1
|
Anyachor CP, Orish CN, Ezejiofor AN, Cirovic A, Cirovic A, Dooka BD, Ezealisiji KM, Noundou XS, Orisakwe OE. Silica Nanoparticles from Melon Seed Husk Abrogated Binary Metal(loid) Mediated Cerebellar Dysfunction by Attenuation of Oxido-inflammatory Response and Upregulation of Neurotrophic Factors in Male Albino Rats. CEREBELLUM (LONDON, ENGLAND) 2024:10.1007/s12311-024-01747-1. [PMID: 39331240 DOI: 10.1007/s12311-024-01747-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Silica nanoparticles (SiNPs) have been touted for their role in the management of non-communicable diseases. Their neuroprotective benefits against heavy metal-induced neurotoxicity remain largely unexplored. This is a comparative evaluation of the oxido-inflammatory and neurotrophic effects of Ni, Al, and Ni/Al mixture on the cerebellum of male albino rats with or without treatment with SiNPs generated from melon seed husk. The study complied with the ARRIVE guidelines for reporting in vivo experiments. A total of 91, 7-9 week-old weight-matched male Sprague rats (to avoid sex bias) were randomly divided into 13 different dosing groups where Group 1 served as the control. Other groups received 0.2 mg/kg Ni, 1 mg/kg Al, and 0.2 mg/kg Ni + 1 mg/kg Al mixture with or without different doses of SiNP for 90 days. Rotarod performance was carried out. Oxidative stress markers, Ni, Al, Ca, Fe, Mg, neurotrophic factors, amyloid beta (Aβ-42), cyclooxygenase-2 (COX-2), and acetylcholinesterase (AChE) were determined in the cerebellum. SiNPs from melon seed husk caused a significant decrease in Aβ-42 level and activities of AChE and COX-2 and a significant increase in brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) mediated by Ni, Al, and Ni/Al mixture exposure in rats. Neurotoxicity of the Ni/Al mixture is via heightened neuronal lipoperoxidative damage, decreased Mg, and increased Fe, and co-administration of SiNPs from melon seed husk with the Ni/Al mixture attenuated some of these biochemical changes in the cerebellum.
Collapse
Affiliation(s)
- Chidinma P Anyachor
- African Centre of Excellence for Public Health and Toxicological Research (ACE‑PUTOR), University of Port Harcourt, PMB, Port Harcourt, Choba, 5323, Nigeria
| | - Chinna N Orish
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, PMB, Choba, Port Harcourt, 5323, Nigeria.
| | - Anthonet N Ezejiofor
- African Centre of Excellence for Public Health and Toxicological Research (ACE‑PUTOR), University of Port Harcourt, PMB, Port Harcourt, Choba, 5323, Nigeria
| | - Ana Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, Belgrade, Serbia
| | - Aleksandar Cirovic
- Faculty of Medicine, Institute of Anatomy, University of Belgrade, Belgrade, Serbia
| | - Baridoo Donatus Dooka
- African Centre of Excellence for Public Health and Toxicological Research (ACE‑PUTOR), University of Port Harcourt, PMB, Port Harcourt, Choba, 5323, Nigeria
| | - Kenneth M Ezealisiji
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Port Harcourt, PMB, Choba, Port Harcourt, 5323, Nigeria
| | - Xavier Siwe Noundou
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, MEDUNSA, Box 218, 0204, Pretoria, South Africa
| | - Orish E Orisakwe
- African Centre of Excellence for Public Health and Toxicological Research (ACE‑PUTOR), University of Port Harcourt, PMB, Port Harcourt, Choba, 5323, Nigeria.
- Advanced Research Centre, European University of Lefke, Lefke, Mersin, TR-10, Northern Cyprus, Turkey.
| |
Collapse
|
2
|
Navaseelan L, Retinasamy T, Shaikh MF, Arulsamy A. High Mobility Group Box-1 (HMGB1), a Key Mediator of Cognitive Decline in Neurotrauma with a Potential for Targeted Therapy: A Comprehensive Review. FRONT BIOSCI-LANDMRK 2024; 29:322. [PMID: 39344324 DOI: 10.31083/j.fbl2909322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/26/2024] [Accepted: 07/11/2024] [Indexed: 10/01/2024]
Abstract
Neurotrauma plays a significant role in secondary injuries by intensifying the neuroinflammatory response in the brain. High Mobility Group Box-1 (HMGB1) protein is a crucial neuroinflammatory mediator involved in this process. Numerous studies have hypothesized about the underlying pathophysiology of HMGB1 and its role in cognition, but a definitive link has yet to be established. Elevated levels of HMGB1 in the hippocampus and serum have been associated with declines in cognitive performance, particularly in spatial memory and learning. This review also found that inhibiting HMGB1 can improve cognitive deficits following neurotrauma. Interestingly, HMGB1 levels are linked to the modulation of neuroplasticity and may offer neuroprotective effects in the later stages of neurotraumatic events. Consequently, administering HMGB1 during the acute phase may help reduce neuroinflammatory effects that lead to cognitive deficits in the later stages of neurotrauma. However, further research is needed to understand the time-dependent regulation of HMGB1 and the clinical implications of treatments targeting HMGB1 after neurotrauma.
Collapse
Affiliation(s)
- Locshiny Navaseelan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, NSW 2800, Australia
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| |
Collapse
|
3
|
Schmitz F, Durán-Carabali LE, Rieder AS, Silveira JS, Ramires Junior OV, Bobermin LD, Quincozes-Santos A, Alves VS, Coutinho-Silva R, Savio LEB, Coelho DM, Vargas CR, Netto CA, Wyse ATS. Methylphenidate Exposing During Neurodevelopment Alters Amino Acid Profile, Astrocyte Marker and Glutamatergic Excitotoxicity in the Rat Striatum. Neurotox Res 2024; 42:39. [PMID: 39190189 DOI: 10.1007/s12640-024-00718-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024]
Abstract
There is a public health concern about the use of methylphenidate (MPH) since the higher prescription for young individuals and non-clinical purposes is addressed to the limited understanding of its neurochemical and psychiatric consequences. This study aimed to evaluate the impact of early and chronic MPH treatment on the striatum focusing on amino acid profile, glutamatergic excitotoxicity, redox status, neuroinflammation and glial cell responses. Male Wistar rats were treated with MPH (2.0 mg/kg) or saline solution from the 15th to the 44th postnatal day. Biochemical and histological analyses were conducted after the last administration. MPH altered the amino acid profile in the striatum, increasing glutamate and ornithine levels, while decreasing the levels of serine, phenylalanine, and branched-chain amino acids (leucine, valine, and isoleucine). Glutamate uptake and Na+,K+-ATPase activity were decreased in the striatum of MPH-treated rats as well as increased ATP levels, as indicator of glutamatergic excitotoxicity. Moreover, MPH caused lipid peroxidation and nitrative stress, increased TNF alpha expression, and induced high levels of astrocytes, and led to a decrease in BDNF levels. In summary, our results suggest that chronic early-age treatment with MPH induces parallel activation of damage-associated pathways in the striatum and increases its vulnerability during the juvenile period. In addition, data presented here contribute to shedding light on the mechanisms underlying MPH-induced striatal damage and its potential implications for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Felipe Schmitz
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse´S Lab), Department of Biochemistry, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luz Elena Durán-Carabali
- Graduate Program in Biological Sciences: Physiology, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Alessandra Schmitt Rieder
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse´S Lab), Department of Biochemistry, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Josiane S Silveira
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse´S Lab), Department of Biochemistry, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Osmar Vieira Ramires Junior
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse´S Lab), Department of Biochemistry, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Larissa D Bobermin
- Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - André Quincozes-Santos
- Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vinícius S Alves
- Laboratory of Immunophysiology, Biophysics, Institute Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics, Institute Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Eduardo B Savio
- Laboratory of Immunophysiology, Biophysics, Institute Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniella M Coelho
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carmen R Vargas
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse´S Lab), Department of Biochemistry, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
4
|
Harris S, Chinnery HR, Semple BD, Mychasiuk R. Shaking Up Our Approach: The Need for Characterization and Optimization of Pre-clinical Models of Infant Abusive Head Trauma. J Neurotrauma 2024; 41:1853-1870. [PMID: 38497766 DOI: 10.1089/neu.2023.0598] [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] [Indexed: 03/19/2024] Open
Abstract
Traumatic brain injuries (TBIs) are a large societal and individual burden. In the first year of life, the vast majority of these injuries are the result of inflicted abusive events by a trusted caregiver. Abusive head trauma (AHT) in infants, formerly known as shaken baby syndrome, is the leading cause of inflicted mortality and morbidity in this population. In this review we address clinical diagnosis, symptoms, prognosis, and neuropathology of AHT, emphasizing the burden of repetitive AHT. Next, we consider existing animal models of AHT, and we evaluate key features of an ideal model, highlighting important developmental milestones in children most vulnerable to AHT. We draw on insights from other injury models, such as repetitive, mild TBIs (RmTBIs), post-traumatic epilepsy (PTE), hypoxic-ischemic injuries, and maternal neglect, to speculate on key knowledge gaps and underline important new opportunities in pre-clinical AHT research. Finally, potential treatment options to facilitate healthy development in children following an AHT are considered. Together, this review aims to drive the field toward optimized, well-characterized animal models of AHT, which will allow for greater insight into the underlying neuropathological and neurobehavioral consequences of AHT.
Collapse
Affiliation(s)
- Sydney Harris
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Science, University of Melbourne, Parkville, Victoria, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Prahran, Victoria, Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Prahran, Victoria, Australia
| |
Collapse
|
5
|
Donovan M, Frentz M, Lozano AR, Rao S, Rodriguez M, Noble-Haeusslein LJ. The Emerging Landscape of the Cerebellum after a Pediatric Traumatic Brain Injury: From Diaschisis to Sociality. ADVANCES IN NEUROBIOLOGY 2024; 42:165-177. [PMID: 39432042 DOI: 10.1007/978-3-031-69832-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
There is an expanding interest in the cerebellum in the context of focal and diffuse traumatic injuries to the cerebral cortex. In the adult brain, preclinical studies have revealed acute as well as progressive loss of Purkinje cells in the cerebellum coincident with microglial activation. This pathogenesis, remote to the site of the primary injury, is termed "diaschisis." Here we consider traumatic injuries to the developing brain, where the cerebellum likewise undergoes neurodegeneration. As injury is superimposed on a young brain, long-term adverse consequences may reflect diaschisis that is compounded by disruption of brain development.
Collapse
Affiliation(s)
- Michael Donovan
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Morgan Frentz
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Anakaren Romero Lozano
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Shripriya Rao
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | | | - Linda J Noble-Haeusslein
- Departments of Neurology and Psychology, Dell Medical School and the College of Liberal Arts, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
6
|
Leonard J, Ladner L, Harris EA, de Jager C, Theus MH. The Neuroimmune Interface: Age-Related Responses to Traumatic Brain Injury. ADVANCES IN NEUROBIOLOGY 2024; 42:241-262. [PMID: 39432046 DOI: 10.1007/978-3-031-69832-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Traumatic Brain Injury (TBI) is a significant public health issue, with diverse consequences across the lifespan. This comprehensive review explores the complex interplay between age-related responses and the immune system following TBI. TBI exhibits distinct effects in pediatric, adult, and elderly populations, with profound implications for recovery and long-term outcomes. The immune system, as a key player in the post-TBI inflammatory cascade, exerts age-dependent influences on inflammation, neuroinflammation, and tissue repair. We examine the evolving understanding of age-related neuroinflammatory responses, cytokine profiles, and the role of immune cells, such as microglia and T cells, in the context of TBI. Furthermore, we evaluate the therapeutic implications of age-specific immunomodulation strategies toward mitigating TBI-associated neuropathology. This review consolidates the current knowledge on age-related immune responses in TBI, shedding light on potential avenues for tailored therapeutic interventions across the age spectrum. Understanding these nuanced responses is crucial for optimizing patient care and enhancing recovery outcomes in the aftermath of traumatic brain injury.
Collapse
Affiliation(s)
- John Leonard
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Liliana Ladner
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Elizabeth A Harris
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Caroline de Jager
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Michelle H Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
7
|
Ulgen Tekerek N, Dursun O, Asilioglu Yener N, Yildizdas D, Anıl AB, Kendirli T, Koker A, Karalok S, Aksoy A, Kinik Kaya E, Ekinci F, Incecik F, Olgac Dundar N, Durak F, Botan E, Havan M, Sahin S, Duman O, Haspolat S. Posttraumatic epilepsy in critically ill children with traumatic brain injury. Childs Nerv Syst 2023; 39:3207-3214. [PMID: 37480521 DOI: 10.1007/s00381-023-06087-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
PURPOSE The aim of this study was to determine the clinical, laboratory, and radiological factors related with posttraumatic epilepsy (PTE). METHODS The study is a multicenter descriptive cross-sectional cohort study. Children who followed up for TBI in the pediatric intensive care unit between 2014 and 2021 were included. Demographic data and clinical and radiological parameters were recorded from electronic case forms. All patients who were in the 6-month posttraumatic period were evaluated by a neurologist for PTE. RESULTS Four hundred seventy-seven patients were included. The median age at the time of trauma was 66 (IQR 27-122) months, and 298 (62.5%) were male. Two hundred eighty (58.7%) patients had multiple traumas. The mortality rate was 11.7%. The mean duration of hospitalization, pediatric intensive care unit hospitalization and mechanical ventilation, Rotterdam score, PRISM III score, and GCS at admission were higher in patients with epilepsy (p < 0.05). The rate of epilepsy was higher in patients with severe TBI, cerebral edema on tomography and clinical findings of increased intracranial pressure, blood transfusion in the intensive care unit, multiple intracranial hemorrhages, and intubated patients (p < 0.05). In logistic regression analysis, the presence of intracranial hemorrhage in more than one compartment of the brain (OR 6.13, 95%CI 3.05-12.33) and the presence of seizures (OR 9.75, 95%CI 4.80-19.83) were independently significant in terms of the development of epilepsy (p < 0.001). CONCLUSIONS In this multicenter cross-sectional study, intracranial hemorrhages in more than one compartment and clinical seizures during intensive care unit admission were found to be independent risk factors for PTE development in pediatric intensive care unit patients with TBI.
Collapse
Affiliation(s)
- Nazan Ulgen Tekerek
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Akdeniz University, Antalya, Turkey.
- Akdeniz University Hospital, Dumlupınar Boulevard, Konyaalti, Antalya, 07059, Turkey.
| | - Oguz Dursun
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Nazik Asilioglu Yener
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Dincer Yildizdas
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ayşe Berna Anıl
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Katip Celebi University, İzmir, Turkey
| | - Tanil Kendirli
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Alper Koker
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Selen Karalok
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ayşe Aksoy
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Elif Kinik Kaya
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Faruk Ekinci
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Faruk Incecik
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Nihal Olgac Dundar
- Division of Pediatric Neurology, Department of Pediatrics, Katip Celebi Faculty of Medicine, İzmir, Turkey
| | - Fatih Durak
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Katip Celebi University, İzmir, Turkey
| | - Edin Botan
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Merve Havan
- Division of Pediatric Intensive Care, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Suleyman Sahin
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Ozgur Duman
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Senay Haspolat
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
8
|
Patient-Centered Approaches to Cognitive Assessment in Acute TBI. Curr Neurol Neurosci Rep 2023; 23:59-66. [PMID: 36705882 DOI: 10.1007/s11910-023-01253-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 01/28/2023]
Abstract
PURPOSE OF THE REVIEW The purpose of this article is to help clinicians understand how underlying pathophysiologies and medical comorbidities associated with acute traumatic brain injury (TBI) can impact assessment of cognition during the initial stages of recovery. Clinicians can use information from this article to develop assessment plans rooted in patient-centered care. RECENT FINDINGS The authors conducted a review of the literature related to the assessment of cognition in acute TBI, focusing on pathophysiology, medical comorbidities, and assessment approaches. Results indicated that TBI pathophysiologies associated with white and gray matter changes make many patients vulnerable to cognitive deficits. Acute comorbidities such as psychological and pain status influence cognitive abilities as well. The current approaches to cognitive assessment can be limited in many ways, though by using the patient's neuropathological profile, noted comorbidities, and other patient specific factors, clinicians can potentially improve the effectiveness of assessment.
Collapse
|
9
|
Mariajoseph FP, Chen Z, Sekhar P, Rewell SS, O'Brien TJ, Antonic‐Baker A, Semple BD. Incidence and risk factors of posttraumatic epilepsy following pediatric traumatic brain injury: A systematic review and meta-analysis. Epilepsia 2022; 63:2802-2812. [PMID: 35996866 PMCID: PMC9826023 DOI: 10.1111/epi.17398] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/31/2022] [Accepted: 08/18/2022] [Indexed: 01/11/2023]
Abstract
Posttraumatic epilepsy (PTE) is a well-known chronic complication following traumatic brain injury (TBI). Despite some evidence that age at the time of injury may influence the likelihood of PTE, the incidence of PTE in pediatric populations remains unclear. We therefore conducted a systematic review to determine the overall reported incidence of PTE, and explore potential risk factors associated with PTE after pediatric TBI. A comprehensive literature search of the PubMed, Embase, and Web of Science databases was conducted, including randomized controlled trials and cohort studies assessing the incidence of PTE in TBI pediatric patients. We excluded studies with a sample size of <10 patients and those in which a pediatric cohort was not clearly discernable. The review was conducted in accordance with the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. We found that the overall incidence of PTE following pediatric TBI was 10% (95% confidence interval [CI] = 5.9%-15%). Subgroup analysis of a small number of studies demonstrated that the occurrence of early seizures (cumulative incidence ratio [CIR] = 7.28, 95% CI = 1.09-48.4, p = .040), severe TBI (CIR = 1.81, 95% CI = 1.23-2.67, p < .001), and intracranial hemorrhage (CIR = 1.60, 95% CI = 1.06-2.40, p = .024) increased the risk of PTE in this population. Other factors, including male sex and neurosurgical intervention, were nonsignificantly associated with a higher incidence of PTE. In conclusion, PTE is a significant chronic complication following childhood TBI, similar to in the adult population. Further standardized investigation into clinical risk factors and management guidelines is warranted. PROSPERO ID# CRD42021245802.
Collapse
Affiliation(s)
| | - Zhibin Chen
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
| | - Praba Sekhar
- School of Clinical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Sarah S. Rewell
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia,Department of NeurologyAlfred HealthPrahranVictoriaAustralia
| | - Terence J. O'Brien
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia,Department of NeurologyAlfred HealthPrahranVictoriaAustralia,Department of Medicine (Royal Melbourne Hospital)University of MelbourneParkvilleVictoriaAustralia
| | - Ana Antonic‐Baker
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
| | - Bridgette D. Semple
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia,Department of NeurologyAlfred HealthPrahranVictoriaAustralia,Department of Medicine (Royal Melbourne Hospital)University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
10
|
Zou D, Qin J, Hu W, Wei Z, Zhan Y, He Y, Zhao C, Li L. Macrophages Rapidly Seal off the Punctured Zebrafish Larval Brain through a Vital Honeycomb Network Structure. Int J Mol Sci 2022; 23:ijms231810551. [PMID: 36142462 PMCID: PMC9503817 DOI: 10.3390/ijms231810551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
There is accumulating evidence that macrophages play additional important roles in tissue damage besides their typical phagocytosis. Although the aggregation of macrophages on injured sites has long been observed, few researchers have focused on the role of the overall structure of macrophage aggregation. In this study, we developed a standardized traumatic brain injury (TBI) model in zebrafish larvae to mimic edema and brain tissue spillage symptoms after severe brain trauma. Using time-lapse imaging, we showed that macrophages/microglia in zebrafish larvae responded rapidly and dominated the surface of injured tissue, forming a meaningful honeycomb network structure through their compact aggregation and connection. Disrupting this structure led to fatal edema-like symptoms with severe loss of brain tissue. Using the RNA-Seq, together with the manipulation of in vitro cell lines, we found that collagen IV was indispensable to the formation of honeycomb network structures. Our study thus revealed a novel perspective regarding macrophages forming a protective compact structure with collagen IV. This honeycomb network structure acted as a physical barrier to prevent tissue loss and maintain brain homeostasis after TBI. This study may provide new evidence of macrophages’ function for the rapid protection of brain tissue after brain injury.
Collapse
Affiliation(s)
- Dandan Zou
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Jie Qin
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Wenlong Hu
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Zongfang Wei
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Yandong Zhan
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Yuepeng He
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Congjian Zhao
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, School of Biomedical Engineering and Informatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Li Li
- Research Center of Stem Cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Correspondence:
| |
Collapse
|
11
|
Mishra N, Agarwal R. Research models of sulfur mustard- and nitrogen mustard-induced ocular injuries and potential therapeutics. Exp Eye Res 2022; 223:109209. [PMID: 35961426 DOI: 10.1016/j.exer.2022.109209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022]
Abstract
Sulfur mustard (SM) is a notorious, bifunctional alkylating vesicant that was first used in warfare during World War I in 1917 and since then has been deployed in numerous skirmishes with its most recent documented use being during the Middle Eastern conflicts. Apart from its use in combat and terrorist activities, continual threat of accidental exposure from old stockpiles and improperly discarded munitions is ever present, especially to the innocent and unassuming civilian populations. SM can cause devastating injuries, depending on the dosage of SM exposure, route of exposure, as well as the physiological conditions of the individuals exposed. The most common routes of exposure are ocular, dermal, and exposure to the lungs and respiratory tissues through inhalation. Eyes are the most susceptible organ to SM-induced toxicities owing to their high moisture content and rapidly dividing cells. Additionally, ocular injury causes the most expeditious disablement of individuals even upon whole-body exposures. Therefore, it is imperative to understand the mechanisms underlying SM-induced ocular toxicity and design therapeutic interventions to prevent/mitigate ocular injuries. Ocular SM exposure may cause a wide range of symptoms such as inflammation, lacrimation, itching, dryness, photophobia, edema of the cornea/sclera/retina/iris, conjunctivitis, degradation of the corneal layer, fusion of two or more ocular layers, neovascularization, fibrosis, and temporary or permanent structural damage to one or more ocular layers. These symptoms may lead to vision impairments, resulting in partial or complete blindness that may be permanent. The highly toxic and exceedingly notorious nature of SM makes it a highly regulated chemical, requiring very expensive licensing, security, and safety requirements; thus, the more easily accessible analogue, nitrogen mustard (NM) that mimics SM-induced toxicity and injuries is employed in plethora of studies conducted in different animal models and culture systems. This review provides a comprehensive account of the injuries and symptoms that occur upon ocular SM exposures in human patients as well as studies in animal (in vivo, ex vivo) and cell (in vitro) models of SM and NM ocular exposures. Special emphasis has been laid on highlighting the strengths and lacunae in the research as well as the possible unexplored avenues of mechanisms underlying mustard-induced ocular injury that can be explored in future research endeavors. Furthermore, development of therapeutic interventions and targets of interest in the ocular system exposed to SM and NM, based on studies in human patients as well as in vivo, ex vivo, and in vitro models has been discussed in great depth, providing a valuable knowledge database to delineate pathways associated with vesicant-induced toxicity, and strategies/diagnostic tools against SM-induced toxicity.
Collapse
Affiliation(s)
- Neha Mishra
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
12
|
Goswami DG, Mishra N, Kant R, Agarwal C, Ammar DA, Petrash JM, Tewari-Singh N, Agarwal R. Effect of dexamethasone treatment at variable therapeutic windows in reversing nitrogen mustard-induced corneal injuries in rabbit ocular in vivo model. Toxicol Appl Pharmacol 2022; 437:115904. [PMID: 35108561 PMCID: PMC8849585 DOI: 10.1016/j.taap.2022.115904] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/25/2022]
Abstract
Nitrogen mustard (NM) is an analogue of the potent vesicating agent sulfur mustard, with well-established ocular injury models in rabbit eyes to study vesicant-induced ocular toxicity. The effects of NM-exposure to eyes may include irritation, redness, inflammation, fibrosis, epithelial degradation, blurred vision, partial/complete blindness, which may be temporary or permanent, depending on the route, duration, and dosage of exposure. Effective countermeasures against vesicant exposure are presently not available and are warranted in case of any terrorist activity or accidental leakage from stockpiles. Herein, our focus was to evaluate whether dexamethasone (DEX), an FDA approved potent corticosteroid with documented anti-inflammatory activities, could be an effective treatment modality. Accordingly, utilizing NM-induced corneal injuries in rabbit ocular in vivo model, we examined and compared the efficacy of DEX treatments when administration was started at early (2 h), intermediate (4 h), and late (6 h) therapeutic windows of intervention after NM-exposure and administered every 8 h thereafter. The effects of NM-exposure and DEX treatments were evaluated on clinical (corneal opacity, ulceration, and neovascularization), biological (epithelial thickness, epithelial-stromal separation, blood vessels density, and inflammatory cell and keratocyte counts) and molecular (COX-2 and VEGF expression) parameters, at day 1, 3, 7 and 14. Results indicated that DEX treatment markedly and effectively reversed the NM-induced injury markers in rabbit corneas. Early administration of DEX at 2 h was found to be most effective in reversing NM-induced corneal injuries, followed by DEX 4 h and DEX 6 h administration initiation, indicating that DEX has best efficacy at the early therapeutic window in our study model.
Collapse
Affiliation(s)
- Dinesh G. Goswami
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Neha Mishra
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Rama Kant
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - David A. Ammar
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - J. Mark Petrash
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America,Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
13
|
WANG FD, LI J, ZHAI X, CHEN R, WANG F. Methane-rich saline restores brain SOD activity and alleviates cognitive impairment in rats with traumatic brain injury. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.54921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Jie LI
- Xi'an Jiaotong University, China
| | - Xu ZHAI
- Xi'an Jiaotong University, China
| | - Rui CHEN
- Xi'an Jiaotong University, China
| | | |
Collapse
|
14
|
Huang CT, Wen YT, Desai TD, Tsai RK. Intravitreal Injection of Long-Acting Pegylated Granulocyte Colony-Stimulating Factor Provides Neuroprotective Effects via Antioxidant Response in a Rat Model of Traumatic Optic Neuropathy. Antioxidants (Basel) 2021; 10:1934. [PMID: 34943037 PMCID: PMC8750325 DOI: 10.3390/antiox10121934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022] Open
Abstract
Traumatic optic neuropathy (TON) may cause severe visual loss following direct or indirect head trauma which may result in optic nerve injuries and therefore contribute to the subsequent loss of retinal ganglion cells by inflammatory mediators and reactive oxygen species (ROS). Granulocyte colony-stimulating factor (G-CSF) provides the anti-inflammatory and anti-oxidative actions but has a short half-life and also induces leukocytosis upon typical systemic administration. The purpose of the present study was to investigate the relationship between the anti-oxidative response and neuroprotective effects of long-acting pegylated human G-CSF (PEG-G-CSF) in a rat model of optic nerve crush (ONC). Adult male Wistar rats (150-180 g) were chosen to have a sham operation in one eye and have ONC in the other. PEG-G-CSF or phosphate-buffered saline (PBS control) was immediately administered after ONC by intravitreal injection (IVI). We found the IVI of PEG-G-CSF does not induce systemic leukocytosis, but increases survival of RGCs and preserves the visual function after ONC. TUNEL assays showed fewer apoptotic cells in the retina in the PEG-G-CSF-treated eyes. The number of sorely ED1-positive cells was attenuated at the lesion site in the PEG-G-CSF-treated eyes. Immunoblotting showed up-regulation of p-Akt1, Nrf2, Sirt3, and HO-1 in the ON of the PEG-G-CSF-treated eyes. Our results demonstrated that one IVI of long-acting PEG-G-CSF is neuroprotective in the rONC. PEG-G-CSF activates the p-Akt1/Nrf2/Sirt3 and the p-Akt1/Nrf2/HO-1 axes to provide the antioxidative action and further attenuated RGC apoptosis and neuroinflammation. This provides crucial preclinical information for the development of alternative therapy with IVI of PEG-G-CSF in TON.
Collapse
Affiliation(s)
- Chin-Te Huang
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
- Department of Ophthalmology, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
| | - Tushar Dnyaneshwar Desai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
| | - Rong-Kung Tsai
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien 970, Taiwan
| |
Collapse
|
15
|
Somebang K, Rudolph J, Imhof I, Li L, Niemi EC, Shigenaga J, Tran H, Gill TM, Lo I, Zabel BA, Schmajuk G, Wipke BT, Gyoneva S, Jandreski L, Craft M, Benedetto G, Plowey ED, Charo I, Campbell J, Ye CJ, Panter SS, Nakamura MC, Eckalbar W, Hsieh CL. CCR2 deficiency alters activation of microglia subsets in traumatic brain injury. Cell Rep 2021; 36:109727. [PMID: 34551293 PMCID: PMC8594931 DOI: 10.1016/j.celrep.2021.109727] [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: 05/30/2019] [Revised: 05/25/2021] [Accepted: 08/27/2021] [Indexed: 12/24/2022] Open
Abstract
In traumatic brain injury (TBI), a diversity of brain resident and peripherally derived myeloid cells have the potential to worsen damage and/or to assist in healing. We define the heterogeneity of microglia and macrophage phenotypes during TBI in wild-type (WT) mice and Ccr2−/− mice, which lack macrophage influx following TBI and are resistant to brain damage. We use unbiased single-cell RNA sequencing methods to uncover 25 microglia, monocyte/macrophage, and dendritic cell subsets in acute TBI and normal brains. We find alterations in transcriptional profiles of microglia subsets in Ccr2−/− TBI mice compared to WT TBI mice indicating that infiltrating monocytes/macrophages influence microglia activation to promote a type I IFN response. Preclinical pharmacological blockade of hCCR2 after injury reduces expression of IFN-responsive gene, Irf7, and improves outcomes. These data extend our understanding of myeloid cell diversity and crosstalk in brain trauma and identify therapeutic targets in myeloid subsets. By single-cell RNA sequencing of traumatically injured and normal brains from wild-type and Ccr2−/− mice, Somebang et al. define microglia, macrophage, and dendritic cell phenotypes in TBI. Targeting mouse and/or human CCR2 reduces specific TBI brain CNS myeloid compartments, dampens type I interferon responses, and improves cognition after TBI.
Collapse
Affiliation(s)
- Kerri Somebang
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA
| | - Joshua Rudolph
- School of Medicine, Lung Biology Center, Division of Pulmonology, UCSF, San Francisco, CA, USA
| | - Isabella Imhof
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA
| | - Luyi Li
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA
| | - Erene C Niemi
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA
| | - Judy Shigenaga
- San Francisco VA Health Care System, San Francisco, CA, USA; Department of Medicine, Division of Endocrinology and Metabolism, UCSF, San Francisco, CA, USA
| | - Huy Tran
- San Francisco VA Health Care System, San Francisco, CA, USA
| | | | - Iris Lo
- Gladstone Institutes, San Francisco, CA, USA
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA; Palo Alto VA Health Care System, Palo Alto, CA, USA
| | - Gabriela Schmajuk
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | - Chun Jimmie Ye
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; Institute for Human Genetics, Department of Epidemiology and Biostatistics, Institute of Computational Health Sciences, University of California, San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - S Scott Panter
- San Francisco VA Health Care System, San Francisco, CA, USA; Department of Neurological Surgery, UCSF, San Francisco, CA, USA
| | - Mary C Nakamura
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA
| | - Walter Eckalbar
- School of Medicine, Lung Biology Center, Division of Pulmonology, UCSF, San Francisco, CA, USA
| | - Christine L Hsieh
- Department of Medicine, Division of Rheumatology, University of California, San Francisco (UCSF), San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA.
| |
Collapse
|
16
|
Semple BD, Raghupathi R. A Pro-social Pill? The Potential of Pharmacological Treatments to Improve Social Outcomes After Pediatric Traumatic Brain Injury. Front Neurol 2021; 12:714253. [PMID: 34489853 PMCID: PMC8417315 DOI: 10.3389/fneur.2021.714253] [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: 05/24/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of injury-induced disability in young children worldwide, and social behavior impairments in this population are a significant challenge for affected patients and their families. The protracted trajectory of secondary injury processes triggered by a TBI during early life-alongside ongoing developmental maturation-offers an extended time window when therapeutic interventions may yield functional benefits. This mini-review explores the scarce but promising pre-clinical literature to date demonstrating that social behavior impairments after early life brain injuries can be modified by drug therapies. Compounds that provide broad neuroprotection, such as those targeting neuroinflammation, oxidative stress, axonal injury and/or myelination, may prevent social behavior impairments by reducing secondary neuropathology. Alternatively, targeted treatments that promote affiliative behaviors, exemplified by the neuropeptide oxytocin, may reduce the impact of social dysfunction after pediatric TBI. Complementary literature from other early life neurodevelopmental conditions such as hypoxic ischemic encephalopathy also provides avenues for future research in neurotrauma. Knowledge gaps in this emerging field are highlighted throughout, toward the goal of accelerating translational research to support optimal social functioning after a TBI during early childhood.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neuroscience, Monash University, Prahran, VIC, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Ramesh Raghupathi
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States.,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
17
|
Bernardi RB, Zanchi ACT, Damaceno-Rodrigues NR, Veras MM, Saldiva PHN, Barros HMT, Rhoden CR. The impact of chronic exposure to air pollution over oxidative stress parameters and brain histology. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:47407-47417. [PMID: 33890219 DOI: 10.1007/s11356-021-14023-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/16/2021] [Indexed: 06/12/2023]
Abstract
Air pollution (AP) triggers neuroinflammation and lipoperoxidation involved in physiopathology of several neurodegenerative diseases. Our study aims to investigate the effect of chronic exposure to ambient AP in oxidative stress (OS) parameters and number of neurons and microglial cells of the cortex and striatum. Seventy-two male Wistar rats were distributed in four groups of exposure: control group (FA), exposed throughout life to filtered air; group PA-FA, pre-natal exposed to polluted air until weaning and then to filtered air; group FA-PA, pre-natal exposed to filtered air until weaning and then to polluted air; and group PA, exposed throughout life to polluted air. After 150 days of exposure, the rats were euthanized for biochemical and histological determinations. The malondialdehyde concentration in the cortex and striatum was significantly higher in the PA group. The activity of superoxide dismutase was significantly decreased in the cortex of all groups exposed to AP while activity of catalase was not modified in the cortex or striatum. The total glutathione concentration was lower in the cortex and higher in the striatum of the FA-PA group. The number of neurons or microglia in the striatum did not differ between FA and PA. On the other hand, neurons and microglia cell numbers were significantly higher in the cortex of the FA-PA group. Our findings suggest that the striatum and cortex have dissimilar thresholds to react to AP exposure and different adaptable responses to chronically AP-induced OS. At least for the cortex, changing to a non-polluted ambient early in life was able to avoid and/or reverse the OS, although some alterations in enzymatic antioxidant system may be permanent. As a result, it is important to clarify the effects of AP in the cortical organization and function because of limited capacity of brain tissue to deal with threatening environments.
Collapse
Affiliation(s)
- Rosane Bossle Bernardi
- Laboratory of OS and Atmospheric Pollution, Health Basic Sciences Department, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.
- Post-Graduate Course in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.
- Pharmacology Division, Basic Health Sciences Department, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.
| | - Ana Cláudia Tedesco Zanchi
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | | | - Mariana Matera Veras
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Paulo Hilário Nascimento Saldiva
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
- Post-Graduate Course in Physiopathology, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Helena Maria Tannhauser Barros
- Post-Graduate Course in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
- Pharmacology Division, Basic Health Sciences Department, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Cláudia Ramos Rhoden
- Laboratory of OS and Atmospheric Pollution, Health Basic Sciences Department, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
- Post-Graduate Course in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
- Pharmacology Division, Basic Health Sciences Department, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| |
Collapse
|
18
|
Trivedi A, Tercovich KG, Casbon AJ, Raber J, Lowell C, Noble-Haeusslein LJ. Neutrophil-specific deletion of Syk results in recruitment-independent stabilization of the barrier and a long-term improvement in cognitive function after traumatic injury to the developing brain. Neurobiol Dis 2021; 157:105430. [PMID: 34153467 PMCID: PMC11302380 DOI: 10.1016/j.nbd.2021.105430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/14/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
While traumatic brain injury (TBI) is the leading cause of death and disability in children, we have yet to identify those pathogenic events that determine the extent of recovery. Neutrophils are best known as "first responders" to sites of infection and trauma where they become fully activated, killing pathogens via proteases that are released during degranulation. However, this activational state may generate substantial toxicity in the young brain after TBI that is partially due to developmentally regulated inadequate antioxidant reserves. Neutrophil degranulation is triggered via a downstream signaling pathway that is dependent on spleen tyrosine kinase (Syk). To test the hypothesis that the activational state of neutrophils is a determinant of early pathogenesis and long-term recovery, we compared young, brain-injured conditional knockouts of Syk (sykf/fMRP8-cre+) to congenic littermates (sykf/f). Based upon flow cytometry, there was an extended recruitment of distinct leukocyte subsets, including Ly6G+/Ly6C- and Ly6G+/Ly6Cint, over the first several weeks post-injury which was similar between genotypes. Subsequent assessment of the acutely injured brain revealed a reduction in blood-brain barrier disruption to both high and low molecular weight dextrans and reactive oxygen species in sykf/fMRP8-cre+ mice compared to congenic littermates, and this was associated with greater preservation of claudin 5 and neuronal integrity, as determined by Western blot analyses. At adulthood, motor learning was less affected in brain-injured sykf/fMRP8-cre+ mice as compared to sykf/f mice. Performance in the Morris Water Maze revealed a robust improvement in hippocampal-dependent acquisition and short and long-term spatial memory retention in sykf/fMRP8-cre+ mice. Subsequent analyses of swim path lengths during hidden platform training and probe trials showed greater thigmotaxis in brain-injured sykf/f mice than sham sykf/f mice and injured sykf/fMRP8-cre+ mice. Our results establish the first mechanistic link between the activation state of neutrophils and long-term functional recovery after traumatic injury to the developing brain. These results also highlight Syk kinase as a novel therapeutic target that could be further developed for the brain-injured child.
Collapse
Affiliation(s)
- Alpa Trivedi
- Departments of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94143, USA; Departments of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Kayleen G Tercovich
- Departments of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Amy Jo Casbon
- Departments of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Clifford Lowell
- Departments of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Linda J Noble-Haeusslein
- Departments of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Departments of Neurology and Psychology, The Dell Medical School and the College of Liberal Arts, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
19
|
Parker KN, Donovan MH, Smith K, Noble-Haeusslein LJ. Traumatic Injury to the Developing Brain: Emerging Relationship to Early Life Stress. Front Neurol 2021; 12:708800. [PMID: 34484104 PMCID: PMC8416304 DOI: 10.3389/fneur.2021.708800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Despite the high incidence of brain injuries in children, we have yet to fully understand the unique vulnerability of a young brain to an injury and key determinants of long-term recovery. Here we consider how early life stress may influence recovery after an early age brain injury. Studies of early life stress alone reveal persistent structural and functional impairments at adulthood. We consider the interacting pathologies imposed by early life stress and subsequent brain injuries during early brain development as well as at adulthood. This review outlines how early life stress primes the immune cells of the brain and periphery to elicit a heightened response to injury. While the focus of this review is on early age traumatic brain injuries, there is also a consideration of preclinical models of neonatal hypoxia and stroke, as each further speaks to the vulnerability of the brain and reinforces those characteristics that are common across each of these injuries. Lastly, we identify a common mechanistic trend; namely, early life stress worsens outcomes independent of its temporal proximity to a brain injury.
Collapse
Affiliation(s)
- Kaila N. Parker
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Michael H. Donovan
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Kylee Smith
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Linda J. Noble-Haeusslein
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
20
|
He J, Mao J, Hou L, Jin S, Wang X, Ding Z, Jin Z, Guo H, Dai R. Minocycline attenuates neuronal apoptosis and improves motor function after traumatic brain injury in rats. Exp Anim 2021; 70:563-569. [PMID: 34349080 PMCID: PMC8614018 DOI: 10.1538/expanim.21-0028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Minocycline is a type of tetracycline antibiotic with broad-spectrum antibacterial activity that has been demonstrated to protect the brain against a series of central nervous system
diseases. However, the precise mechanisms of these neuroprotective actions remain unknown. In the present study, we found that minocycline treatment significantly reduced HT22 cell apoptosis
in a mechanical cell injury model. In addition, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining confirmed the neuroprotective effects of minocycline in
vivo through the inhibition of apoptosis in a rat model of controlled cortical impact (CCI) brain injury. The western blotting analysis revealed that minocycline treatment
significantly downregulated the pro-apoptotic proteins BAX and cleaved caspase-3 and upregulated the anti-apoptotic protein BCL-2. Furthermore, the beam-walking test showed that the
administration of minocycline ameliorated traumatic brain injury (TBI)-induced deficits in motor function. Taken together, these findings suggested that minocycline attenuated neuronal
apoptosis and improved motor function following TBI.
Collapse
Affiliation(s)
- Jihong He
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences
| | | | - Lei Hou
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences
| | - Shimin Jin
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences
| | - Xiaodong Wang
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences
| | - Zhaoqi Ding
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences
| | - Zhene Jin
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences
| | - Hua Guo
- The Second Affiliated Hospital of Nanchang University
| | - Rongxiao Dai
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences
| |
Collapse
|
21
|
Cacciottola L, Donnez J, Dolmans MM. Can Endometriosis-Related Oxidative Stress Pave the Way for New Treatment Targets? Int J Mol Sci 2021; 22:ijms22137138. [PMID: 34281188 PMCID: PMC8267660 DOI: 10.3390/ijms22137138] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Endometriosis is a disease of reproductive age characterized by chronic pelvic pain and infertility. Its pathogenesis is complex and still partially unexplained. However, there is increasing evidence of the role of chronic inflammation, immune system dysregulation, and oxidative stress in its development and progression. The latter appears to be involved in multiple aspects of the disease. Indeed, disease progression sustained by a hyperproliferative phenotype can be related to reactive oxygen species (ROS) imbalance, as numerous experiments using drugs to counteract hyperproliferation have shown in recent years. Chronic pelvic pain is also associated with cell function dysregulation favoring chronic inflammation and oxidative stress, specifically involving macrophages and mast cell activation. Moreover, there is increasing evidence of a role for ROS and impaired mitochondrial function not only as deleterious effectors of the ovarian reserve in patients with endometriomas but also in terms of oocyte quality and, hence, embryo development impairment. Targeting oxidative stress looks to be a promising strategy to both curb endometriotic lesion progression and alleviate endometriosis-associated symptoms of chronic pain and infertility. More investigations are nevertheless needed to develop effective therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Luciana Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Jacques Donnez
- Society for Research into Infertility, 1150 Brussels, Belgium;
- Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium;
- Department of Gynecology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Correspondence: ; Tel.: +32-(0)2-764-5287
| |
Collapse
|
22
|
Liviskie C, McPherson C, Luecke C. Assessment and Management of Delirium in the Pediatric Intensive Care Unit: A Review. J Pediatr Intensive Care 2021; 12:94-105. [PMID: 37082469 PMCID: PMC10113017 DOI: 10.1055/s-0041-1730918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022] Open
Abstract
AbstractMany critically ill patients suffer from delirium which is associated with significant morbidity and mortality. There is a paucity of data about the incidence, symptoms, or treatment of delirium in the pediatric intensive care unit (PICU). Risk factors for delirium are common in the PICU including central nervous system immaturity, developmental delay, mechanical ventilation, and use of anticholinergic agents, corticosteroids, vasopressors, opioids, or benzodiazepines. Hypoactive delirium is the most common subtype in pediatric patients; however, hyperactive delirium has also been reported. Various screening tools are validated in the pediatric population, with the Cornell Assessment of Pediatric Delirium (CAPD) applicable to the largest age range and able to detect signs and symptoms consistent with both hypo- and hyperactive delirium. Treatment of delirium should always include identification and reversal of the underlying etiology, reserving pharmacologic management for those patients without symptom resolution, or with significant impact to medical care. Atypical antipsychotics (olanzapine, quetiapine, and risperidone) should be used first-line in patients requiring pharmacologic treatment owing to their apparent efficacy and low incidence of reported adverse effects. The choice of atypical antipsychotic should be based on adverse effect profile, available dosage forms, and consideration of medication interactions. Intravenous haloperidol may be a potential treatment option in patients unable to tolerate oral medications and with significant symptoms. However, given the high incidence of serious adverse effects with intravenous haloperidol, routine use should be avoided. Dexmedetomidine should be used when sedation is needed and when clinically appropriate, given the positive impact on delirium. Additional well-designed trials assessing screening and treatment of PICU delirium are needed.
Collapse
Affiliation(s)
- Caren Liviskie
- Department of Pharmacy, St. Louis Children's Hospital, St. Louis, Missouri, United States
| | - Christopher McPherson
- Department of Pharmacy, St. Louis Children's Hospital, St. Louis, Missouri, United States
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Caitlyn Luecke
- Department of Pharmacy, St. Louis Children's Hospital, St. Louis, Missouri, United States
| |
Collapse
|
23
|
Schober ME, Requena DF, Ohde JW, Maves S, Pauly JR. Docosahexaenoic acid decreased inflammatory gene expression, but not 18-kDa translocator protein binding, in rat pup brain after controlled cortical impact. J Trauma Acute Care Surg 2021; 90:866-873. [PMID: 33728886 PMCID: PMC8068600 DOI: 10.1097/ta.0000000000003084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Traumatic brain injury is the leading cause of acquired neurologic disability in children. In our model of pediatric traumatic brain injury, controlled cortical impact (CCI) in rat pups, docosahexaenoic acid (DHA) improved lesion volume and cognitive testing as late as postinjury day (PID) 50. Docosahexaenoic acid decreased proinflammatory messenger RNA (mRNA) in microglia and macrophages at PIDs 3 and 7, but not 30. We hypothesized that DHA affected inflammatory markers differentially relative to impact proximity, early and persistently after CCI. METHODS To provide a temporal snapshot of regional neuroinflammation, we measured 18-kDa translocator protein (TSPO) binding using whole brain autoradiography at PIDs 3, 7, 30, and 50. Guided by TSPO results, we measured mRNA levels in contused cortex and underlying hippocampus for genes associated with proinflammatory and inflammation-resolving states at PIDs 2 and 3. RESULTS Controlled cortical impact increased TSPO binding at all time points, most markedly at PID 3 and in regions closest to impact, not blunted by DHA. Controlled cortical impact increased cortical and hippocampal mRNA proinflammatory markers, blunted by DHA at PID 2 in hippocampus. CONCLUSION Controlled cortical impact increased TSPO binding in the immature brain in a persistent manner more intensely with more severe injury, not altered by DHA. Controlled cortical impact increased PIDs 2 and 3 mRNA levels of proinflammatory and inflammation-resolving genes. Docosahexaenoic acid decreased proinflammatory markers associated with inflammasome activation at PID 2. We speculate that DHA's salutary effects on long-term outcomes result from early effects on the inflammasome. Future studies will examine functional effects of DHA on microglia both early and late after CCI.
Collapse
Affiliation(s)
- Michelle Elena Schober
- From the Primary Children's Hospital (M.E.S.), and Division of Critical Care, Department of Pediatrics (M.E.S., D.F.R., S.M.), University of Utah, Salt Lake City, Utah; and Department of Pharmaceutical Sciences (J.W.O., J.K.P.), College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | | | | | | | | |
Collapse
|
24
|
Sharma R, Zamani A, Dill LK, Sun M, Chu E, Robinson MJ, O'Brien TJ, Shultz SR, Semple BD. A systemic immune challenge to model hospital-acquired infections independently regulates immune responses after pediatric traumatic brain injury. J Neuroinflammation 2021; 18:72. [PMID: 33731173 PMCID: PMC7968166 DOI: 10.1186/s12974-021-02114-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/16/2021] [Indexed: 11/10/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of disability in young children, yet the factors contributing to poor outcomes in this population are not well understood. TBI patients are highly susceptible to nosocomial infections, which are mostly acquired within the first week of hospitalization, and such infections may modify TBI pathobiology and recovery. In this study, we hypothesized that a peripheral immune challenge such as lipopolysaccharide (LPS)—mimicking a hospital-acquired infection—would worsen outcomes after experimental pediatric TBI, by perpetuating the inflammatory immune response. Methods Three-week-old male mice received either a moderate controlled cortical impact or sham surgery, followed by a single LPS dose (1 mg/kg i.p.) or vehicle (0.9% saline) at 4 days post-surgery, then analysis at 5 or 8 days post-injury (i.e., 1 or 4 days post-LPS). Results LPS-treated mice exhibited a time-dependent reduction in general activity and social investigation, and increased anxiety, alongside substantial body weight loss, indicating transient sickness behaviors. Spleen-to-body weight ratios were also increased in LPS-treated mice, indicative of persistent activation of adaptive immunity at 4 days post-LPS. TBI + LPS mice showed an impaired trajectory of weight gain post-LPS, reflecting a synergistic effect of TBI and the LPS-induced immune challenge. Flow cytometry analysis demonstrated innate immune cell activation in blood, brain, and spleen post-LPS; however, this was not potentiated by TBI. Cytokine protein levels in serum, and gene expression levels in the brain, were altered in response to LPS but not TBI across the time course. Immunofluorescence analysis of brain sections revealed increased glia reactivity due to injury, but no additive effect of LPS was observed. Conclusions Together, we found that a transient, infection-like systemic challenge had widespread effects on the brain and immune system, but these were not synergistic with prior TBI in pediatric mice. These findings provide novel insight into the potential influence of a secondary immune challenge to the injured pediatric brain, with future studies needed to elucidate the chronic effects of this two-hit insult. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02114-1.
Collapse
Affiliation(s)
- Rishabh Sharma
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Larissa K Dill
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Erskine Chu
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Marcus J Robinson
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia. .,Department of Neurology, Alfred Health, Prahran, VIC, Australia. .,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
25
|
Abstract
Delirium is a frequent complication of critical illness in adult and pediatric populations and is associated with significant morbidity and mortality. Little is known about the incidence, risk, symptoms, or treatment of delirium in the NICU. Only 4 cases of NICU delirium have been reported, but many pediatric studies include infants. The Cornell Assessment of Pediatric Delirium tool has been validated in neonatal and infant populations for identification of delirium. Initial treatment should focus on identification and reversal of the cause, with pharmacologic management reserved for patients with symptoms that do not resolve or that significantly impact medical care. Routine use of intravenous haloperidol should be avoided because of the high incidence of serious adverse effects, but it may be considered in patients with significant symptoms who are unable to take oral medications. Atypical antipsychotics (olanzapine, quetiapine, and risperidone) appear to be efficacious with a low incidence of adverse effects. Risperidone has weight-based dosing and a liquid dosage form available, making it a good option for use in the NICU. Additional data from large cohorts of NICU patients routinely screened for delirium, and treated as indicated, are needed.
Collapse
|
26
|
White matter changes following experimental pediatric traumatic brain injury: an advanced diffusion-weighted imaging investigation. Brain Imaging Behav 2021; 15:2766-2774. [PMID: 33411159 DOI: 10.1007/s11682-020-00433-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/24/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
Pediatric traumatic brain injury (pTBI) is a major community health concern. Due to ongoing maturation, injury to the brain at a young age can have devastating consequences in later life. However, how pTBI affects brain development, including white matter maturation, is still poorly understood. Here, we used advanced diffusion weighted imaging (DWI) to assess chronic white matter changes after experimental pTBI. Mice at post-natal day 21 sustained a TBI using the controlled cortical impact model and magnetic resonance imaging (MRI) was performed at 6 months post-injury using a 4.7 T Bruker scanner. Four diffusion shells with 81 directions and b-values of 1000, 3000, 5000, and 7000s/mm2 were acquired and analyzed using MRtrix3 software. Advanced DWI metrics, including fiber density, fiber cross-section and a combined fiber density and cross-section measure, were investigated together with three track-weighted images (TWI): the average pathlength map, mean curvature and the track density image. These advanced metrics were compared to traditional diffusion tensor imaging (DTI) metrics which indicated that TBI injured mice had reduced fractional anisotropy and increased radial diffusivity in the white matter when compared to age-matched sham controls. Consistent with previous findings, fiber density and TWI metrics appeared to be more sensitive to white matter changes than DTI metrics, revealing widespread reductions in fiber density and TWI metrics in pTBI mice compared to sham controls. These results provide additional support for the use of advanced DWI metrics in assessing white matter degeneration following injury and highlight the chronic outcomes that can follow pTBI.
Collapse
|
27
|
Ginsberg Y, Gutzeit O, Hadad S, Divon MY, Khatib N, Fainaru O, Weiner Z, Beloosesky R. Maternal Progesterone Treatment Reduces Maternal Inflammation-Induced Fetal Brain Injury in a Mouse Model of Preterm Birth. Reprod Sci 2021; 28:166-176. [PMID: 32833191 DOI: 10.1007/s43032-020-00272-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/22/2020] [Indexed: 01/08/2023]
Abstract
Maternal natural vaginal progesterone (nVP) administration has been shown to reduce the risk of preterm birth (PTB). The largest randomized trial of nVP for PTB (OPPTIMUM) noted a sonographic reduction in neonatal brain injury following nVP treatment. We investigated the neuroinflammatory protective effect of maternal nVP in a mouse model for maternal inflammation. Pregnant mice (n = 24) were randomized to nVP (1 mg/day) or vehicle from days 13-16 of gestation. At days 15 and 16, lipopolysaccharide (30 μg) or saline were administered. Mice were sacrificed 4 h following the last injection. Fetal brains and placentas were collected. Levels of NF-κB, nNOS, IL-6, and TNFα were determined by Western blot. Maternal lipopolysaccharide significantly increased fetal brain levels of IL-6 (0.33 ± 0.02 vs. 0.11 ± 0.01 u), TNFα (0.3 ± 0.02 vs. 0.10 ± 0.01 u), NF-κB (0.32 ± 0.01 vs. 0.17 ± 0.01 u), and nNOS (0.24 ± 0.04 vs. 0.08 ± 0.01 u), and reduced the total glutathione levels (0.014 ± 0.001 vs. 0.026 ± 0.001 pmol/μl; p < 0.01) compared with control. Maternal nVP significantly reduced fetal brain levels of IL-6 (0.14 ± 0.01 vs. 0.33 ± 0.02 u), TNFα (0.2 ± 0.06 vs. 0.3 ± 0.02 u), NF-κB (0.16 ± 0.01 vs 0.32 ± 0.01 u), and nNOS (0.14 ± 0.01 vs 0.24 ± 0.04 u), and prevented the reduction of fetal brain total glutathione levels (0.022 ± 0.001 vs. 0.014 ± 0.001 pmol/μl; p < 0.01) to levels similar to controls. A similar pattern was demonstrated in the placenta. Maternal nVP for PTB may protect the fetal brain from inflammation-induced brain injury by inhibiting specific inflammatory and oxidative pathways in both brain and placenta.
Collapse
Affiliation(s)
- Yuval Ginsberg
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel.
| | - Ola Gutzeit
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Salim Hadad
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Michael Y Divon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Lenox Hill Hospital, Northwell Health, New York City, NY, USA
| | - Nizar Khatib
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Ofer Fainaru
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Ron Beloosesky
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| |
Collapse
|
28
|
Preterm birth and sustained inflammation: consequences for the neonate. Semin Immunopathol 2020; 42:451-468. [PMID: 32661735 PMCID: PMC7508934 DOI: 10.1007/s00281-020-00803-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022]
Abstract
Almost half of all preterm births are caused or triggered by an inflammatory process at the feto-maternal interface resulting in preterm labor or rupture of membranes with or without chorioamnionitis (“first inflammatory hit”). Preterm babies have highly vulnerable body surfaces and immature organ systems. They are postnatally confronted with a drastically altered antigen exposure including hospital-specific microbes, artificial devices, drugs, nutritional antigens, and hypoxia or hyperoxia (“second inflammatory hit”). This is of particular importance to extremely preterm infants born before 28 weeks, as they have not experienced important “third-trimester” adaptation processes to tolerate maternal and self-antigens. Instead of a balanced adaptation to extrauterine life, the delicate co-regulation between immune defense mechanisms and immunosuppression (tolerance) to allow microbiome establishment is therefore often disturbed. Hence, preterm infants are predisposed to sepsis but also to several injurious conditions that can contribute to the onset or perpetuation of sustained inflammation (SI). This is a continuing challenge to clinicians involved in the care of preterm infants, as SI is regarded as a crucial mediator for mortality and the development of morbidities in preterm infants. This review will outline the (i) role of inflammation for short-term consequences of preterm birth and (ii) the effect of SI on organ development and long-term outcome.
Collapse
|
29
|
Wang R, He M, Xu J. Serum bilirubin level correlates with mortality in patients with traumatic brain injury. Medicine (Baltimore) 2020; 99:e21020. [PMID: 32629724 PMCID: PMC7337601 DOI: 10.1097/md.0000000000021020] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/14/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023] Open
Abstract
As a catabolic product of hemoglobin, bilirubin has been confirmed playing an important role in the development of various central nervous system disease. The aim of this study is to explore the correlation between serum bilirubin level and mortality in patients with traumatic brain injury (TBI).Patients admitted with traumatic brain injury (TBI) in our hospital between January 2015 and January 2018 were enrolled in this study. Clinical and laboratory data of 361 patients were retrospectively collected to explore the independent risk factors of mortality.The comparison of baseline characteristics showed that non-survivors had lower Glasgow Coma Scale (GCS) (P < .001) and higher level of serum total bilirubin (TBIL) (P < .001) and direct bilirubin (DBIL) (P < .001). We found that only GCS (P < .001), glucose (P < .001), lactate dehydrogenase (LDH) (P = .042) and DBIL (P = .005) were significant risk factors in multivariate logistic regression analysis. GCS and DBIL had comparable AUC value (0.778 vs 0.750, P > .05) on predicting mortality in TBI patients. The AUC value of the combination of GCS and DBIL is higher than the single value of these two factors (P < .05). Moreover, predictive model 1 consisted of GCS, glucose, LDH and DBIL had the highest AUC value of 0.894.DBIL is a significant risk factor of mortality in TBI patients. Assessing the level of DBIL is beneficial for physicians to evaluate severity and predict outcome for TBI patients.
Collapse
Affiliation(s)
| | - Min He
- Department of Critical Care Medicine
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
30
|
Sutherland TC, Geoffroy CG. The Influence of Neuron-Extrinsic Factors and Aging on Injury Progression and Axonal Repair in the Central Nervous System. Front Cell Dev Biol 2020; 8:190. [PMID: 32269994 PMCID: PMC7109259 DOI: 10.3389/fcell.2020.00190] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
In the aging western population, the average age of incidence for spinal cord injury (SCI) has increased, as has the length of survival of SCI patients. This places great importance on understanding SCI in middle-aged and aging patients. Axon regeneration after injury is an area of study that has received substantial attention and made important experimental progress, however, our understanding of how aging affects this process, and any therapeutic effort to modulate repair, is incomplete. The growth and regeneration of axons is mediated by both neuron intrinsic and extrinsic factors. In this review we explore some of the key extrinsic influences on axon regeneration in the literature, focusing on inflammation and astrogliosis, other cellular responses, components of the extracellular matrix, and myelin proteins. We will describe how each element supports the contention that axonal growth after injury in the central nervous system shows an age-dependent decline, and how this may affect outcomes after a SCI.
Collapse
Affiliation(s)
- Theresa C Sutherland
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| |
Collapse
|
31
|
Warnock A, Toomey LM, Wright AJ, Fisher K, Won Y, Anyaegbu C, Fitzgerald M. Damage Mechanisms to Oligodendrocytes and White Matter in Central Nervous System Injury: The Australian Context. J Neurotrauma 2020; 37:739-769. [DOI: 10.1089/neu.2019.6890] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Andrew Warnock
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Lillian M. Toomey
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Alexander J. Wright
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Katherine Fisher
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Yerim Won
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Chidozie Anyaegbu
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| |
Collapse
|
32
|
Schober ME, Requena DF, Maschek JA, Cox J, Parra L, Lolofie A. Effects of controlled cortical impact and docosahexaenoic acid on rat pup fatty acid profiles. Behav Brain Res 2020; 378:112295. [PMID: 31618622 PMCID: PMC6897326 DOI: 10.1016/j.bbr.2019.112295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/23/2019] [Accepted: 10/08/2019] [Indexed: 12/15/2022]
Abstract
Traumatic brain injury (TBI) is the leading cause of acquired neurologic disability in children, particularly in those under four years old. During this period, rapid brain growth demands higher Docosahexaenoic Acid (DHA) intake. DHA is an essential fatty acid and brain cell component derived almost entirely from the diet. DHA improved neurologic outcomes and decreased inflammation after controlled cortical impact (CCI) in 17-day old (P17) rats, our established model of pediatric TBI. In adult rodents, TBI decreases brain DHA. We hypothesized that CCI would decrease rat brain DHA at post injury day (PID) 60, blunted by 0.1% DHA diet. We quantitated fatty acids using Gas Chromatography-Mass Spectrometry. We provided 0.1% DHA before CCI to ensure high DHA in dam milk. We compared brain DHA in rats after 60 days of regular (REG) or DHA diet to SHAM pups on REG diet. Brain DHA decreased in REGCCI, not in DHACCI, relative to SHAMREG. In a subsequent experiment, we gave rat pups DHA or vehicle intraperitoneally after CCI followed by DHA or REG diet for 60 days. REG increased brain Docosapentaenoic Acid (n-6 DPA, a brain DHA deficiency marker) relative to SHAMDHA and DHACCI pups (p < 0.001, diet effect). DHA diet nearly doubled DHA and decreased n-6 DPA in blood but did not increase brain DHA content (p < 0.0001, diet effect). We concluded that CCI or craniotomy alone induces a mild DHA deficit as shown by increased brain DPA.
Collapse
Affiliation(s)
- Michelle E Schober
- Department of Pediatrics, Division of Critical Care, Salt Lake City, UT, 84132, United States.
| | - Daniela F Requena
- Department of Pediatrics, Division of Critical Care, Salt Lake City, UT, 84132, United States.
| | - J Alan Maschek
- Metabolomics, Mass Spectrometry and Proteomics Core of the University of Utah, Salt Lake City, UT, 84132, United States.
| | - James Cox
- Department of Biochemistry, Salt Lake City, UT, 84132, United States; Diabetes and Metabolism Research Center, Salt Lake City, UT, 84132, United States; Metabolomics, Mass Spectrometry and Proteomics Core of the University of Utah, Salt Lake City, UT, 84132, United States.
| | - Leonardo Parra
- Department of Biology, Howard Hughes Medical Institute, Salt Lake City, UT, 84132, United States.
| | - Alyssa Lolofie
- Department of Pediatrics, Division of Critical Care, Salt Lake City, UT, 84132, United States.
| |
Collapse
|
33
|
Affiliation(s)
- Matilde Otero-Losada
- National Research Council. Argentina Marcelo T. de Alvear 2270 3P Buenos Aires (1122), Argentina
| | - Francisco Capani
- National Research Council. Argentina Marcelo T. de Alvear 2270 3P Buenos Aires (1122), Argentina
| |
Collapse
|
34
|
Toro-Urrego N, Turner LF, Avila-Rodriguez MF. New Insights into Oxidative Damage and Iron Associated Impairment in Traumatic Brain Injury. Curr Pharm Des 2020; 25:4737-4746. [DOI: 10.2174/1381612825666191111153802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
:
Traumatic Brain Injury is considered one of the most prevalent causes of death around the world; more
than seventy millions of individuals sustain the condition per year. The consequences of traumatic brain injury on
brain tissue are complex and multifactorial, hence, the current palliative treatments are limited to improve patients’
quality of life. The subsequent hemorrhage caused by trauma and the ongoing oxidative process generated
by biochemical disturbances in the in the brain tissue may increase iron levels and reactive oxygen species. The
relationship between oxidative damage and the traumatic brain injury is well known, for that reason, diminishing
factors that potentiate the production of reactive oxygen species have a promissory therapeutic use. Iron chelators
are molecules capable of scavenging the oxidative damage from the brain tissue and are currently in use for ironoverload-
derived diseases.
:
Here, we show an updated overview of the underlying mechanisms of the oxidative damage after traumatic brain
injury. Later, we introduced the potential use of iron chelators as neuroprotective compounds for traumatic brain
injury, highlighting the action mechanisms of iron chelators and their current clinical applications.
Collapse
Affiliation(s)
- Nicolas Toro-Urrego
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Liliana F. Turner
- Grupo Modelos Experimentales para las Ciencias Zoohumanas - Departamento de Biología Facultad de Ciencias, Universidad del Tolima- Ibagué, Tolima, Colombia
| | - Marco F. Avila-Rodriguez
- Grupo Modelos Experimentales para las Ciencias Zoohumanas - Departamento de Ciencias Clínicas- Facultad de Ciencias de la Salud, Universidad del Tolima- Ibagué, Tolima, Colombia
| |
Collapse
|
35
|
Wright DK, Brady RD, Kamnaksh A, Trezise J, Sun M, McDonald SJ, Mychasiuk R, Kolbe SC, Law M, Johnston LA, O'Brien TJ, Agoston DV, Shultz SR. Repeated mild traumatic brain injuries induce persistent changes in plasma protein and magnetic resonance imaging biomarkers in the rat. Sci Rep 2019; 9:14626. [PMID: 31602002 PMCID: PMC6787341 DOI: 10.1038/s41598-019-51267-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/28/2019] [Indexed: 01/05/2023] Open
Abstract
A single mild traumatic brain injury (mTBI) typically causes only transient symptoms, but repeated mTBI (RmTBI) is associated with cumulative and chronic neurological abnormalities. Clinical management of mTBI is challenging due to the heterogeneous, subjective and transient nature of symptoms, and thus would be aided by objective biomarkers. Promising biomarkers including advanced magnetic resonance imaging (MRI) and plasma levels of select proteins were examined here in a rat model of RmTBI. Rats received either two mild fluid percussion or sham injuries administered five days apart. Rats underwent MRI and behavioral testing 1, 3, 5, 7, and 30 days after the second injury and blood samples were collected on days 1, 7, and 30. Structural and diffusion-weighted MRI revealed that RmTBI rats had abnormalities in the cortex and corpus callosum. Proteomic analysis of plasma found that RmTBI rats had abnormalities in markers indicating axonal and vascular injury, metabolic and mitochondrial dysfunction, and glial reactivity. These changes occurred in the presence of ongoing cognitive and sensorimotor deficits in the RmTBI rats. Our findings demonstrate that RmTBI can result in chronic neurological abnormalities, provide insight into potential contributing pathophysiological mechanisms, and supports the use of MRI and plasma protein measures as RmTBI biomarkers.
Collapse
Affiliation(s)
- David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Alaa Kamnaksh
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Jack Trezise
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Scott C Kolbe
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Leigh A Johnston
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3052, Australia.,Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia. .,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
36
|
Targeting high-mobility group box protein 1 (HMGB1) in pediatric traumatic brain injury: Chronic neuroinflammatory, behavioral, and epileptogenic consequences. Exp Neurol 2019; 320:112979. [DOI: 10.1016/j.expneurol.2019.112979] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/29/2019] [Accepted: 06/18/2019] [Indexed: 11/18/2022]
|
37
|
Lee SW, Jang MS, Jeong SH, Kim H. Exploratory, cognitive, and depressive-like behaviors in adult and pediatric mice exposed to controlled cortical impact. Clin Exp Emerg Med 2019; 6:125-137. [PMID: 31261483 PMCID: PMC6614057 DOI: 10.15441/ceem.18.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/17/2018] [Indexed: 12/13/2022] Open
Abstract
Objective Sequelae of behavioral impairments associated with human traumatic brain injury (TBI) include neurobehavioral problems. We compared exploratory, cognitive, and depressive-like behaviors in pediatric and adult male mice exposed to controlled cortical impact (CCI). Methods Pediatric (21 to 25 days old) and adult (8 to 12 weeks old) male C57Bl/6 mice underwent CCI at a 2-mm depth of deflection. Hematoxylin and eosin staining was performed 3 to 7 days after recovery from CCI, and injury volume was analyzed using ImageJ. Neurobehavioral characterization after CCI was performed using the Barnes maze test (BMT), passive avoidance test, open-field test, light/dark test, tail suspension test, and rotarod test. Acutely and subacutely (3 and 7 days after CCI, respectively), CCI mice showed graded injury compared to sham mice for all analyzed deflection depths. Results Time-dependent differences in injury volume were noted between 3 and 7 days following 2-mm TBI in adult mice. In the BMT, 2-mm TBI adults showed spatial memory deficits compared to sham adults (P<0.05). However, no difference in spatial learning and memory was found between sham and 2-mm CCI groups among pediatric mice. The open-field test, light/dark test, and tail suspension test did not reveal differences in anxiety-like behaviors in both age groups. Conclusion Our findings revealed a graded injury response in both age groups. The BMT was an efficient cognitive test for assessing spatial/non-spatial learning following CCI in adult mice; however, spatial learning impairments in pediatric mice could not be assessed.
Collapse
Affiliation(s)
- Suk-Woo Lee
- Department of Emergency Medicine, Chungbuk National University Hospital, Cheongju, Korea.,Department of Emergency Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Mun-Sun Jang
- Department of Emergency Medicine, Chungbuk National University College of Medicine, Cheongju, Korea.,Department of Emergency Medical Technology, Chungbuk Health & Science University, Cheongju, Korea
| | - Seong-Hae Jeong
- Department of Neurology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hoon Kim
- Department of Emergency Medicine, Chungbuk National University Hospital, Cheongju, Korea.,Department of Emergency Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| |
Collapse
|
38
|
Kostic D, Carlson R, Henke D, Rohn K, Tipold A. Evaluation of IL-1β levels in epilepsy and traumatic brain injury in dogs. BMC Neurosci 2019; 20:29. [PMID: 31208341 PMCID: PMC6580646 DOI: 10.1186/s12868-019-0509-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/03/2019] [Indexed: 11/20/2022] Open
Abstract
Background Epilepsy is a common neurological disease in dogs affecting approximately 0.6–0.75% of the canine population. There is much evidence of neuroinflammation presence in epilepsy, creating new possibilities for the treatment of the disease. An increased expression of interleukin-1 beta (IL-1β) was reported in epileptogenic foci. We hypothesized that there is an elevation of IL-1β in serum and CSF of dogs with epilepsy, as well as in serum of dogs with TBI, reflecting involvement of this cytokine in pathophysiology of naturally occurring canine epilepsy in a clinical setting. Results IL-1β levels were evaluated in CSF and serum of six healthy and 51 dogs with epilepsy (structural and idiopathic). In 16 dogs with TBI, only serum was tested. IL-1β concentrations in CSF were not detectable. Serum values were not elevated in dogs with TBI in comparison to healthy controls (p > 0.05). However, dogs with epilepsy had increased levels of IL-1β in serum (p = 0.003) regardless of the underlying cause of the disease (p = 0.0045). There was no significant relationship between the variables and IL-1β levels. Statistically noticeable (p = 0.0630) was that approximately 10% of dog with epilepsy (R2 = 0.105) had increased seizure frequency and IL-1β elevation. Conclusion Increased IL-1β levels were detected in the peripheral blood in dogs with idiopathic and structural epilepsy leading to the assumption that there is an involvement of inflammation in pathophysiology of epilepsy which should be considered in the search for new therapeutic strategies for this disease. However, to better understand the pathogenic role of this cytokine in epilepsy, further evaluation of IL-1β in brain tissue is desired.
Collapse
Affiliation(s)
- Draginja Kostic
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, Buenteweg 9, 30559, Hannover, Germany. .,Centre for Systems Neuroscience, Hannover, Germany.
| | - Regina Carlson
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, Buenteweg 9, 30559, Hannover, Germany
| | - Diana Henke
- Animal Clinic am Hasenberg, Stuttgart, Germany
| | - Karl Rohn
- Institute of Biometry, Epidemiology, and Information Processing, University of Veterinary Medicine, Hannover, Germany
| | - Andrea Tipold
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, Buenteweg 9, 30559, Hannover, Germany.,Centre for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
39
|
Badaut J, Adami A, Huang L, Obenaus A. Noninvasive magnetic resonance imaging stratifies injury severity in a rodent model of male juvenile traumatic brain injury. J Neurosci Res 2019; 98:129-140. [PMID: 30916808 DOI: 10.1002/jnr.24415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 02/06/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022]
Abstract
Age and severity are significant predictors of traumatic brain injury (TBI) outcomes in the immature brain. TBI studies have segregated TBI injury into three severity groups: mild, moderate, and severe. While mild TBI is most frequent form in children and adults, there is debate over the indicators used to denote mild injury. Clinically, magnetic resonance imaging (MRI) and computed tomography (CT) are used to diagnose the TBI severity when medically warranted. Herein, we induced mild, moderate, and severe TBI in juvenile rats (jTBI) using the controlled cortical impact model. We characterized the temporal and spatial injury after graded jTBI in vivo using high-field MRI at 0.25 (6 hr), 1 and 3 days post-injury (dpi) with comparative histology. Susceptibility-weighted imaging (SWI) for blood and T2-weighted imaging (T2WI) for edema were quantified over the 0.25-3 dpi. Edema volumes increased linearly with severity at 0.25 dpi that slowly continued to decrease over the 3 dpi. In contrast, blood volumes did not decrease over time. Mild TBI had the least amount of blood visible on SWI. Fluoro-jade B (FJB) staining for cell death confirmed increased cellular death with increasing severity and increased FJB + cells in the corpus callosum (CC). Interestingly, the strongest correlation was observed for cell death and the presence of extravascular blood. A clear understanding of acute brain injury (jTBI) and how blood/edema contribute to mild, moderate, and severe jTBI is needed prior to embarking on therapeutic interventions. Noninvasive imaging should be used in mild jTBI to verify lack of overt injury.
Collapse
Affiliation(s)
- Jerome Badaut
- CNRS UMR5287, University of Bordeaux, Bordeaux, France.,Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Arash Adami
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lei Huang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, California
| | - André Obenaus
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California.,Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, UC Riverside, Riverside, California.,Department of Pediatrics, University of California Irvine, Irvine, California
| |
Collapse
|
40
|
von Leden RE, Parker KN, Bates AA, Noble-Haeusslein LJ, Donovan MH. The emerging role of neutrophils as modifiers of recovery after traumatic injury to the developing brain. Exp Neurol 2019; 317:144-154. [PMID: 30876905 DOI: 10.1016/j.expneurol.2019.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/03/2019] [Accepted: 03/08/2019] [Indexed: 12/16/2022]
Abstract
The innate immune response plays a critical role in traumatic brain injury (TBI), contributing to ongoing pathogenesis and worsening long-term outcomes. Here we focus on neutrophils, one of the "first responders" to TBI. These leukocytes are recruited to the injured brain where they release a host of toxic molecules including free radicals, proteases, and pro-inflammatory cytokines, all of which promote secondary tissue damage. There is mounting evidence that the developing brain is more vulnerable to injury that the adult brain. This vulnerability to greater damage from TBI is, in part, attributed to relatively low antioxidant reserves coupled with an early robust immune response. The latter is reflected in enhanced sensitivity to cytokines and a prolonged recruitment of neutrophils into both cortical and subcortical regions. This review considers the contribution of neutrophils to early secondary pathogenesis in the injured developing brain and raises the distinct possibility that these leukocytes, which exhibit phenotypic plasticity, may also be poised to support wound healing. We provide a basic review of the development, life cycle, and granular contents of neutrophils and evaluate their potential as therapeutic targets for early neuroprotection and functional recovery after injury at early age. While neutrophils have been broadly studied in neurotrauma, we are only beginning to appreciate their diverse roles in the developing brain and the extent to which their acute manipulation may result in enduring neurological recovery when TBI is superimposed upon brain development.
Collapse
Affiliation(s)
- Ramona E von Leden
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1701 Trinity St., Austin, TX 78712, USA.
| | - Kaila N Parker
- Department of Psychology, Behavioral Neuroscience, The University of Texas at Austin, 108 E. Dean Keeton St., Austin, TX 78712, USA.
| | - Adrian A Bates
- Institute for Neuroscience, The University of Texas at Austin, 100 E. 24(th) St., Austin, TX 78712, USA.
| | - Linda J Noble-Haeusslein
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1701 Trinity St., Austin, TX 78712, USA; Department of Psychology, Behavioral Neuroscience, The University of Texas at Austin, 108 E. Dean Keeton St., Austin, TX 78712, USA; Institute for Neuroscience, The University of Texas at Austin, 100 E. 24(th) St., Austin, TX 78712, USA.
| | - Michael H Donovan
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1701 Trinity St., Austin, TX 78712, USA.
| |
Collapse
|
41
|
Agoston DV, Kamnaksh A. Protein biomarkers of epileptogenicity after traumatic brain injury. Neurobiol Dis 2019; 123:59-68. [PMID: 30030023 PMCID: PMC6800147 DOI: 10.1016/j.nbd.2018.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a major risk factor for acquired epilepsy. Post-traumatic epilepsy (PTE) develops over time in up to 50% of patients with severe TBI. PTE is mostly unresponsive to traditional anti-seizure treatments suggesting distinct, injury-induced pathomechanisms in the development of this condition. Moderate and severe TBIs cause significant tissue damage, bleeding, neuron and glia death, as well as axonal, vascular, and metabolic abnormalities. These changes trigger a complex biological response aimed at curtailing the physical damage and restoring homeostasis and functionality. Although a positive correlation exists between the type and severity of TBI and PTE, there is only an incomplete understanding of the time-dependent sequelae of TBI pathobiologies and their role in epileptogenesis. Determining the temporal profile of protein biomarkers in the blood (serum or plasma) and cerebrospinal fluid (CSF) can help to identify pathobiologies underlying the development of PTE, high-risk individuals, and disease modifying therapies. Here we review the pathobiological sequelae of TBI in the context of blood- and CSF-based protein biomarkers, their potential role in epileptogenesis, and discuss future directions aimed at improving the diagnosis and treatment of PTE.
Collapse
Affiliation(s)
- Denes V Agoston
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, USA.
| | - Alaa Kamnaksh
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, USA
| |
Collapse
|
42
|
Zhang L, Wang H, Zhou X, Mao L, Ding K, Hu Z. Role of mitochondrial calcium uniporter-mediated Ca 2+ and iron accumulation in traumatic brain injury. J Cell Mol Med 2019; 23:2995-3009. [PMID: 30756474 PMCID: PMC6433723 DOI: 10.1111/jcmm.14206] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 12/28/2018] [Accepted: 01/16/2019] [Indexed: 12/17/2022] Open
Abstract
Previous studies have suggested that the cellular Ca2+ and iron homeostasis, which can be regulated by mitochondrial calcium uniporter (MCU), is associated with oxidative stress, apoptosis and many neurological diseases. However, little is known about the role of MCU‐mediated Ca2+ and iron accumulation in traumatic brain injury (TBI). Under physiological conditions, MCU can be inhibited by ruthenium red (RR) and activated by spermine (Sper). In the present study, we used RR and Sper to reveal the role of MCU in mouse and neuron TBI models. Our results suggested that the Ca2+ and iron concentrations were obviously increased after TBI. In addition, TBI models showed a significant generation of reactive oxygen species (ROS), decrease in adenosine triphosphate (ATP), deformation of mitochondria, up‐regulation of deoxyribonucleic acid (DNA) damage and increase in apoptosis. Blockage of MCU by RR prevented Ca2+ and iron accumulation, abated the level of oxidative stress, improved the energy supply, stabilized mitochondria, reduced DNA damage and decreased apoptosis both in vivo and in vitro. Interestingly, Sper did not increase cellular Ca2+ and iron concentrations, but suppressed the Ca2+ and iron accumulation to benefit the mice in vivo. However, Sper had no significant impact on TBI in vitro. Taken together, our data demonstrated for the first time that blockage of MCU‐mediated Ca2+ and iron accumulation was essential for TBI. These findings indicated that MCU could be a novel therapeutic target for treating TBI.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Lei Mao
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhigang Hu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| |
Collapse
|
43
|
Pavlova V, Filipova E, Uzunova K, Kalinov K, Vekov T. Pioglitazone Therapy and Fractures: Systematic Review and Meta- Analysis. Endocr Metab Immune Disord Drug Targets 2019; 18:502-507. [PMID: 29683100 DOI: 10.2174/1871530318666180423121833] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/22/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Thiazolidinediones are a group of synthetic medications used in type 2 diabetes treatment. Among available thiazolidinediones, pioglitazone is gaining increased attention due to its lower cardiovascular risk in type 2 diabetes mellitus sufferers and seems a promising future therapy. Accumulating evidence suggests that diabetic patients may exert bone fractures due to such treatments. Simultaneously, the female population is thought to be at greater risk. Still, the safety outcomes of pioglitazone treatment especially in terms of fractures are questionable and need to be clarified. METHODS We searched MEDLINE, Scopus, PsyInfo, eLIBRARY.ru electronic databases and clinical trial registries for studies reporting an association between pioglitazone and bone fractures in type 2 diabetes mellitus patients published before Feb 15, 2016. Among 1536 sources that were initially identified, six studies including 3172 patients proved relevant for further analysis. RESULT Pooled analysis of the included studies demonstrated that after treatment with pioglitazone patients with type 2 diabetes mellitus had no significant increase in fracture risk [odds ratio (OR): 1.18, 95% confidence interval (CI): 0.82 to 1.71, p=0.38] compared to other antidiabetic drugs or placebo. Additionally, no association was found between the risk of fractures and pioglitazone therapy duration. The gender of the patients involved was not relevant to the risk of fractures, too. CONCLUSION Pioglitazone treatment in diabetic patients does not increase the incidence of bone fractures. Moreover, there is no significant association between patients' fractures, their gender and the period of exposure to pioglitazone. Additional longitudinal studies need to be undertaken to obtain more detailed information on bone fragility and pioglitazone therapy.
Collapse
Affiliation(s)
- Velichka Pavlova
- Science Department, Tchaikapharma High-Quality Medicines, Inc., 1 G.M. Dimitrov Blvd, 1172 Sofia, Bulgaria
| | - Elena Filipova
- Science Department, Tchaikapharma High-Quality Medicines, Inc., 1 G.M. Dimitrov Blvd, 1172 Sofia, Bulgaria
| | - Katya Uzunova
- Science Department, Tchaikapharma High-Quality Medicines, Inc., 1 G.M. Dimitrov Blvd, 1172 Sofia, Bulgaria
| | - Krassimir Kalinov
- Department of Informatics, New Bulgarian University, 21 Montevideo Street, 1618 Sofia, Bulgaria
| | - Toni Vekov
- Medical University, Faculty of Pharmacy, Dean, Pleven, Bulgaria
| |
Collapse
|
44
|
Guignet M, Lein PJ. Neuroinflammation in organophosphate-induced neurotoxicity. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
45
|
Madathil SK, Wilfred BS, Urankar SE, Yang W, Leung LY, Gilsdorf JS, Shear DA. Early Microglial Activation Following Closed-Head Concussive Injury Is Dominated by Pro-Inflammatory M-1 Type. Front Neurol 2018; 9:964. [PMID: 30498469 PMCID: PMC6249371 DOI: 10.3389/fneur.2018.00964] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022] Open
Abstract
Microglial activation is a pathological hallmark of traumatic brain injury (TBI). Following brain injury, activated microglia/macrophages adopt different phenotypes, generally categorized as M-1, or classically activated, and M-2, or alternatively activated. While the M-1, or pro-inflammatory phenotype is detrimental to recovery, M-2, or the anti-inflammatory phenotype, aids in brain repair. Recent findings also suggest the existence of mixed phenotype following brain injury, where activated microglia simultaneously express both M-1 and M-2 markers. The present study sought to determine microglial activation states at early time points (6-72 h) following single or repeated concussive injury in rats. Closed-head concussive injury was modeled in rats using projectile concussive impact injury, with either single or repeated impacts (4 impacts, 1 h apart). Brain samples were examined using immunohistochemical staining, inflammatory gene profiling and real-time polymerase chain reaction analyses to detect concussive injury induced changes in microglial activation and phenotype in cortex and hippocampal regions. Our findings demonstrate robust microglial activation following concussive brain injury. Moreover, we show that multiple concussions induced a unique rod-shaped microglial morphology that was also observed in other diffuse brain injury models. Histological studies revealed a predominance of MHC-II positive M-1 phenotype in the post-concussive microglial milieu following multiple impacts. Although there was simultaneous expression of M-1 and M-2 markers, gene expression results indicate a clear dominance in M-1 pro-inflammatory markers following both single and repeated concussions. While the increase in M-1 markers quickly resolved after a single concussion, they persisted following repeated concussions, indicating a pro-inflammatory environment induced by multiple concussions that may delay recovery and contribute to long-lasting consequences of concussion.
Collapse
Affiliation(s)
- Sindhu K Madathil
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Bernard S Wilfred
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Sarah E Urankar
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Weihong Yang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Lai Yee Leung
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Janice S Gilsdorf
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Deborah A Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
46
|
Xie BS, Wang YQ, Lin Y, Mao Q, Feng JF, Gao GY, Jiang JY. Inhibition of ferroptosis attenuates tissue damage and improves long-term outcomes after traumatic brain injury in mice. CNS Neurosci Ther 2018; 25:465-475. [PMID: 30264934 DOI: 10.1111/cns.13069] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/30/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS Ferroptosis, a new form of iron-dependent programmed cell death, has been shown to be involved in a range of diseases. However, the role of ferroptosis in traumatic brain injury (TBI) has yet to be elucidated. We aimed to investigate whether ferroptosis is induced after TBI and whether the inhibition of ferroptosis would protect against traumatic brain injury in a controlled cortical impact injury (CCI) mouse model. METHODS After establishing the TBI model in mice, we determined the biochemical and morphological changes associated with ferroptosis, including iron accumulation with Perl's staining, neuronal cell death with Fluoro-Jade B (FJB) staining, iron metabolism dysfunction with Western blotting, reactive oxygen species (ROS) accumulation with malondialdehyde (MDA) assays, and shrunken mitochondria with transmission electron microscopy. Furthermore, a specific inhibitor of ferroptosis, ferrostatin-1(fer-1), was administrated by cerebral ventricular injection after CCI. We used cresyl violet (CV) staining to assess lesion volume, along with the Morris water maze and beam walk test to evaluate long-term outcomes. RESULTS TBI was followed by iron accumulation, dysfunctional iron metabolism, the upregulation of ferroptosis-related genes, reduced glutathione peroxidase (GPx) activity, and the accumulation of lipid-reactive oxygen species (ROS). Three days (d) after TBI, transmission electron microscopy (TEM) confirmed that the mitochondria had shrunk a typical characteristic of ferroptosis. Importantly, the administration of Fer-1 by cerebral ventricular injection significantly reduced iron deposition and neuronal degeneration while attenuating injury lesions and improving long-term motor and cognitive function. CONCLUSION This study demonstrated an effective method with which to treat TBI by targeting ferroptosis.
Collapse
Affiliation(s)
- Bao-Shu Xie
- Department of Neurosurgery, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Head Trauma, Shanghai, China
| | - Yi-Qin Wang
- Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Lin
- Department of Neurosurgery, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Head Trauma, Shanghai, China
| | - Qing Mao
- Department of Neurosurgery, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun-Feng Feng
- Department of Neurosurgery, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Head Trauma, Shanghai, China
| | - Guo-Yi Gao
- Department of Neurosurgery, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Head Trauma, Shanghai, China
| | - Ji-Yao Jiang
- Department of Neurosurgery, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Head Trauma, Shanghai, China
| |
Collapse
|
47
|
Neuroinflammation and Seizures After Pediatric Brain Injury: What a Headache! Epilepsy Curr 2018; 18:246-247. [PMID: 30254521 DOI: 10.5698/1535-7597.18.4.246] [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
|
48
|
Khalaf S, Ahmad AS, Chamara KR, Doré S. Unique Properties Associated with the Brain Penetrant Iron Chelator HBED Reveal Remarkable Beneficial Effects after Brain Trauma. J Neurotrauma 2018; 36:43-53. [PMID: 29743006 PMCID: PMC6306957 DOI: 10.1089/neu.2017.5617] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Iron is postulated to contribute to secondary injury after brain trauma through various pathways including oxidative stress and inflammation. Therefore, one goal is to limit iron toxicity by either directly limiting iron activity, or limiting the secondary cascade mediated by iron, therefore rescuing the brain from damage after trauma. The N,N'-Di(2-hydroxybenzyl)ethylenediamine-N,N'-diacetic acid monohydrochloride (HBED) is a unique iron chelator that has the ability to cross the intact blood-brain barrier; it has a higher affinity to iron, and it has a longer half-life than most commonly used chelators. A controlled-cortical impact model of traumatic brain injury (TBI) was induced in mice. Mice were subcutaneously injected with HBED immediately after TBI, then at 12 h after, followed by a twice-a-day regimen until an end-point of 3 days. Neurobehavioral tests were performed daily. Cortical injury volume, hemispheric enlargement, and hippocampal swelling were quantified. Perls' iron immunostaining along with markers of gliosis, oxidative stress, and aquaporin (AQP) 4 were also performed. Data revealed that HBED treatment significantly decreases motor deficits and improves recovery after TBI. It also reduces cortical injury volume by 36.6 ± 6.8% (p < 0.001), hippocampal swelling by 23.4 ± 3.8% (p < 0.05), and total hemispheric volume by 13.3 ± 2.7% (p < 0.01). These effects are related to a reduction in microgliosis and oxidiative stress markers in the impacted corpus callosum area by 39.8 ± 7.3%, and by 80.5 ± 0.8% (p < 0.05), respectively. AQP4 staining is also attenuated in the hippocampus of HBED-treated mice. Therefore, our results suggest that HBED should be considered as a therapeutic tool to facilitate the recovery process following brain trauma.
Collapse
Affiliation(s)
- Saher Khalaf
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Abdullah Shafique Ahmad
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - K.V.D. Ranga Chamara
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
- Departments of Neurology, Psychiatry, Pharmaceutics, Psychology, and Neuroscience, University of Florida, Gainesville, Florida
| |
Collapse
|
49
|
CE: Original Research: Recognizing Delirium in Hospitalized Children: A Systematic Review of the Evidence on Risk Factors and Characteristics. Am J Nurs 2018. [PMID: 29543606 DOI: 10.1097/01.naj.0000532069.55339.f9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
: Purpose: The purpose of this study was to examine the evidence regarding the risk factors for and characteristics of acute pediatric delirium in hospitalized children. METHODS The systematic review method within an epidemiological framework of person, place, and time was used. Fifty-two studies were selected for initial retrieval. Of these, after assessment for methodological quality, 21 studies involving 2,616 subjects were included in the review. RESULTS Findings revealed five primary characteristics seen in children experiencing delirium: agitation, disorientation, hallucinations, inattention, and sleep-wake cycle disturbances. Children who were more seriously ill, such as those in a pediatric ICU (PICU) and those with a high Pediatric Risk of Mortality II (PRISM II) score, and children who were mechanically ventilated were at greater risk for development of delirium. Those with a developmental delay or a preexisting anxiety disorder were also more prone to delirium. Although delirium symptoms fluctuate, most episodes occurred at night. Boys were slightly more susceptible than girls, though this difference was not significant. A key finding of this review was that delirium is multifactorial, related to treatment (mechanical ventilation, for example) and to a hospital environment (such as a PICU) that deprives patients of normal sleep-wake cycles and familiar routines. CONCLUSION These findings will be useful in efforts to achieve earlier recognition and better management or prevention of pediatric delirium. This may also help to prevent unnecessary laboratory testing and imaging studies, which can cause children and parents unnecessary pain and anxiety and increase hospital costs.
Collapse
|
50
|
Chen J, Hu J, Liu H, Xiong Y, Zou Y, Huang W, Shao M, Wu J, Yu L, Wang X, Wang X, Lin L. FGF21 Protects the Blood-Brain Barrier by Upregulating PPARγ via FGFR1/β-klotho after Traumatic Brain Injury. J Neurotrauma 2018; 35:2091-2103. [PMID: 29648978 DOI: 10.1089/neu.2017.5271] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Blood-brain barrier (BBB) disruption and dysfunction result in brain edema, which is responsible for more than half of all deaths after severe traumatic brain injury (TBI). Fibroblast growth factor 21 (FGF21) has a potential neuroprotective function in the brain. However, the effects and underlying possible mechanism of action on BBB integrity following TBI remain unknown. The purpose of the current study was to determine the effects of FGF21 on BBB protection and TBI treatment. The effects of recombinant human FGF21 (rhFGF21) on BBB integrity and on tight junction (TJ) and adhesion junction (AJ) proteins were investigated both in a TBI mouse model and an in vitro BBB disruption model established with tumor necrosis factor alpha (TNF-α)-induced human brain microvascular endothelial cells (HBMECs). The ability of rhFGF21 to form an FGF21/FGFR1/β-klotho complex was confirmed by in vitro β-klotho small interfering RNA (siRNA) transfection and FGFR1 co-immunoprecipitation. In addition, the specific FGFR1 and peroxisome proliferator-activated receptor gamma (PPARγ) inhibitors PD173074 and GW9662, respectively, were applied to further explore the possible mechanism of rhFGF21 in BBB maintenance after TBI. rhFGF21 markedly reduced neurofunctional behavior deficits and cerebral edema degree, preserved BBB integrity, and recued brain tissue loss and neuron apoptosis in the mouse model after TBI. Both in vivo and in vitro, rhFGF21 upregulated TJ and AJ proteins, thereby preserving the BBB. Moreover, rhFGF21 activated PPARγ in TNF-α-induced HBMECs through formation of an FGF21/FGFR1/β-klotho complex. rhFGF21 protected the BBB through FGF21/FGFR1/β-klotho complex formation and PPARγ activation, which upregulated TJ and AJ proteins.
Collapse
Affiliation(s)
- Jun Chen
- 1 School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Jian Hu
- 1 School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Huan Liu
- 1 School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Ye Xiong
- 2 The First Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Yuchi Zou
- 2 The First Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Wenting Huang
- 3 School of the First Clinical Medical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Mingjie Shao
- 3 School of the First Clinical Medical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Jiamin Wu
- 1 School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Li Yu
- 4 The Affiliated Hospital of Medical School of Ningbo University , Ningbo, Zhejiang, China
| | - Xiaojie Wang
- 1 School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Xue Wang
- 1 School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Li Lin
- 1 School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou, Zhejiang, China
| |
Collapse
|