Sandro Rizoli
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ismail Mahmood
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Mushreq Al-Ani
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ahad Kanbar
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Abubaker Alaieb
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Suhail Hakim
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Basil Younis
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ibrahim Taha
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Hisham Jogol
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Tariq Siddiqui
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Abdel Aziz Hammo
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Nuri Abdurraheim
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Mohammad Alabdallat
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | | | - Khalid Ahmed
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Sajid Atique
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Irshad H Chaudry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Hassan Al-Thani
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
4
|
Pearson A, Koprivica M, Eisenbaum M, Ortiz C, Browning M, Vincennie T, Tinsley C, Mullan M, Crawford F, Ojo J. PPARγ activation ameliorates cognitive impairment and chronic microglial activation in the aftermath of r-mTBI. J Neuroinflammation 2024; 21:194. [PMID: 39097742 PMCID: PMC11297749 DOI: 10.1186/s12974-024-03173-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/12/2024] [Indexed: 08/05/2024] Open
Abstract
Chronic neuroinflammation and microglial activation are key mediators of the secondary injury cascades and cognitive impairment that follow exposure to repetitive mild traumatic brain injury (r-mTBI). Peroxisome proliferator-activated receptor-γ (PPARγ) is expressed on microglia and brain resident myeloid cell types and their signaling plays a major anti-inflammatory role in modulating microglial responses. At chronic timepoints following injury, constitutive PPARγ signaling is thought to be dysregulated, thus releasing the inhibitory brakes on chronically activated microglia. Increasing evidence suggests that thiazolidinediones (TZDs), a class of compounds approved from the treatment of diabetes mellitus, effectively reduce neuroinflammation and chronic microglial activation by activating the peroxisome proliferator-activated receptor-γ (PPARγ). The present study used a closed-head r-mTBI model to investigate the influence of the TZD Pioglitazone on cognitive function and neuroinflammation in the aftermath of r-mTBI exposure. We revealed that Pioglitazone treatment attenuated spatial learning and memory impairments at 6 months post-injury and reduced the expression of reactive microglia and astrocyte markers in the cortex, hippocampus, and corpus callosum. We then examined whether Pioglitazone treatment altered inflammatory signaling mechanisms in isolated microglia and confirmed downregulation of proinflammatory transcription factors and cytokine levels. To further investigate microglial-specific mechanisms underlying PPARγ-mediated neuroprotection, we generated a novel tamoxifen-inducible microglial-specific PPARγ overexpression mouse line and examined its influence on microglial phenotype following injury. Using RNA sequencing, we revealed that PPARγ overexpression ameliorates microglial activation, promotes the activation of pathways associated with wound healing and tissue repair (such as: IL10, IL4 and NGF pathways), and inhibits the adoption of a disease-associated microglia-like (DAM-like) phenotype. This study provides insight into the role of PPARγ as a critical regulator of the neuroinflammatory cascade that follows r-mTBI in mice and demonstrates that the use of PPARγ agonists such as Pioglitazone and newer generation TZDs hold strong therapeutic potential to prevent the chronic neurodegenerative sequelae of r-mTBI.
Collapse
Affiliation(s)
- Andrew Pearson
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK.
| | - Milica Koprivica
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Max Eisenbaum
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
| | - Camila Ortiz
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
| | | | - Tessa Vincennie
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Cooper Tinsley
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
- James A. Haley Veterans' Hospital, 13000 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Joseph Ojo
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
- James A. Haley Veterans' Hospital, 13000 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| |
Collapse
|
5
|
Mohd Khairi NAA, Hanafi MH, Kassim NK, Ibrahim AH, W Ahmad WMA. The Levels of Biomarkers Interleukin 1 (IL-1) and Brain-Derived Neurotrophic Factor (BDNF) in Non-Invasive Conventional Rehabilitation and Robotic Rehabilitation Among Brain Injury Patients: A Narrative Review. Cureus 2024; 16:e68332. [PMID: 39355076 PMCID: PMC11444801 DOI: 10.7759/cureus.68332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2024] [Indexed: 10/03/2024] Open
Abstract
Acquired brain injury (ABI) is becoming increasingly common in Malaysia as a result of a rise in both strokes and accidents. The present review aims to explore the levels of serum inflammatory markers of interleukin-1 (IL-1) and brain-derived neurotrophic factor (BDNF) following conventional and robotic rehabilitation regimes among ABI patients and the association between serum biomarkers with the Medical Research Council (MRC) scale for muscle strength. Online databases, namely ScienceDirect, PubMed, and Google Scholar were utilized by using search terms such as 'Definition of brain injury', 'Epidemiology of brain injury', 'Interleukin-1 in stroke', 'BDNF in stroke', 'Interleukin-1 in traumatic brain injury', 'BDNF in traumatic brain injury', 'Interleukin-1 level and robotic rehabilitation', 'BDNF and robotic rehabilitation', 'Interleukin-1 level and neurorehabilitation', and 'BDNF and neurorehabilitation'. All types of articles with different levels of evidence were included along with other relevant review articles. Articles that were not in English and were not available in the full text were excluded. The review identifies similar and no significant improvement in the treatment between conventional rehabilitation and robotic rehabilitation concerning serum biomarkers IL-1 and BDNF. This review also identifies that muscle strength and endurance training improved the level of serum BDNF in brain injury patients. Therefore, this review provides evidence of the levels of IL-1 and BDNF in non-invasive conventional rehabilitation and robotic rehabilitation among brain injury patients, as well as their relation with the MRC scale, to give a good functional outcome that will enhance the quality of life of these groups of individuals.
Collapse
Affiliation(s)
| | - Muhammad Hafiz Hanafi
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | | | - Al Hafiz Ibrahim
- School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, MYS
| | | |
Collapse
|
6
|
Banderwal R, Kadian M, Garg S, Kumar A. 'Comprehensive review of emerging drug targets in traumatic brain injury (TBI): challenges and future scope. Inflammopharmacology 2024:10.1007/s10787-024-01524-w. [PMID: 39023681 DOI: 10.1007/s10787-024-01524-w] [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: 08/03/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Traumatic brain injury (TBI) is a complex brain problem that causes significant morbidity and mortality among people of all age groups. The complex pathophysiology, varied symptoms, and inadequate treatment further precipitate the problem. Further, TBI produces several psychiatric problems and other related complications in post-TBI survival patients, which are often treated symptomatically or inadequately. Several approaches, including neuroprotective agents targeting several pathways of oxidative stress, neuroinflammation, cytokines, immune system GABA, glutamatergic, microglia, and astrocytes, are being tried by researchers to develop effective treatments or magic bullets to manage the condition effectively. The problem of TBI is therefore treated as a challenge among pharmaceutical scientists or researchers to develop drugs for the effective management of this problem. The goal of the present comprehensive review is to provide an overview of the several pharmacological targets, processes, and cellular pathways that researchers are focusing on, along with an update on their current state.
Collapse
Affiliation(s)
- Rittu Banderwal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Sukant Garg
- Department of General Pathology, Dr HS Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, 160014, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC- Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
7
|
Guida F, Iannotta M, Lauritano A, Infantino R, Salviati E, Verde R, Luongo L, Sommella EM, Iannotti FA, Campiglia P, Maione S, Di Marzo V, Piscitelli F. Early biomarkers in the presymptomatic phase of cognitive impairment: changes in the endocannabinoidome and serotonergic pathways in Alzheimer's-prone mice after mTBI. Acta Neuropathol Commun 2024; 12:113. [PMID: 38992700 PMCID: PMC11241935 DOI: 10.1186/s40478-024-01820-0] [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: 03/13/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Despite extensive studies on the neurobiological correlates of traumatic brain injury (TBI), little is known about its molecular determinants on long-term consequences, such as dementia and Alzheimer's disease (AD). METHODS Here, we carried out behavioural studies and an extensive biomolecular analysis, including inflammatory cytokines, gene expression and the combination of LC-HRMS and MALDI-MS Imaging to elucidate the targeted metabolomics and lipidomics spatiotemporal alterations of brains from wild-type and APP-SWE mice, a genetic model of AD, at the presymptomatic stage, subjected to mild TBI. RESULTS We found that brain injury does not affect cognitive performance in APP-SWE mice. However, we detected an increase of key hallmarks of AD, including Aβ1-42 levels and BACE1 expression, in the cortices of traumatized transgenic mice. Moreover, significant changes in the expanded endocannabinoid (eCB) system, or endocannabinoidome (eCBome), occurred, including increased levels of the endocannabinoid 2-AG in APP-SWE mice in both the cortex and hippocampus, and N-acylserotonins, detected for the first time in the brain. The gene expression of enzymes for the biosynthesis and inactivation of eCBs and eCB-like mediators, and some of their main molecular targets, also underwent significant changes. We also identified the formation of heteromers between cannabinoid 1 (CB1) and serotonergic 2A (5HT2A) receptors, whose levels increased in the cortex of APP-SWE mTBI mice, possibly contributing to the exacerbated pathophysiology of AD induced by the trauma. CONCLUSIONS Mild TBI induces biochemical changes in AD genetically predisposed mice and the eCBome may play a role in the pathogenetic link between brain injury and neurodegenerative disorders also by interacting with the serotonergic system.
Collapse
Affiliation(s)
- Francesca Guida
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Monica Iannotta
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Anna Lauritano
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy
| | - Rosmara Infantino
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Emanuela Salviati
- Dipartimento di Farmacia, Università Degli Studi di Salerno, Fisciano, (SA), Italy
| | - Roberta Verde
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy
| | - Livio Luongo
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | | | - Fabio Arturo Iannotti
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy
| | - Pietro Campiglia
- Dipartimento di Farmacia, Università Degli Studi di Salerno, Fisciano, (SA), Italy
| | - Sabatino Maione
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy.
- Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut sur la Nutrition et les Aliments Fonctionnels, Centre NUTRISS, Université Laval, Quebec City, Canada.
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy.
| |
Collapse
|
8
|
Ergul Erkec O, Acikgoz E, Huyut Z, Akyol ME, Ozyurt EO, Keskin S. Ghrelin ameliorates neuronal damage, oxidative stress, inflammatory parameters, and GFAP expression in traumatic brain injury. Brain Inj 2024; 38:514-523. [PMID: 38433464 DOI: 10.1080/02699052.2024.2324012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE This study investigated the effects of ghrelin on oxidative stress, working memory, inflammatory parameters, and neuron degeneration. METHODS TBI was produced with the weight-drop technique. Rats in the G+TBI and TBI+G groups received ghrelin for 7 or 2 days, respectively. The control group received saline. On the 8th day of the study, the brain and blood tissue were taken under anesthesia. RESULTS A significant increase in brain GSH-PX, MDA, IL-1β, TGF-β1, and IL-8 levels and a significant decrease in CAT levels were found in the TBI group compared to the control. Serum MDA, GSH, IL-1β, and IL-8 levels were increased with TBI. Ghrelin treatment after TBI significantly increased the serum GSH, CAT, GSH-PX, and brain GSH and CAT levels, while it significantly decreased the serum MDA, IL-1β, and brain MDA, TGF-β1, and IL-8 levels. Histological evaluations revealed that ghrelin treatment led to a reduction in inflammation, while also significantly ameliorating TBI-induced neuron damage and vascular injuries. Immunohistochemistry staining showed that GFAP staining intensity was significantly increased in the cortex and hippocampus in TBI, and GFAP immunoreactivity was decreased with ghrelin treatment. CONCLUSION The results from this study suggested that ghrelin may have curative effects on TBI.
Collapse
Affiliation(s)
- Ozlem Ergul Erkec
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Mehmet Edip Akyol
- Department of Neurosurgery, Faculty of Mecine, Van Yuzuncu Yil University, Van, Turkey
| | | | - Sıddık Keskin
- Department of Biostatistics, Van Yuzuncu Yıl University, Van, Turkey
| |
Collapse
|
9
|
Özen I, Clausen F, Flygt J, Marklund N, Paul G. Neutralization of Interleukin 1-beta is associated with preservation of thalamic capillaries after experimental traumatic brain injury. Front Neurol 2024; 15:1378203. [PMID: 38765267 PMCID: PMC11100426 DOI: 10.3389/fneur.2024.1378203] [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: 01/29/2024] [Accepted: 04/11/2024] [Indexed: 05/21/2024] Open
Abstract
Introduction Traumatic brain injury to thalamo-cortical pathways is associated with posttraumatic morbidity. Diffuse mechanical forces to white matter tracts and deep grey matter regions induce an inflammatory response and vascular damage resulting in progressive neurodegeneration. Pro-inflammatory cytokines, including interleukin-1β (IL-1β), may contribute to the link between inflammation and the injured capillary network after TBI. This study investigates whether IL-1β is a key contributor to capillary alterations and changes in pericyte coverage in the thalamus and cortex after TBI. Methods Animals were subjected to central fluid percussion injury (cFPI), a model of TBI causing widespread axonal and vascular pathology, or sham injury and randomized to receive a neutralizing anti-IL-1β or a control, anti-cyclosporin A antibody, at 30 min post-injury. Capillary length and pericyte coverage of cortex and thalamus were analyzed by immunohistochemistry at 2- and 7-days post-injury. Results and Conclusion Our results show that early post-injury attenuation of IL-1β dependent inflammatory signaling prevents capillary damage by increasing pericyte coverage in the thalamus.
Collapse
Affiliation(s)
- Ilknur Özen
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Fredrik Clausen
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Johanna Flygt
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Niklas Marklund
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden
| |
Collapse
|
10
|
Fritsch LE, Kelly C, Leonard J, de Jager C, Wei X, Brindley S, Harris EA, Kaloss AM, DeFoor N, Paul S, O'Malley H, Ju J, Olsen ML, Theus MH, Pickrell AM. STING-Dependent Signaling in Microglia or Peripheral Immune Cells Orchestrates the Early Inflammatory Response and Influences Brain Injury Outcome. J Neurosci 2024; 44:e0191232024. [PMID: 38360749 PMCID: PMC10957216 DOI: 10.1523/jneurosci.0191-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 12/16/2023] [Accepted: 01/02/2024] [Indexed: 02/17/2024] Open
Abstract
While originally identified as an antiviral pathway, recent work has implicated that cyclic GMP-AMP-synthase-Stimulator of Interferon Genes (cGAS-STING) signaling is playing a critical role in the neuroinflammatory response to traumatic brain injury (TBI). STING activation results in a robust inflammatory response characterized by the production of inflammatory cytokines called interferons, as well as hundreds of interferon stimulated genes (ISGs). Global knock-out (KO) mice inhibiting this pathway display neuroprotection with evidence that this pathway is active days after injury; yet, the early neuroinflammatory events stimulated by STING signaling remain understudied. Furthermore, the source of STING signaling during brain injury is unknown. Using a murine controlled cortical impact (CCI) model of TBI, we investigated the peripheral immune and microglial response to injury utilizing male chimeric and conditional STING KO animals, respectively. We demonstrate that peripheral and microglial STING signaling contribute to negative outcomes in cortical lesion volume, cell death, and functional outcomes postinjury. A reduction in overall peripheral immune cell and neutrophil infiltration at the injury site is STING dependent in these models at 24 h. Transcriptomic analysis at 2 h, when STING is active, reveals that microglia drive an early, distinct transcriptional program to elicit proinflammatory genes including interleukin 1-β (IL-1β), which is lost in conditional knock-out mice. The upregulation of alternative innate immune pathways also occurs after injury in these animals, which supports a complex relationship between brain-resident and peripheral immune cells to coordinate the proinflammatory response and immune cell influx to damaged tissue after injury.
Collapse
Affiliation(s)
- Lauren E Fritsch
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Roanoke, Virginia 24016
| | - Colin Kelly
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Roanoke, Virginia 24016
| | - John Leonard
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Caroline de Jager
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Roanoke, Virginia 24016
| | - Xiaoran Wei
- Biomedical and Veterinary Sciences Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Samantha Brindley
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Elizabeth A Harris
- Biomedical and Veterinary Sciences Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Alexandra M Kaloss
- Biomedical and Veterinary Sciences Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Nicole DeFoor
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Swagatika Paul
- Biomedical and Veterinary Sciences Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Hannah O'Malley
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Jing Ju
- Biomedical and Veterinary Sciences Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Michelle L Olsen
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Alicia M Pickrell
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| |
Collapse
|
11
|
Ma X, Jiao J, Aierken M, Sun H, Chen L. Hypoxia Inducible Factor-1α Through ROS/NLRP3 Pathway Regulates the Mechanism of Acute Ischemic Stroke Microglia Scorching Mechanism. Biologics 2023; 17:167-180. [PMID: 38145108 PMCID: PMC10748736 DOI: 10.2147/btt.s444714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
Purpose In vitro experiments explored how the hypoxia-induced factor-1α (HIF-1α) regulates the regulation of pyroptosis in microglial cells (BV 2) in acute ischemic stroke through ROS/NLRP3 pathway. Methods The microglia acute phase oxygen-glucose deprivation/reoxygenation (OGD/R) was established, CCK-8 was applied to determine the optimal timing of intervention modeling. HIF-1α was overexpressed with stabilizer GF-4592 and HIF-1α small molecule interfering RNA (HIF-1α-siRNA), which was divided into group A (blank group), group B (OGD/R model group), group C (model+FG-4592 intervention group), group D (model+siRNA negative control group) and group E (model+HIF-1α-siRNA group). Cell proliferation of different groups was measured by CCK-8 assay. Pyroptosis and intracellular ROS levels were measured by flow cell technology. IL-18, IL-1β levels were measured by ELISA. HIF-1α, GSDMD-D, GSDMD-N, clean-Caspase-1 and NLRP3 protein expression levels were measured by Western blot. On the above experiments, ROS and NLRP3 response experiments were performed to explore how HIF-1α regulates pyroptosis through ROS/NLRP3 pathway. Results Hypoxia for 6 h then reoxygenation for 12 h was the optimal intervention time. Compared with groups B and D, cell proliferation in group C was significantly enhanced, pyroptosis, intracellular levels of ROS, IL-18, IL-1β and the expression of GSDMD-D, GSDMD-N, clean-Caspase-1, and NLRP3 proteins were significantly decreased in group C (P < 0.05). However, in group E, the performance of these test indicators were exactly the opposite, and the difference was statistically significant (P < 0.05). Through ROS and NLRP3 response experiments, it was found that HIF-1α Inhibition of Pyroptosis by inhibiting ROS/NLRP3 pathway. Conclusion Overexpression of HIF-1α factor can inhibit microglia pyroptosis. HIF-1α factor has an inhibitory effect on the ROS/NLRP 3 pathway, which can inhibit the pyroptotic process in microglia.
Collapse
Affiliation(s)
- Xin Ma
- Department of Clinical Laboratory, Urumqi Friendship Hospital, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Junxia Jiao
- Department of Clinical Laboratory, Urumqi Friendship Hospital, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Mayila Aierken
- Department of Clinical Laboratory, Urumqi Friendship Hospital, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Hong Sun
- Department of Clinical Laboratory, Urumqi Friendship Hospital, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Li Chen
- Department of Clinical Laboratory, Urumqi Friendship Hospital, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| |
Collapse
|
12
|
Wang JX, Xiao X, He XC, He BD, Liu CM, Teng ZQ. Agomir-331 Suppresses Reactive Gliosis and Neuroinflammation after Traumatic Brain Injury. Cells 2023; 12:2429. [PMID: 37887272 PMCID: PMC10605079 DOI: 10.3390/cells12202429] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/19/2023] [Accepted: 09/23/2023] [Indexed: 10/28/2023] Open
Abstract
Traumatic brain injury usually triggers glial scar formation, neuroinflammation, and neurodegeneration. However, the molecular mechanisms underlying these pathological features are largely unknown. Using a mouse model of hippocampal stab injury (HSI), we observed that miR-331, a brain-enriched microRNA, was significantly downregulated in the early stage (0-7 days) of HSI. Intranasal administration of agomir-331, an upgraded product of miR-331 mimics, suppressed reactive gliosis and neuronal apoptosis and improved cognitive function in HSI mice. Finally, we identified IL-1β as a direct downstream target of miR-331, and agomir-331 treatment significantly reduced IL-1β levels in the hippocampus after acute injury. Our findings highlight, for the first time, agomir-331 as a pivotal neuroprotective agent for early rehabilitation of HSI.
Collapse
Affiliation(s)
- Jin-Xing Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China (X.-C.H.)
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100408, China
| | - Xiao Xiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China (X.-C.H.)
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100408, China
| | - Xuan-Cheng He
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China (X.-C.H.)
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Bao-Dong He
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China (X.-C.H.)
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100408, China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China (X.-C.H.)
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100408, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China (X.-C.H.)
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100408, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| |
Collapse
|
13
|
Lindblad C, Rostami E, Helmy A. Interleukin-1 Receptor Antagonist as Therapy for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1508-1528. [PMID: 37610701 PMCID: PMC10684479 DOI: 10.1007/s13311-023-01421-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury is a common type of acquired brain injury of varying severity carrying potentially deleterious consequences for the afflicted individuals, families, and society. Following the initial, traumatically induced insult, cellular injury processes ensue. These are believed to be amenable to treatment. Among such injuries, neuroinflammation has gained interest and has become a specific focus for both experimental and clinical researchers. Neuroinflammation is elicited almost immediately following trauma, and extend for a long time, possibly for years, after the primary injury. In the acute phase, the inflammatory response is characterized by innate mechanisms such as the activation of microglia which among else mediates cytokine production. Among the earliest cytokines to emerge are the interleukin- (IL-) 1 family members, comprising, for example, the agonist IL-1β and its competitive antagonist, IL-1 receptor antagonist (IL-1ra). Because of its early emergence following trauma and its increased concentrations also after human TBI, IL-1 has been hypothesized to be a tractable treatment target following TBI. Ample experimental data supports this, and demonstrates restored neurological behavior, diminished lesion zones, and an attenuated inflammatory response following IL-1 modulation either through IL-1 knock-out experiments, IL-1β inhibition, or IL-1ra treatment. Of these, IL-1ra treatment is likely the most physiological. In addition, recombinant human IL-1ra (anakinra) is already approved for utilization across a few rheumatologic disorders. As of today, one randomized clinical controlled trial has utilized IL-1ra inhibition as an intervention and demonstrated its safety. Further clinical trials powered for patient outcome are needed in order to demonstrate efficacy. In this review, we summarize IL-1 biology in relation to acute neuroinflammatory processes following TBI with a particular focus on current evidence for IL-1ra treatment both in the experimental and clinical context.
Collapse
Affiliation(s)
- Caroline Lindblad
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Elham Rostami
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, entrance 85 floor 2, Akademiska Sjukhuset, 751 85, Uppsala, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Ozkizilcik A, Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Wiklund L, Sharma HS. Nanowired delivery of antibodies to tau and neuronal nitric oxide synthase together with cerebrolysin attenuates traumatic brain injury induced exacerbation of brain pathology in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 171:83-121. [PMID: 37783564 DOI: 10.1016/bs.irn.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Concussive head injury (CHI) is one of the major risk factors for developing Parkinson's disease in later life of military personnel affecting lifetime functional and cognitive disturbances. Till date no suitable therapies are available to attenuate CHI or PD induced brain pathology. Thus, further exploration of novel therapeutic agents are highly warranted using nanomedicine in enhancing the quality of life of veterans or service members of US military. Since PD or CHI induces oxidative stress and perturbs neurotrophic factors regulation associated with phosphorylated tau (p-tau) deposition, a possibility exists that nanodelivery of agents that could enhance neurotrophic factors balance and attenuate oxidative stress could be neuroprotective in nature. In this review, nanowired delivery of cerebrolysin-a balanced composition of several neurotrophic factors and active peptide fragments together with monoclonal antibodies to neuronal nitric oxide synthase (nNOS) with p-tau antibodies was examined in PD following CHI in model experiments. Our results suggest that combined administration of nanowired antibodies to nNOS and p-tau together with cerebrolysin significantly attenuated CHI induced exacerbation of PD brain pathology. This combined treatment also has beneficial effects in CHI or PD alone, not reported earlier.
Collapse
Affiliation(s)
- Asya Ozkizilcik
- Dept. Biomedical Engineering, University of Arkansas, Fayetteville, AR, United Staes
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; ''RoNeuro'' Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston MA, United States
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
15
|
Timaru-Kast R, Coronel-Castello SP, Krämer TJ, Hugonnet AV, Schäfer MKE, Sebastiani A, Thal SC. AT 1 inhibition mediated neuroprotection after experimental traumatic brain injury is dependent on neutrophils in male mice. Sci Rep 2023; 13:7413. [PMID: 37150755 PMCID: PMC10164737 DOI: 10.1038/s41598-023-33797-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/19/2023] [Indexed: 05/09/2023] Open
Abstract
After traumatic brain injury (TBI) cerebral inflammation with invasion of neutrophils and lymphocytes is a crucial factor in the process of secondary brain damage. In TBI the intrinsic renin-angiotensin system is an important mediator of cerebral inflammation, as inhibition of the angiotensin II receptor type 1 (AT1) reduces secondary brain damage and the invasion of neutrophil granulocytes into injured cerebral tissue. The current study explored the involvement of immune cells in neuroprotection mediated by AT1 inhibition following experimental TBI. Four different cohorts of male mice were examined, investigating the effects of neutropenia (anti-Ly6G antibody mediated neutrophil depletion; C57BL/6), lymphopenia (RAG1 deficiency, RAG1-/-), and their combination with candesartan-mediated AT1 inhibition. The present results showed that reduction of neutrophils and lymphocytes, as well as AT1 inhibition in wild type and RAG1-/- mice, reduced brain damage and neuroinflammation after TBI. However, in neutropenic mice, candesartan did not have an effect. Interestingly, AT1 inhibition was found to be neuroprotective in RAG1-/- mice but not in neutropenic mice. The findings suggest that AT1 inhibition may exert neuroprotection by reducing the inflammation caused by neutrophils, ultimately leading to a decrease in their invasion into cerebral tissue.
Collapse
Affiliation(s)
- Ralph Timaru-Kast
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Shila P Coronel-Castello
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Tobias J Krämer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Faculty of Health, University of Witten/Herdecke, Alfred-Herrhausen-Strasse 50, 58455, Witten, Germany
| | - André V Hugonnet
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Anne Sebastiani
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Department of Anesthesiology, HELIOS University Hospital Wuppertal, University of Witten/Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany
| | - Serge C Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
- Department of Anesthesiology, HELIOS University Hospital Wuppertal, University of Witten/Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany
| |
Collapse
|
16
|
Li QY, Duan YW, Zhou YH, Chen SX, Li YY, Zang Y. NLRP3-Mediated Piezo1 Upregulation in ACC Inhibitory Parvalbumin-Expressing Interneurons Is Involved in Pain Processing after Peripheral Nerve Injury. Int J Mol Sci 2022; 23:13035. [PMID: 36361825 PMCID: PMC9655876 DOI: 10.3390/ijms232113035] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 10/29/2023] Open
Abstract
The anterior cingulate cortex (ACC) is particularly critical for pain information processing. Peripheral nerve injury triggers neuronal hyper-excitability in the ACC and mediates descending facilitation to the spinal dorsal horn. The mechanically gated ion channel Piezo1 is involved in the transmission of pain information in the peripheral nervous system. However, the pain-processing role of Piezo1 in the brain is unknown. In this work, we found that spared (sciatic) nerve injury (SNI) increased Piezo1 protein levels in inhibitory parvalbumin (PV)-expressing interneurons (PV-INs) but not in glutaminergic CaMKⅡ+ neurons, in the bilateral ACC. A reduction in the number of PV-INs but not in the number of CaMKⅡ+ neurons and a significant reduction in inhibitory synaptic terminals was observed in the SNI chronic pain model. Further, observation of morphological changes in the microglia in the ACC showed their activated amoeba-like transformation, with a reduction in process length and an increase in cell body area. Combined with the encapsulation of Piezo1-positive neurons by Iba1+ microglia, the loss of PV-INs after SNI might result from phagocytosis by the microglia. In cellular experiments, administration of recombinant rat TNF-α (rrTNF) to the BV2 cell culture or ACC neuron primary culture elevated the protein levels of Piezo1 and NOD-like receptor (NLR) family pyrin domain containing 3 (NLRP3). The administration of the NLRP3 inhibitor MCC950 in these cells blocked the rrTNF-induced expression of caspase-1 and interleukin-1β (key downstream factors of the activated NLRP3 inflammasome) in vitro and reversed the SNI-induced Piezo1 overexpression in the ACC and alleviated SNI-induced allodynia in vivo. These results suggest that NLRP3 may be the key factor in causing Piezo1 upregulation in SNI, promoting an imbalance between ACC excitation and inhibition by inducing the microglial phagocytosis of PV-INs and, thereby, facilitating spinal pain transmission.
Collapse
Affiliation(s)
- Qiao-Yun Li
- Pain Research Center and Department of Physiology, Zhongshan Medical School of Sun Yat-sen University, 74 Zhongshan Road. 2, Guangzhou 510080, China
| | - Yi-Wen Duan
- Pain Research Center and Department of Physiology, Zhongshan Medical School of Sun Yat-sen University, 74 Zhongshan Road. 2, Guangzhou 510080, China
| | - Yao-Hui Zhou
- Pain Research Center and Department of Physiology, Zhongshan Medical School of Sun Yat-sen University, 74 Zhongshan Road. 2, Guangzhou 510080, China
| | - Shao-Xia Chen
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Yong-Yong Li
- Pain Research Center and Department of Physiology, Zhongshan Medical School of Sun Yat-sen University, 74 Zhongshan Road. 2, Guangzhou 510080, China
| | - Ying Zang
- Pain Research Center and Department of Physiology, Zhongshan Medical School of Sun Yat-sen University, 74 Zhongshan Road. 2, Guangzhou 510080, China
| |
Collapse
|
17
|
Kim CK, Park JS, Kim E, Oh MK, Lee YT, Yoon KJ, Joo KM, Lee K, Park YS. The effects of early exercise in traumatic brain-injured rats with changes in motor ability, brain tissue, and biomarkers. BMB Rep 2022; 55:512-517. [PMID: 36104258 PMCID: PMC9623238 DOI: 10.5483/bmbrep.2022.55.10.097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/06/2022] [Accepted: 08/17/2022] [Indexed: 09/19/2023] Open
Abstract
Traumatic brain injury (TBI) is brain damage which is caused by the impact of external mechanical forces. TBI can lead to the temporary or permanent impairment of physical and cognitive abilities, resulting in abnormal behavior. We recently observed that a single session of early exercise in animals with TBI improved their behavioral performance in the absence of other cognitive abnormalities. In the present study, we investigated the therapeutic effects of continuous exercise during the early stages of TBI in rats. We found that continuous low-intensity exercise in early-stage improves the locomotion recovery in the TBI of animal models; however, it does not significantly enhance short-term memory capabilities. Moreover, continuous early exercise not only reduces the protein expression of cerebral damage-related markers, such as Glial Fibrillary Acid Protein (GFAP), Neuron-Specific Enolase (NSE), S100β, Protein Gene Products 9.5 (PGP9.5), and Heat Shock Protein 70 (HSP70), but it also decreases the expression of apoptosis-related protein BAX and cleaved caspase 3. Furthermore, exercise training in animals with TBI decreases the microglia activation and the expression of inflammatory cytokines in the serum, such as CCL20, IL-13, IL-1α, and IL-1β. These findings thus demonstrate that early exercise therapy for TBI may be an effective strategy in improving physiological function, and that serum protein levels are useful biomarkers for the predicition of the effectiveness of early exercise therapy.[BMB Reports 2022; 55(10): 506-511].
Collapse
Affiliation(s)
- Chung Kwon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Jee Soo Park
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Eunji Kim
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Min-Kyun Oh
- Department of Rehabilitation Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University Graduate School of Medicine, Jinju 52727, Korea
| | - Yong-Taek Lee
- Department of Physical & Rehabilitation Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
| | - Kyung Jae Yoon
- Department of Physical & Rehabilitation Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
| | - Kyeung Min Joo
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Kyunghoon Lee
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Young Sook Park
- Department of Physical & Rehabilitation Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Korea
| |
Collapse
|
18
|
Jacquens A, Needham EJ, Zanier ER, Degos V, Gressens P, Menon D. Neuro-Inflammation Modulation and Post-Traumatic Brain Injury Lesions: From Bench to Bed-Side. Int J Mol Sci 2022; 23:11193. [PMID: 36232495 PMCID: PMC9570205 DOI: 10.3390/ijms231911193] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Head trauma is the most common cause of disability in young adults. Known as a silent epidemic, it can cause a mosaic of symptoms, whether neurological (sensory-motor deficits), psychiatric (depressive and anxiety symptoms), or somatic (vertigo, tinnitus, phosphenes). Furthermore, cranial trauma (CT) in children presents several particularities in terms of epidemiology, mechanism, and physiopathology-notably linked to the attack of an immature organ. As in adults, head trauma in children can have lifelong repercussions and can cause social and family isolation, difficulties at school, and, later, socio-professional adversity. Improving management of the pre-hospital and rehabilitation course of these patients reduces secondary morbidity and mortality, but often not without long-term disability. One hypothesized contributor to this process is chronic neuroinflammation, which could accompany primary lesions and facilitate their development into tertiary lesions. Neuroinflammation is a complex process involving different actors such as glial cells (astrocytes, microglia, oligodendrocytes), the permeability of the blood-brain barrier, excitotoxicity, production of oxygen derivatives, cytokine release, tissue damage, and neuronal death. Several studies have investigated the effect of various treatments on the neuroinflammatory response in traumatic brain injury in vitro and in animal and human models. The aim of this review is to examine the various anti-inflammatory therapies that have been implemented.
Collapse
Affiliation(s)
- Alice Jacquens
- Unité de Neuroanesthésie-Réanimation, Hôpital de la Pitié Salpêtrière 43-87, Boulevard de l’Hôpital, F-75013 Paris, France
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - Edward J. Needham
- Division of Anaesthesia, Addenbrooke’s Hospital, University of Cambridge, Box 93, Hills Road, Cambridge CB2 2QQ, UK
| | - Elisa R. Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Vincent Degos
- Unité de Neuroanesthésie-Réanimation, Hôpital de la Pitié Salpêtrière 43-87, Boulevard de l’Hôpital, F-75013 Paris, France
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - Pierre Gressens
- Inserm, Maladies Neurodéveloppementales et Neurovasculaires, Université Paris Cité, F-75019 Paris, France
| | - David Menon
- Division of Anaesthesia, Addenbrooke’s Hospital, University of Cambridge, Box 93, Hills Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
19
|
Purkinje cell vulnerability induced by diffuse traumatic brain injury is linked to disruption of long-range neuronal circuits. Acta Neuropathol Commun 2022; 10:129. [PMID: 36064443 PMCID: PMC9446851 DOI: 10.1186/s40478-022-01435-3] [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: 06/07/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
Cerebellar dysfunction is commonly observed following traumatic brain injury (TBI). While direct impact to the cerebellum by TBI is rare, cerebellar pathology may be caused by indirect injury via cortico-cerebellar pathways. To address the hypothesis that degeneration of Purkinje cells (PCs), which constitute the sole output from the cerebellum, is linked to long-range axonal injury and demyelination, we used the central fluid percussion injury (cFPI) model of widespread traumatic axonal injury in mice. Compared to controls, TBI resulted in early PC loss accompanied by alterations in the size of pinceau synapses and levels of non-phosphorylated neurofilament in PCs. A combination of vDISCO tissue clearing technique and immunohistochemistry for vesicular glutamate transporter type 2 show that diffuse TBI decreased mossy and climbing fiber synapses on PCs. At 2 days post-injury, numerous axonal varicosities were found in the cerebellum supported by fractional anisotropy measurements using 9.4 T MRI. The disruption and demyelination of the cortico-cerebellar circuits was associated with poor performance of brain-injured mice in the beam-walk test. Despite a lack of direct input from the injury site to the cerebellum, these findings argue for novel long-range mechanisms causing Purkinje cell injury that likely contribute to cerebellar dysfunction after TBI.
Collapse
|
20
|
Crosstalk between PI3K/AKT/KLF4 signaling and microglia M1/M2 polarization as a novel mechanistic approach towards flibanserin repositioning in parkinson's disease. Int Immunopharmacol 2022; 112:109191. [PMID: 36055034 DOI: 10.1016/j.intimp.2022.109191] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 02/07/2023]
Abstract
Balancing microglia M1/M2 polarization has been shown as a prospective therapeutic strategy for Parkinson's disease (PD). Various vital signaling pathways are likely to govern the microglial phenotype. The implication of 5HT1A receptors in neurodegenerative disorders has raised interest in exploring the repositioning of flibanserin (Flib), a 5HT1A agonist, as an effective neuroprotective agent for PD. Therefore, this study was designed to assess the ability of Flib to modulate microglia phenotype switching from M1 to M2 via PI3K/AKT downstream targets in a rotenone model of PD. Rats received rotenone (1.5 mg/kg) every other day and were concurrently treated with Flib (40 mg/kg/day) with or without wortmannin (15 μg/kg/day), a PI3K inhibitor, for 21 days. Flib improved the motor perturbations induced by rotenone, as confirmed by the reversion of histopathological damage and tyrosine hydroxylase immunohistochemical alterations in both the striata and substantia nigra. The molecular signaling of Flib was elaborated by inducing striatal AKT phosphorylation and the expression of its substantial target, KLF4. Flib induced STAT6 phosphorylation to promote M2 polarization as demonstrated by the increased CD163++ microglial count with striatal arginase activity. In parallel, it markedly inhibited M1 activation as evidenced by the reduction in CD86++ microglia count with striatal proinflammatory mediators, IL-1β and iNOS. The pre-administration of wortmannin mostly negated Flib's neuroprotective effects. In conclusion, Flib AKT/ KLF4-dependently amended M1/M2 microglial imbalance to exert a promising neuroprotective effect, highlighting its potential as a revolutionary candidate for conquering PD.
Collapse
|
21
|
Doğanyiğit Z, Erbakan K, Akyuz E, Polat AK, Arulsamy A, Shaikh MF. The Role of Neuroinflammatory Mediators in the Pathogenesis of Traumatic Brain Injury: A Narrative Review. ACS Chem Neurosci 2022; 13:1835-1848. [PMID: 35732021 DOI: 10.1021/acschemneuro.2c00196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) is a debilitating acquired neurological disorder that afflicts nearly 74 million people worldwide annually. TBI has been classified as more than just a single insult because of its associated risk toward various long-term neurological and neurodegenerative disorders. This risk may be triggered by a series of postinjury secondary molecular and cellular pathology, which may be dependent on the severity of the TBI. Among the secondary injury mechanisms, neuroinflammation may be the most crucial as it may exacerbate brain damage and lead to fatal consequences when prolonged. This Review aimed to elucidate the influence of neuroinflammatory mediators on the TBI functional and pathological outcomes, particularly focusing on inflammatory cytokines which were associated with neuronal dysfunctions in the acute and chronic stages of TBI. These cytokines include interleukins (IL) such as IL-1(beta)β, IL-4, IL-6, IL8, IL-10, IL-18, IL-33 and tumor necrosis factor alpha (TNF-α), which have been extensively studied. Apart from these, IL-2, interferon gamma (IFN-γ), and transforming growth factor-beta (TGF-β) may also play a significant role in the pathogenesis of TBI. These neuroinflammatory mediators may trigger a series of pathological events such as cell death, microglial suppression, and increased catecholaminergic activity. Interestingly, in the acute phase of TBI, most of these mediators may also play a neuroprotective role by displaying anti-inflammatory properties, which may convert to a pro-inflammatory action in the chronic stages post TBI. Early identification and treatment of these mediators may help the development of more effective treatment options for TBI.
Collapse
Affiliation(s)
- Züleyha Doğanyiğit
- Department of Histology and Embryology, Faculty of Medicine, Yozgat Bozok University, Yozgat 66100, Turkey
| | - Kaan Erbakan
- Ordu University, Faculty of Medicine, Ordu 52200, Turkey
| | - Enes Akyuz
- University of Health Sciences, Hamidiye International Faculty of Medicine, Department of Biophysics, Istanbul 34668, Turkey
| | | | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| |
Collapse
|
22
|
Intervention of neuroinflammation in the traumatic brain injury trajectory: In vivo and clinical approaches. Int Immunopharmacol 2022; 108:108902. [DOI: 10.1016/j.intimp.2022.108902] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/25/2022] [Accepted: 05/24/2022] [Indexed: 12/11/2022]
|
23
|
Tapp ZM, Cornelius S, Oberster A, Kumar JE, Atluri R, Witcher KG, Oliver B, Bray C, Velasquez J, Zhao F, Peng J, Sheridan J, Askwith C, Godbout JP, Kokiko-Cochran ON. Sleep fragmentation engages stress-responsive circuitry, enhances inflammation and compromises hippocampal function following traumatic brain injury. Exp Neurol 2022; 353:114058. [PMID: 35358498 PMCID: PMC9068267 DOI: 10.1016/j.expneurol.2022.114058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/04/2022] [Accepted: 03/24/2022] [Indexed: 02/08/2023]
Abstract
Traumatic brain injury (TBI) impairs the ability to restore homeostasis in response to stress, indicating hypothalamic-pituitary-adrenal (HPA)-axis dysfunction. Many stressors result in sleep disturbances, thus mechanical sleep fragmentation (SF) provides a physiologically relevant approach to study the effects of stress after injury. We hypothesize SF stress engages the dysregulated HPA-axis after TBI to exacerbate post-injury neuroinflammation and compromise recovery. To test this, male and female mice were given moderate lateral fluid percussion TBI or sham-injury and left undisturbed or exposed to daily, transient SF for 7- or 30-days post-injury (DPI). Post-TBI SF increases cortical expression of interferon- and stress-associated genes characterized by inhibition of the upstream regulator NR3C1 that encodes glucocorticoid receptor (GR). Moreover, post-TBI SF increases neuronal activity in the hippocampus, a key intersection of the stress-immune axes. By 30 DPI, TBI SF enhances cortical microgliosis and increases expression of pro-inflammatory glial signaling genes characterized by persistent inhibition of the NR3C1 upstream regulator. Within the hippocampus, post-TBI SF exaggerates microgliosis and decreases CA1 neuronal activity. Downstream of the hippocampus, post-injury SF suppresses neuronal activity in the hypothalamic paraventricular nucleus indicating decreased HPA-axis reactivity. Direct application of GR agonist, dexamethasone, to the CA1 at 30 DPI increases GR activity in TBI animals, but not sham animals, indicating differential GR-mediated hippocampal action. Electrophysiological assessment revealed TBI and SF induces deficits in Schaffer collateral long-term potentiation associated with impaired acquisition of trace fear conditioning, reflecting dorsal hippocampal-dependent cognitive deficits. Together these data demonstrate that post-injury SF engages the dysfunctional post-injury HPA-axis, enhances inflammation, and compromises hippocampal function. Therefore, external stressors that disrupt sleep have an integral role in mediating outcome after brain injury.
Collapse
Affiliation(s)
- Zoe M Tapp
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Sydney Cornelius
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Alexa Oberster
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA
| | - Julia E Kumar
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Ravitej Atluri
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Kristina G Witcher
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Chelsea Bray
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - John Velasquez
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Fangli Zhao
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Juan Peng
- Center for Biostatistics, The Ohio State University, 320-55 Lincoln Tower, 1800 Cannon Drive, Columbus, OH 43210, USA.
| | - John Sheridan
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA; Division of Biosciences, College of Dentistry, The Ohio State University, 305 W. 12(th) Ave, Columbus, OH 43210, USA.
| | - Candice Askwith
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Jonathan P Godbout
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Olga N Kokiko-Cochran
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| |
Collapse
|
24
|
Nicotinamide mononucleotide (NMN) and NMN-rich product supplementation alleviate p-chlorophenylalanine-induced sleep disorders. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
25
|
Role of Inflammation in Traumatic Brain Injury-Associated Risk for Neuropsychiatric Disorders: State of the Evidence and Where Do We Go From Here. Biol Psychiatry 2022; 91:438-448. [PMID: 34955170 DOI: 10.1016/j.biopsych.2021.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/01/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, there has been an increasing awareness that traumatic brain injury (TBI) and concussion substantially increase the risk for developing psychiatric disorders. Even mild TBI increases the risk for depression and anxiety disorders such as posttraumatic stress disorder by two- to threefold, predisposing patients to further functional impairment. This strong epidemiological link supports examination of potential mechanisms driving neuropsychiatric symptom development after TBI. One potential mechanism for increased neuropsychiatric symptoms after TBI is via inflammatory processes, as central nervous system inflammation can last years after initial injury. There is emerging preliminary evidence that TBI patients with posttraumatic stress disorder or depression exhibit increased central and peripheral inflammatory markers compared with TBI patients without these comorbidities. Growing evidence has demonstrated that immune signaling in animals plays an integral role in depressive- and anxiety-like behaviors after severe stress or brain injury. In this review, we will 1) discuss current evidence for chronic inflammation after TBI in the development of neuropsychiatric symptoms, 2) highlight potential microglial activation and cytokine signaling contributions, and 3) discuss potential promise and pitfalls for immune-targeted interventions and biomarker strategies to identify and treat TBI patients with immune-related neuropsychiatric symptoms.
Collapse
|
26
|
Moss LD, Sode D, Patel R, Lui A, Hudson C, Patel NA, Bickford PC. Intranasal delivery of exosomes from human adipose derived stem cells at forty-eight hours post injury reduces motor and cognitive impairments following traumatic brain injury. Neurochem Int 2021; 150:105173. [PMID: 34453976 PMCID: PMC8511339 DOI: 10.1016/j.neuint.2021.105173] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
The neuroprotective role of human adipose-derived stems cells (hASCs) has raised great interest in regenerative medicine due to their ability to modulate their surrounding environment. Our group has demonstrated that exosomes derived from hASC (hASCexo) are a cell-free regenerative approach to long term recovery following traumatic brain injury (TBI). Previously, we demonstrated the efficacy of exosome treatment with intravenous delivery at 3 h post TBI in rats. Here, we show efficacy of exosomes through intranasal delivery at 48 h post TBI in mice lengthening the therapeutic window of treatment and therefore increasing possible translation to clinical studies. Our findings demonstrate significant recovery of motor impairment assessed by an elevated body swing test in mice treated with exosomes containing MALAT1 compared to both TBI mice without exosomes and exosomes depleted of MALAT1. Significant cognitive improvement was seen in the reversal trial of 8 arm radial arm water maze in mice treated with exosomes containing MALAT1. Furthermore, cortical damage was significantly reduced in mice treated with exosomes containing MALAT1 as well as decreased MHCII+ staining of microglial cells. Mice without exosomes or treated with exosomes depleted of MALAT1 did not show similar recovery. Results demonstrate both inflammation related genes and NRTK3 (TrkC) are target genes modulated by hASC exosomes and further that MALAT1 in hASC exosomes regulates expression of full length TrkC thereby activating the MAPK pathway and promoting recovery. Exosomes are a promising therapeutic approach following TBI with a therapeutic window of at least 48 h and contain long noncoding RNA's, specifically MALAT1 that play a vital role in the mechanism of action.
Collapse
Affiliation(s)
- Lauren D Moss
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Derek Sode
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Rekha Patel
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Ashley Lui
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Charles Hudson
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Niketa A Patel
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA; Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Paula C Bickford
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA; James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA.
| |
Collapse
|
27
|
Sjöström EO, Culot M, Leickt L, Åstrand M, Nordling E, Gosselet F, Kaiser C. Transport study of interleukin-1 inhibitors using a human in vitro model of the blood-brain barrier. Brain Behav Immun Health 2021; 16:100307. [PMID: 34589799 PMCID: PMC8474601 DOI: 10.1016/j.bbih.2021.100307] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/24/2021] [Indexed: 01/15/2023] Open
Abstract
The proinflammatory cytokine Interleukin-1 (IL-1), with its two isoforms α and β, has important roles in multiple pathogenic processes in the central nervous system. The present study aimed to evaluate and compare the blood-to-brain distribution of anakinra (IL-1 receptor antagonist), bermekimab (IL-1α antagonist) and canakinumab (IL-1β antagonist). A human in vitro model of the blood-brain barrier derived from human umbilical cord blood stem cells was used, where isolated CD34+ cells co-cultured with bovine pericytes were matured into polarized brain-like endothelial cells. Transport rates of the three test items were evaluated after 180 min incubation at concentrations 50, 250 and 1250 nM in a transwell system. We report herein that anakinra passes the human brain-like endothelial monolayer at a 4-7-fold higher rate than the monoclonal antibodies tested. Both antibodies had similar transport rates at all concentrations. No dose-dependent effects in transport rates were observed, nor any saturation effects at supraphysiological concentrations. The larger propensity of anakinra to pass this model of the human blood-brain barrier supports existing data and confirms that anakinra can reach the brain compartment at clinically relevant concentrations. As anakinra inhibits the actions of both IL-1α and IL-1β, it blocks all effects of IL-1 downstream signaling. The results herein further add to the growing body of evidence of the potential utility of anakinra to treat neuroinflammatory disorders. Anakinra has a larger propensity to pass the in vitro BBB than monoclonal antibodies targeting the IL-1 system. Implications for targeting inflammation in cerebral ischemia and neurological sequelae of autoinflammatory diseases. Novel and comparative study of biologics in a human in vitro BBB model shows relevance and validity.
Collapse
Affiliation(s)
| | - Maxime Culot
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Lisa Leickt
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Mikael Åstrand
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Erik Nordling
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Fabien Gosselet
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Christina Kaiser
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| |
Collapse
|
28
|
Siebold L, Krueger AC, Abdala JA, Figueroa JD, Bartnik-Olson B, Holshouser B, Wilson CG, Ashwal S. Cosyntropin Attenuates Neuroinflammation in a Mouse Model of Traumatic Brain Injury. Front Mol Neurosci 2020; 13:109. [PMID: 32670020 PMCID: PMC7332854 DOI: 10.3389/fnmol.2020.00109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/22/2020] [Indexed: 12/21/2022] Open
Abstract
Aim: Traumatic brain injury (TBI) is a leading cause of mortality/morbidity and is associated with chronic neuroinflammation. Melanocortin receptor agonists including adrenocorticotropic hormone (ACTH) ameliorate inflammation and provide a novel therapeutic approach. We examined the effect of long-acting cosyntropin (CoSyn), a synthetic ACTH analog, on the early inflammatory response and functional outcome following experimental TBI. Methods: The controlled cortical impact model was used to induce TBI in mice. Mice were assigned to injury and treatment protocols resulting in four experimental groups including sham + saline, sham + CoSyn, TBI + saline, and TBI + CoSyn. Treatment was administered subcutaneously 3 h post-injury and daily injections were given for up to 7 days post-injury. The early inflammatory response was evaluated at 3 days post-injury through the evaluation of cytokine expression (IL1β and TNFα) and immune cell response. Quantification of immune cell response included cell counts of microglia/macrophages (Iba1+ cells) and neutrophils (MPO+ cells) in the cortex and hippocampus. Behavioral testing (n = 10–14 animals/group) included open field (OF) and novel object recognition (NOR) during the first week following injury and Morris water maze (MWM) at 10–15 days post-injury. Results: Immune cell quantification showed decreased accumulation of Iba1+ cells in the perilesional cortex and CA1 region of the hippocampus for CoSyn-treated TBI animals compared to saline-treated. Reduced numbers of MPO+ cells were also found in the perilesional cortex and hippocampus in CoSyn treated TBI mice compared to their saline-treated counterparts. Furthermore, CoSyn treatment reduced IL1β expression in the cortex of TBI mice. Behavioral testing showed a treatment effect of CoSyn for NOR with CoSyn increasing the discrimination ratio in both TBI and Sham groups, indicating increased memory performance. CoSyn also decreased latency to find platform during the early training period of the MWM when comparing CoSyn to saline-treated TBI mice suggesting moderate improvements in spatial memory following CoSyn treatment. Conclusion: Reduced microglia/macrophage accumulation and neutrophil infiltration in conjunction with moderate improvements in spatial learning in our CoSyn treated TBI mice suggests a beneficial anti-inflammatory effect of CoSyn following TBI.
Collapse
Affiliation(s)
- Lorraine Siebold
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Amy C Krueger
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jonathan A Abdala
- The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Johnny D Figueroa
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Brenda Bartnik-Olson
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Barbara Holshouser
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Christopher G Wilson
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States.,Department of Pediatrics, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Stephen Ashwal
- Department of Pediatrics, Loma Linda University Medical Center, Loma Linda, CA, United States
| |
Collapse
|