1
|
Zima L, Moore AN, Smolen P, Kobori N, Noble B, Robinson D, Hood KN, Homma R, Al Mamun A, Redell JB, Dash PK. The evolving pathophysiology of TBI and the advantages of temporally-guided combination therapies. Neurochem Int 2024; 180:105874. [PMID: 39366429 DOI: 10.1016/j.neuint.2024.105874] [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: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Several clinical and experimental studies have demonstrated that traumatic brain injury (TBI) activates cascades of biochemical, molecular, structural, and pathological changes in the brain. These changes combine to contribute to the various outcomes observed after TBI. Given the breadth and complexity of changes, combination treatments may be an effective approach for targeting multiple detrimental pathways to yield meaningful improvements. In order to identify targets for therapy development, the temporally evolving pathophysiology of TBI needs to be elucidated in detail at both the cellular and molecular levels, as it has been shown that the mechanisms contributing to cognitive dysfunction change over time. Thus, a combination of individual mechanism-based therapies is likely to be effective when maintained based on the time courses of the cellular and molecular changes being targeted. In this review, we will discuss the temporal changes of some of the key clinical pathologies of human TBI, the underlying cellular and molecular mechanisms, and the results from preclinical and clinical studies aimed at mitigating their consequences. As most of the pathological events that occur after TBI are likely to have subsided in the chronic stage of the disease, combination treatments aimed at attenuating chronic conditions such as cognitive dysfunction may not require the initiation of individual treatments at a specific time. We propose that a combination of acute, subacute, and chronic interventions may be necessary to maximally improve health-related quality of life (HRQoL) for persons who have sustained a TBI.
Collapse
Affiliation(s)
- Laura Zima
- Departments of Neurosurgery, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Anthony N Moore
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Paul Smolen
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Nobuhide Kobori
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Brian Noble
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Dustin Robinson
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Kimberly N Hood
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Ryota Homma
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Amar Al Mamun
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - John B Redell
- Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA
| | - Pramod K Dash
- Departments of Neurosurgery, The University of Texas McGovern Medical School, Houston, TX, USA; Departments of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
2
|
Machado CA, Oliveira BDS, de Barros JLVM, Fernandes HDB, de Brito Toscano EC, Kangussu LM, Guimarães PPG, Simões E Silva AC, Teixeira AL, de Miranda AS. Involvement of Renin-Angiotensin system (RAS) components in mild traumatic brain injury. Brain Res 2024; 1846:149266. [PMID: 39374839 DOI: 10.1016/j.brainres.2024.149266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
The Renin Angiotensin System (RAS) plays a pathophysiological role in traumatic brain injury (TBI) but the evidence of its involvement in mild TBI (mTBI) is still limited. We aimed at investigating the levels of components from both the classical and counter-regulatory axis of the RAS in a mTBI animal model. Mice with mTBI displayed enhanced ACE/Ang II/AT1R axis ipsilateral- and contralaterally to the trauma in the hippocampus and prefrontal cortex during acute (24 and 72 h) and later (30 days) timepoints. Increase in Ang-(1-7) levels alongside reduction in Mas receptor expression in hippocampus and prefrontal cortex was also observed after injury. Conversely, mTBI-mice presented higher expression of AT2 receptor in the contralateral hippocampus and the ipsilateral prefrontal cortex. Importantly, treatment with telmisartan, an AT1R blocker, and perindopril, an ACE inhibitor, were able to prevent mTBI-associated locomotor activity impairment and anxiety-like behavior, corroborating the involvement of RAS in the pathophysiology of mTBI. We provided original evidence that components of classical and alternative RAS axes undergo alterations in key brain areas following a mTBI in a time and hemisphere dependent manner. Our findings also open new avenues for investigating the therapeutic potential of RAS components in mTBI.
Collapse
Affiliation(s)
- Caroline Amaral Machado
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Bruna da Silva Oliveira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Heliana de Barros Fernandes
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Lucas Miranda Kangussu
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antonio Lucio Teixeira
- The Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aline Silva de Miranda
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
3
|
Ritter K, Somnuke P, Hu L, Griemert EV, Schäfer MKE. Current state of neuroprotective therapy using antibiotics in human traumatic brain injury and animal models. BMC Neurosci 2024; 25:10. [PMID: 38424488 PMCID: PMC10905838 DOI: 10.1186/s12868-024-00851-6] [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: 09/25/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
TBI is a leading cause of death and disability in young people and older adults worldwide. There is no gold standard treatment for TBI besides surgical interventions and symptomatic relief. Post-injury infections, such as lower respiratory tract and surgical site infections or meningitis are frequent complications following TBI. Whether the use of preventive and/or symptomatic antibiotic therapy improves patient mortality and outcome is an ongoing matter of debate. In contrast, results from animal models of TBI suggest translational perspectives and support the hypothesis that antibiotics, independent of their anti-microbial activity, alleviate secondary injury and improve neurological outcomes. These beneficial effects were largely attributed to the inhibition of neuroinflammation and neuronal cell death. In this review, we briefly outline current treatment options, including antibiotic therapy, for patients with TBI. We then summarize the therapeutic effects of the most commonly tested antibiotics in TBI animal models, highlight studies identifying molecular targets of antibiotics, and discuss similarities and differences in their mechanistic modes of action.
Collapse
Affiliation(s)
- Katharina Ritter
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), Mainz, 55131, Germany
| | - Pawit Somnuke
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), Mainz, 55131, Germany
- Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Lingjiao Hu
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), Mainz, 55131, Germany
- Department of Gastroenterology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Eva-Verena Griemert
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), Mainz, 55131, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), Mainz, 55131, Germany.
- Focus Program Translational Neurosciences (FTN, Johannes Gutenberg-University Mainz, Mainz, Germany.
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg- University Mainz, Mainz, Germany.
| |
Collapse
|
4
|
Vedaei F, Newberg AB, Alizadeh M, Zabrecky G, Navarreto E, Hriso C, Wintering N, Mohamed FB, Monti D. Treatment effects of N-acetyl cysteine on resting-state functional MRI and cognitive performance in patients with chronic mild traumatic brain injury: a longitudinal study. Front Neurol 2024; 15:1282198. [PMID: 38299014 PMCID: PMC10829764 DOI: 10.3389/fneur.2024.1282198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Mild traumatic brain injury (mTBI) is a significant public health concern, specially characterized by a complex pattern of abnormal neural activity and functional connectivity. It is often associated with a broad spectrum of short-term and long-term cognitive and behavioral symptoms including memory dysfunction, headache, and balance difficulties. Furthermore, there is evidence that oxidative stress significantly contributes to these symptoms and neurophysiological changes. The purpose of this study was to assess the effect of N-acetylcysteine (NAC) on brain function and chronic symptoms in mTBI patients. Fifty patients diagnosed with chronic mTBI participated in this study. They were categorized into two groups including controls (CN, n = 25), and patients receiving treatment with N-acetyl cysteine (NAC, n = 25). NAC group received 50 mg/kg intravenous (IV) medication once a day per week. In the rest of the week, they took one 500 mg NAC tablet twice per day. Each patient underwent rs-fMRI scanning at two timepoints including the baseline and 3 months later at follow-up, while the NAC group received a combination of oral and IV NAC over that time. Three rs-fMRI metrics were measured including fractional amplitude of low frequency fluctuations (fALFF), degree centrality (DC), and functional connectivity strength (FCS). Neuropsychological tests were also assessed at the same day of scanning for each patient. The alteration of rs-fMRI metrics and cognitive scores were measured over 3 months treatment with NAC. Then, the correlation analysis was executed to estimate the association of rs-fMRI measurements and cognitive performance over 3 months (p < 0.05). Two significant group-by-time effects demonstrated the changes of rs-fMRI metrics particularly in the regions located in the default mode network (DMN), sensorimotor network, and emotional circuits that were significantly correlated with cognitive function recovery over 3 months treatment with NAC (p < 0.05). NAC appears to modulate neural activity and functional connectivity in specific brain networks, and these changes could account for clinical improvement. This study confirmed the short-term therapeutic efficacy of NAC in chronic mTBI patients that may contribute to understanding of neurophysiological effects of NAC in mTBI. These findings encourage further research on long-term neurobehavioral assessment of NAC assisting development of therapeutic plans in mTBI.
Collapse
Affiliation(s)
- Faezeh Vedaei
- Department of Radiology, Jefferson Integrated Magnetic Resonance Imaging Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Andrew B. Newberg
- Department of Radiology, Jefferson Integrated Magnetic Resonance Imaging Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Mahdi Alizadeh
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - George Zabrecky
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Emily Navarreto
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Chloe Hriso
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Nancy Wintering
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| | - Feroze B. Mohamed
- Department of Radiology, Jefferson Integrated Magnetic Resonance Imaging Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Daniel Monti
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
5
|
Fesharaki-Zadeh A. Navigating the Complexities of Traumatic Encephalopathy Syndrome (TES): Current State and Future Challenges. Biomedicines 2023; 11:3158. [PMID: 38137378 PMCID: PMC10740836 DOI: 10.3390/biomedicines11123158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a unique neurodegenerative disease that is associated with repetitive head impacts (RHI) in both civilian and military settings. In 2014, the research criteria for the clinical manifestation of CTE, traumatic encephalopathy syndrome (TES), were proposed to improve the clinical identification and understanding of the complex neuropathological phenomena underlying CTE. This review provides a comprehensive overview of the current understanding of the neuropathological and clinical features of CTE, proposed biomarkers of traumatic brain injury (TBI) in both research and clinical settings, and a range of treatments based on previous preclinical and clinical research studies. Due to the heterogeneity of TBI, there is no universally agreed-upon serum, CSF, or neuroimaging marker for its diagnosis. However, as our understanding of this complex disease continues to evolve, it is likely that there will be more robust, early diagnostic methods and effective clinical treatments. This is especially important given the increasing evidence of a correlation between TBI and neurodegenerative conditions, such as Alzheimer's disease and CTE. As public awareness of these conditions grows, it is imperative to prioritize both basic and clinical research, as well as the implementation of necessary safe and preventative measures.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Department of Neurology and Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
6
|
Clark RSB, Empey PE, Kochanek PM, Bell MJ. N-Acetylcysteine and Probenecid Adjuvant Therapy for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1529-1537. [PMID: 37596428 PMCID: PMC10684451 DOI: 10.1007/s13311-023-01422-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 08/20/2023] Open
Abstract
N-Acetylcysteine (NAC) has shown promise as a putative neurotherapeutic for traumatic brain injury (TBI). Yet, many such promising compounds have limited ability to cross the blood-brain barrier (BBB), achieve therapeutic concentrations in brain, demonstrate target engagement, among other things, that have hampered successful translation. A pharmacologic strategy for overcoming poor BBB permeability and/or efflux out of the brain of organic acid-based, small molecule therapeutics such as NAC is co-administration with a targeted or nonselective membrane transporter inhibitor. Probenecid is a classic ATP-binding cassette and solute carrier inhibitor that blocks transport of organic acids, including NAC. Accordingly, combination therapy using probenecid as an adjuvant with NAC represents a logical neurotherapeutic strategy for treatment of TBI (and other CNS diseases). We have completed a proof-of-concept pilot study using this drug combination in children with severe TBI-the Pro-NAC Trial (ClinicalTrials.gov NCT01322009). In this review, we will discuss the background and rationale for combination therapy with probenecid and NAC in TBI, providing justification for further clinical investigation.
Collapse
Affiliation(s)
- Robert S B Clark
- Departments of Critical Care Medicine and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| | - Philip E Empey
- Department of Pharmacy & Therapeutics, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Departments of Critical Care Medicine, Anesthesiology, and Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael J Bell
- Division of Critical Care Medicine, Department of Pediatrics, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
7
|
Bergold PJ, Furhang R, Lawless S. Treating Traumatic Brain Injury with Minocycline. Neurotherapeutics 2023; 20:1546-1564. [PMID: 37721647 PMCID: PMC10684850 DOI: 10.1007/s13311-023-01426-9] [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] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Traumatic brain injury (TBI) results in both rapid and delayed brain damage. The speed, complexity, and persistence of TBI present large obstacles to drug development. Preclinical studies from multiple laboratories have tested the FDA-approved anti-microbial drug minocycline (MINO) to treat traumatic brain injury. At concentrations greater than needed for anti-microbial action, MINO readily inhibits microglial activation. MINO has additional pleotropic effects including anti-inflammatory, anti-oxidant, and anti-apoptotic activities. MINO inhibits multiple proteins that promote brain injury including metalloproteases, caspases, calpain, and polyADP-ribose-polymerase-1. At these elevated doses, MINO is well tolerated and enters the brain even when the blood-brain barrier is intact. Most preclinical studies with a first dose of MINO at less than 1 h after injury have shown improved multiple outcomes after TBI. Fewer studies with more delayed dosing have yielded similar results. A small number of clinical trials for TBI have established the safety of MINO and suggested some drug efficacy. Studies are also ongoing that either improve MINO pharmacology or combine MINO with other drugs to increase its therapeutic efficacy against TBI. This review builds upon a previous, recent review by some of the authors (Lawless and Bergold, Neural Regen Res 17:2589-92, 2022). The present review includes the additional preclinical studies examining the efficacy of minocycline in preclinical TBI models. This review also includes recommendations for a clinical trial to test MINO to treat TBI.
Collapse
Affiliation(s)
- Peter J Bergold
- Graduate Programs in Neural and Behavioral Sciences, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA.
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA.
| | - Rachel Furhang
- Graduate Programs in Neural and Behavioral Sciences, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA
| | - Siobhán Lawless
- Graduate Programs in Neural and Behavioral Sciences, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA
| |
Collapse
|
8
|
Ezzat GM, Nassar AY, Bakr MH, Mohamed S, Nassar GA, Kamel AA. Acetylated Oligopeptide and N-acetyl cysteine Protected Against Oxidative Stress, Inflammation, Testicular-Blood Barrier Damage, and Testicular Cell Death in Iron-Overload Rat Model. Appl Biochem Biotechnol 2023; 195:5053-5071. [PMID: 36947366 DOI: 10.1007/s12010-023-04457-2] [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] [Accepted: 03/15/2023] [Indexed: 03/23/2023]
Abstract
Multiple organs, including the testes, are damaged by iron overload. It has been shown that N-acetyl cysteine (NAC) influences oxidative stress in iron overload. The present study aimed to evaluate the roles of acetylated peptide (AOP) and NAC in the inhibition of iron-overload induced-testicular damage. At the beginning of the experiment, NAC (150 mg /kg) was given for a week to all 40 rats. Then, four groups were formed by dividing the animals (10 rats/group). Group I included healthy control rats. Group II (iron overload) was given intraperitoneal iron dextran (60 mg/kg/day) 5 days a week for 4 weeks. Group III (NAC) was given NAC orally at a dose of 150 mg/kg/day for 4 weeks in addition to iron dextran. Group IV (AOP) was given AOP orally at a dose of 150 mg/kg/day for 4 weeks besides iron dextran. When the experiment time was over, testosterone serum level, testicular B cell lymphoma-2 (BCL-2) and protein kinase B (PKB) protein levels, nuclear factor kappa-B (NF-κB), and Beclin1 mRNA expression levels, and malondialdehyde (MDA), and reduced glutathione (GSH) were determined by ELISA, quantitative reverse transcription-PCR, and chemical methods. Finally, histopathological examinations and immunohistochemical detection of claudin-1 and CD68 were performed. The iron overload group exhibited decreased testosterone, BCL-2, PKB, claudin-1, and GSH and increased MDA, NF-κB, Beclin1, and CD68, while both NAC and AOP treatments protected against the biochemical and histopathological disturbances occurring in the iron overload model. We concluded that NAC and AOP can protect against testes damage by iron overload via their antioxidant, anti-inflammatory, antiapoptotic, and ant-autophagic properties. The NAC and AOP may be used as preventative measures against iron overload-induced testicular damage.
Collapse
Affiliation(s)
- Ghada M Ezzat
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Ahmed Y Nassar
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwa H Bakr
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Shimma Mohamed
- Department of Medical Biochemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Gamal A Nassar
- Metabolic and Genetic disorders unit, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amira A Kamel
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
9
|
Wang J, Venugopal J, Silaghi P, Su EJ, Guo C, Lawrence DA, Eitzman DT. Beta1-receptor blockade attenuates atherosclerosis progression following traumatic brain injury in apolipoprotein E deficient mice. PLoS One 2023; 18:e0285499. [PMID: 37235594 PMCID: PMC10218730 DOI: 10.1371/journal.pone.0285499] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with cardiovascular mortality in humans. Enhanced sympathetic activity following TBI may contribute to accelerated atherosclerosis. The effect of beta1-adrenergic receptor blockade on atherosclerosis progression induced by TBI was studied in apolipoprotein E deficient mice. Mice were treated with metoprolol or vehicle following TBI or sham operation. Mice treated with metoprolol experienced a reduced heart rate with no difference in blood pressure. Six weeks following TBI, mice were sacrificed for analysis of atherosclerosis. Total surface area and lesion thickness, analyzed at the level of the aortic valve, was found to be increased in mice receiving TBI with vehicle treatment but this effect was ameliorated in TBI mice receiving metoprolol. No effect of metoprolol on atherosclerosis was observed in mice receiving only sham operation. In conclusion, accelerated atherosclerosis following TBI is reduced with beta-adrenergic receptor antagonism. Beta blockers may be useful to reduce vascular risk associated with TBI.
Collapse
Affiliation(s)
- Jintao Wang
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jessica Venugopal
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Paul Silaghi
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Enming J. Su
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Chiao Guo
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Daniel A. Lawrence
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Daniel T. Eitzman
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
10
|
Lynch DG, Narayan RK, Li C. Multi-Mechanistic Approaches to the Treatment of Traumatic Brain Injury: A Review. J Clin Med 2023; 12:jcm12062179. [PMID: 36983181 PMCID: PMC10052098 DOI: 10.3390/jcm12062179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Despite extensive research efforts, the majority of trialed monotherapies to date have failed to demonstrate significant benefit. It has been suggested that this is due to the complex pathophysiology of TBI, which may possibly be addressed by a combination of therapeutic interventions. In this article, we have reviewed combinations of different pharmacologic treatments, combinations of non-pharmacologic interventions, and combined pharmacologic and non-pharmacologic interventions for TBI. Both preclinical and clinical studies have been included. While promising results have been found in animal models, clinical trials of combination therapies have not yet shown clear benefit. This may possibly be due to their application without consideration of the evolving pathophysiology of TBI. Improvements of this paradigm may come from novel interventions guided by multimodal neuromonitoring and multimodal imaging techniques, as well as the application of multi-targeted non-pharmacologic and endogenous therapies. There also needs to be a greater representation of female subjects in preclinical and clinical studies.
Collapse
Affiliation(s)
- Daniel G. Lynch
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
| | - Raj K. Narayan
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Neurosurgery, St. Francis Hospital, Roslyn, NY 11576, USA
| | - Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
- Department of Neurosurgery, Northwell Health, Manhasset, NY 11030, USA
- Correspondence:
| |
Collapse
|
11
|
N-acetylcysteine aggravates seizures while improving depressive-like and cognitive impairment comorbidities in the WAG/Rij rat model of absence epilepsy. Mol Neurobiol 2022; 59:2702-2714. [PMID: 35167014 DOI: 10.1007/s12035-021-02720-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022]
Abstract
N-acetylcysteine (NAC) is an antioxidant with some demonstrated efficacy in a range of neuropsychiatric disorders. NAC has shown anticonvulsant effects in animal models. NAC effects on absence seizures are still not uncovered, and considering its clinical use as a mucolytic in patients with lung diseases, people with epilepsy are also likely to be exposed to the drug. Therefore, we aimed to study the effects of NAC on absence seizures in the WAG/Rij rat model of absence epilepsy with neuropsychiatric comorbidities. The effects of NAC chronic treatment in WAG/Rij rats were evaluated on: absence seizures at 15 and 30 days by EEG recordings and animal behaviour at 30 days on neuropsychiatric comorbidities. Furthermore, the mechanism of action of NAC was evaluated by analysing brain expression levels of some possible key targets: the excitatory amino acid transporter 2, cystine-glutamate antiporter, metabotropic glutamate receptor 2, the mechanistic target of rapamycin and p70S6K as well as levels of total glutathione. Our results demonstrate that in WAG/Rij rats, NAC treatment significantly increased the number and duration of SWDs, aggravating absence epilepsy while ameliorating neuropsychiatric comorbidities. NAC treatment was linked to an increase in brain mGlu2 receptor expression with this being likely responsible for the observed absence seizure-promoting effects. In conclusion, while confirming the positive effects on animal behaviour induced by NAC also in epileptic animals, we report the aggravating effects of NAC on absence seizures which could have some serious consequences for epilepsy patients with the possible wider use of NAC in clinical therapeutics.
Collapse
|
12
|
Davis CK, Vemuganti R. Antioxidant therapies in traumatic brain injury. Neurochem Int 2022; 152:105255. [PMID: 34915062 PMCID: PMC11884749 DOI: 10.1016/j.neuint.2021.105255] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022]
Abstract
Oxidative stress plays a crucial role in traumatic brain injury (TBI) pathogenesis. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) formed in excess after TBI synergistically contribute to secondary brain damage together with lipid peroxidation products (reactive aldehydes) and inflammatory mediators. Furthermore, oxidative stress, endoplasmic reticulum stress and inflammation potentiate each other. Following TBI, excessive oxidative stress overloads the endogenous cellular antioxidant system leading to cell death. To combat oxidative stress, several antioxidant therapies were tested in preclinical animal models of TBI. These include free radical scavengers, activators of antioxidant systems, Inhibitors of free radical generating enzymes and antioxidant enzymes. Many of these therapies showed promising outcomes including reduced edema, blood-brain barrier (BBB) protection, smaller contusion volume, and less inflammation. In addition, many antioxidant therapies also promoted better sensory, motor, and cognitive functional recovery after TBI. Overall, preventing oxidative stress is a viable therapeutic option to minimize the secondary damage and to improve the quality of life after TBI.
Collapse
Affiliation(s)
- Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton Memorial Veteran Administration Hospital, Madison, WI, USA.
| |
Collapse
|
13
|
Velayudhan PS, Schwab N, Hazrati LN, Wheeler AL. Temporal patterns of microglial activation in white matter following experimental mild traumatic brain injury: a systematic literature review. Acta Neuropathol Commun 2021; 9:197. [PMID: 34924026 PMCID: PMC8684664 DOI: 10.1186/s40478-021-01297-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022] Open
Abstract
Mild traumatic brain injuries (mTBIs) are a prevalent form of injury that can result in persistent neurological impairments. Microglial activation has become increasingly recognized as a key process regulating the pathology of white matter in a wide range of brain injury and disease contexts. As white matter damage is known to be a major contributor to the impairments that follow mTBI, microglia have rightfully become a common target of investigation for the development of mTBI therapies and biomarkers. Recent work has demonstrated that the efficacy of microglial manipulation as a therapeutic intervention following injury or disease is highly time-sensitive, emphasizing the importance of advancing our understanding of the dynamics of post-mTBI microglial activation from onset to resolution. Current reporting of microglial activation in experimental studies of mTBI is non-standardized, which has limited our ability to identify concrete patterns of post-mTBI microglial activation over time. In this review, we examine preclinical studies of mTBI that report on microglial activation in white matter regions to summarize our current understanding of these patterns. Specifically, we summarize timecourses of post-mTBI microglial activation in white matter regions of the brain, identify factors that influence this activation, examine the temporal relationship between microglial activation and other post-mTBI assessments, and compare the relative sensitivities of various methods for detecting microglial activation. While the lack of replicated experimental conditions has limited the extent of conclusions that can confidently be drawn, we find that microglia are activated over a wide range of timecourses following mTBI and that microglial activation is a long-lasting outcome of mTBI that may resolve after most typical post-mTBI assessments, with the exception of those measuring oligodendrocyte lineage cell integrity. We identify several understudied parameters of post-mTBI microglial activation in white matter, such as the inclusion of female subjects. This review summarizes our current understanding of the progression of microglial activation in white matter structures following experimental mTBI and offers suggestions for important future research directions.
Collapse
Affiliation(s)
- Prashanth S Velayudhan
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Nicole Schwab
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Lili-Naz Hazrati
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anne L Wheeler
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
14
|
Kyyriäinen J, Kajevu N, Bañuelos I, Lara L, Lipponen A, Balosso S, Hämäläinen E, Das Gupta S, Puhakka N, Natunen T, Ravizza T, Vezzani A, Hiltunen M, Pitkänen A. Targeting Oxidative Stress with Antioxidant Duotherapy after Experimental Traumatic Brain Injury. Int J Mol Sci 2021; 22:10555. [PMID: 34638900 PMCID: PMC8508668 DOI: 10.3390/ijms221910555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 01/23/2023] Open
Abstract
We assessed the effect of antioxidant therapy using the Food and Drug Administration-approved respiratory drug N-acetylcysteine (NAC) or sulforaphane (SFN) as monotherapies or duotherapy in vitro in neuron-BV2 microglial co-cultures and validated the results in a lateral fluid-percussion model of TBI in rats. As in vitro measures, we assessed neuronal viability by microtubule-associated-protein 2 immunostaining, neuroinflammation by monitoring tumor necrosis factor (TNF) levels, and neurotoxicity by measuring nitrite levels. In vitro, duotherapy with NAC and SFN reduced nitrite levels to 40% (p < 0.001) and neuroinflammation to -29% (p < 0.001) compared with untreated culture. The treatment also improved neuronal viability up to 72% of that in a positive control (p < 0.001). The effect of NAC was negligible, however, compared with SFN. In vivo, antioxidant duotherapy slightly improved performance in the beam walking test. Interestingly, duotherapy treatment decreased the plasma interleukin-6 and TNF levels in sham-operated controls (p < 0.05). After TBI, no treatment effect on HMGB1 or plasma cytokine levels was detected. Also, no treatment effects on the composite neuroscore or cortical lesion area were detected. The robust favorable effect of duotherapy on neuroprotection, neuroinflammation, and oxidative stress in neuron-BV2 microglial co-cultures translated to modest favorable in vivo effects in a severe TBI model.
Collapse
Affiliation(s)
- Jenni Kyyriäinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| | - Natallie Kajevu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| | - Ivette Bañuelos
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| | - Leonardo Lara
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| | - Anssi Lipponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
- Department of Health Security, Finnish Institute for Health and Welfare, FI-70701 Kuopio, Finland
| | - Silvia Balosso
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milano, Italy; (S.B.); (T.R.); (A.V.)
| | - Elina Hämäläinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| | - Shalini Das Gupta
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| | - Noora Puhakka
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (T.N.); (M.H.)
| | - Teresa Ravizza
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milano, Italy; (S.B.); (T.R.); (A.V.)
| | - Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milano, Italy; (S.B.); (T.R.); (A.V.)
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (T.N.); (M.H.)
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (N.K.); (I.B.); (L.L.); (A.L.); (E.H.); (S.D.G.); (N.P.)
| |
Collapse
|
15
|
Whitney K, Nikulina E, Rahman SN, Alexis A, Bergold PJ. Delayed dosing of minocycline plus N-acetylcysteine reduces neurodegeneration in distal brain regions and restores spatial memory after experimental traumatic brain injury. Exp Neurol 2021; 345:113816. [PMID: 34310944 DOI: 10.1016/j.expneurol.2021.113816] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
Multiple drugs to treat traumatic brain injury (TBI) have failed clinical trials. Most drugs lose efficacy as the time interval increases between injury and treatment onset. Insufficient therapeutic time window is a major reason underlying failure in clinical trials. Few drugs have been developed with therapeutic time windows sufficiently long enough to treat TBI because little is known about which brain functions can be targeted if therapy is delayed hours to days after injury. We identified multiple injury parameters that are improved by first initiating treatment with the drug combination minocycline (MINO) plus N-acetylcysteine (NAC) at 72 h after injury (MN72) in a mouse closed head injury (CHI) experimental TBI model. CHI produces spatial memory deficits resulting in impaired performance on Barnes maze, hippocampal neuronal loss, and bilateral damage to hippocampal neurons, dendrites, spines and synapses. MN72 treatment restores Barnes maze acquisition and retention, protects against hippocampal neuronal loss, limits damage to dendrites, spines and synapses, and accelerates recovery of microtubule associated protein 2 (MAP2) expression, a key protein in maintaining proper dendritic architecture and synapse density. These data show that in addition to the structural integrity of the dendritic arbor, spine and synapse density can be successfully targeted with drugs first dosed days after injury. Retention of substantial drug efficacy even when first dosed 72 h after injury makes MINO plus NAC a promising candidate to treat clinical TBI.
Collapse
Affiliation(s)
- Kristen Whitney
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Elena Nikulina
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Syed N Rahman
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Alisia Alexis
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Peter J Bergold
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America.
| |
Collapse
|
16
|
Ho MH, Yen CH, Hsieh TH, Kao TJ, Chiu JY, Chiang YH, Hoffer BJ, Chang WC, Chou SY. CCL5 via GPX1 activation protects hippocampal memory function after mild traumatic brain injury. Redox Biol 2021; 46:102067. [PMID: 34315111 PMCID: PMC8327355 DOI: 10.1016/j.redox.2021.102067] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/12/2021] [Accepted: 07/08/2021] [Indexed: 12/23/2022] Open
Abstract
Traumatic brain injury (TBI) is a prevalent head injury worldwide which increases the risk of neurodegenerative diseases. Increased reactive oxygen species (ROS) and inflammatory chemokines after TBI induces secondary effects which damage neurons. Targeting NADPH oxidase or increasing redox systems are ways to reduce ROS and damage. Earlier studies show that C–C motif chemokine ligand 5 (CCL5) has neurotrophic functions such as promoting neurite outgrowth as well as reducing apoptosis. Although CCL5 levels in blood are associated with severity in TBI patients, the function of CCL5 after brain injury is unclear. In the current study, we induced mild brain injury in C57BL/6 (wildtype, WT) mice and CCL5 knockout (CCL5-KO) mice using a weight-drop model. Cognitive and memory functions in mice were analyzed by Novel-object-recognition and Barnes Maze tests. The memory performance of both WT and KO mice were impaired after mild injury. Cognition and memory function in WT mice quickly recovered after 7 days but recovery took more than 14 days in CCL5-KO mice. FJC, NeuN and Hypoxyprobe staining revealed large numbers of neurons damaged by oxidative stress in CCL5-KO mice after mTBI. NADPH oxidase activity show increased ROS generation together with reduced glutathione peroxidase-1 (GPX1) and glutathione (GSH) activity in CCL5-KO mice; this was opposite to that seen in WT mice. CCL5 increased GPX1 expression and reduced intracellular ROS levels which subsequently increased cell survival both in primary neuron cultures and in an overexpression model using SHSY5Y cell. Memory impairment in CCL5-KO mice induced by TBI could be rescued by i.p. injection of the GSH precursor – N-acetylcysteine (NAC) or intranasal delivery of recombinant CCL5 into mice after injury. We conclude that CCL5 is an important molecule for GPX1 antioxidant activation during post-injury day 1–3, and protects hippocampal neurons from ROS as well as improves memory function after trauma.
Collapse
Affiliation(s)
- Man-Hau Ho
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research, Taiwan; Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan; Department of Biological Science and Technology, National Pingtung University of Science and Technology, Neipu, Pingtung, 91201, Taiwan
| | - Chia-Hung Yen
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Neipu, Pingtung, 91201, Taiwan
| | - Tsung-Hsun Hsieh
- School of Physical Therapy and Graduate Institute of Rehabilitation Science, Chang Gung University, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Tzu-Jen Kao
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research, Taiwan; Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jing-Yuan Chiu
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yung-Hsiao Chiang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, 11031, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei, 11031, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei, 11031, Taiwan
| | - Barry J Hoffer
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research, Taiwan; Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei, 11031, Taiwan; Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Scientist Emeritus, National Institutes of Health, USA
| | - Wen-Chang Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Szu-Yi Chou
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research, Taiwan; Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
17
|
Hiskens MI, Vella RK, Schneiders AG, Fenning AS. Minocycline improves cognition and molecular measures of inflammation and neurodegeneration following repetitive mTBI. Brain Inj 2021; 35:831-841. [PMID: 33818227 DOI: 10.1080/02699052.2021.1909139] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/01/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To compare the neuroprotective effects of minocycline treatment in a murine model of mTBI on measures of spatial learning and memory, neuroinflammation, excitotoxicity, and neurodegeneration. DESIGN Adult male C57BL/6 J mice were randomly assigned into vehicle control, vehicle with repetitive mTBI, minocycline without mTBI, or minocycline with repetitive mTBI groups. METHODS A validated mouse model of repetitive impact-induced rotational acceleration was used to deliver 15 mTBIs across 23 days. Cognition was assessed via Morris water maze (MWM) testing, and mRNA analysis investigated MAPT, GFAP, AIF1, GRIA1, TARDBP, TNF, and NEFL genes. Assessment was undertaken 48 h and 3 months following final mTBI. RESULTS In the chronic phase of recovery, MWM testing revealed impairment in the vehicle mTBI group compared to unimpacted controls (p < .01) that was not present in the minocycline mTBI group, indicating chronic neuroprotection. mRNA analysis revealed AIF1 elevation in the acute cortex (p < .01) and chronic hippocampus (p < .01) of the vehicle mTBI group, with minocycline treatment leading to improved markers of microglial activation and inflammation in the chronic stage of recovery. CONCLUSIONS These data suggest that minocycline treatment alleviated some mTBI pathophysiology and clinical features at chronic time-points.
Collapse
Affiliation(s)
- Matthew I Hiskens
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton 4701, Australia
| | - Rebecca K Vella
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton 4701, Australia
| | - Anthony G Schneiders
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton 4701, Australia
| | - Andrew S Fenning
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton 4701, Australia
| |
Collapse
|
18
|
Tras B, Eser Faki H, Ozdemir Kutahya Z, Bahcivan E, Dik B, Bozkurt B, Uney K. Treatment and protective effects of metalloproteinase inhibitors alone and in combination with N-Acetyl cysteine plus vitamin E in rats exposed to aflatoxin B 1. Toxicon 2021; 194:79-85. [PMID: 33617885 DOI: 10.1016/j.toxicon.2021.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 11/19/2022]
Abstract
This study was conducted to investigate the effects of matrix metalloproteinase (MMP) inhibitors dexamethasone and minocycline administrations -both single and in combination with N-acetylcysteine (NAC) and vitamin E-on the tissue distribution and lethal dose (LD)50 of aflatoxin (AF)B1 in rats. We performed this study on male Wistar rats (8-10 weeks) in two phases. In the first phase, rats were administered dexamethasone (5 and 20 mg/kg) and minocycline (45 and 90 mg/kg), both as single treatments and in combination with NAC (200 mg/kg) and vitamin E (600 mg/kg); these treatments followed AFB1 administration (2 mg/kg). In the second phase, the therapeutic effect value (TEV) was calculated to determine the treatment effect on the LD50 level of AFB1. The tissue affinity of AFB1 from high to low was liver, kidney, intestine, brain, heart, spleen, lung, testis, and vitreous humor, respectively. Dexamethasone at the 20 mg/kg dose significantly reduced AFB1 concentrations in the plasma and the other tissues, except for the vitreous humor. The effects of minocycline on the plasma and tissue concentrations of AFB1 varied by dose and tissue. The combinations of dexamethasone or minocycline with NAC and vitamin E increased the AFB1 concentrations in the plasma and all tissues, except for vitreous humor and liver. In male rats, the LD50 value of AFB1 was 11.86 mg/kg. The TEV of dexamethasone (20 mg/kg) was calculated to be 1.5. Dexamethasone can be administered in repeated doses at ≥20 mg/kg to increase survival in AFB1 poisoning.
Collapse
Affiliation(s)
- Bunyamin Tras
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey
| | - Hatice Eser Faki
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey
| | - Zeynep Ozdemir Kutahya
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Cukurova, 01930, Adana, Turkey
| | - Emre Bahcivan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Kafkas, 36000, Kars, Turkey
| | - Burak Dik
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey.
| | - Banu Bozkurt
- Department of Ophthalmology, Faculty of Medicine, University of Selcuk, 42031, Konya, Turkey
| | - Kamil Uney
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031, Konya, Turkey
| |
Collapse
|
19
|
Jia Y, Wang G, Ye Y, Kang E, Chen H, Guo Z, He X. Niche Cells Crosstalk In Neuroinflammation After Traumatic Brain Injury. Int J Biol Sci 2021; 17:368-378. [PMID: 33390856 PMCID: PMC7757042 DOI: 10.7150/ijbs.52169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is recognized as the disease with high morbidity and disability around world in spite of the work ongoing in neural protection. Due to heterogeneity among the patients, it's still hard to acquire satisfying achievements in clinic. Neuroinflammation, which exists since primary injury occurs, with elusive duality, appear to be of significance from recovery of injury to neurogenesis. In recent years, studied have revealed that communication in neurogenic niche is more than “cell to cell” communication, and study on NSCs represent it as central role in the progress of neural regeneration. Hence, the neuroinflammation-affecting crosstalk after TBI, and clarifying definitive role of NSCs in the course of regeneration is a promising subject for researchers, for its great potential in overcoming the frustrating status quo in clinic, promoting welfare of TBI patient.
Collapse
Affiliation(s)
- Yibin Jia
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Guanyi Wang
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Yuqing Ye
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China.,Department of Neurosurgery, PLA 163rd Hospital (Second Affiliated Hospital of Hunan Normal University), Changsha 410000, China
| | - Enming Kang
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Huijun Chen
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Zishuo Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China
| | - Xiaosheng He
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| |
Collapse
|
20
|
Roberson SW, Patel MB, Dabrowski W, Ely EW, Pakulski C, Kotfis K. Challenges of Delirium Management in Patients with Traumatic Brain Injury: From Pathophysiology to Clinical Practice. Curr Neuropharmacol 2021; 19:1519-1544. [PMID: 33463474 PMCID: PMC8762177 DOI: 10.2174/1570159x19666210119153839] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/12/2020] [Accepted: 01/13/2021] [Indexed: 11/22/2022] Open
Abstract
Traumatic brain injury (TBI) can initiate a very complex disease of the central nervous system (CNS), starting with the primary pathology of the inciting trauma and subsequent inflammatory and CNS tissue response. Delirium has long been regarded as an almost inevitable consequence of moderate to severe TBI, but more recently has been recognized as an organ dysfunction syndrome with potentially mitigating interventions. The diagnosis of delirium is independently associated with prolonged hospitalization, increased mortality and worse cognitive outcome across critically ill populations. Investigation of the unique problems and management challenges of TBI patients is needed to reduce the burden of delirium in this population. In this narrative review, possible etiologic mechanisms behind post-traumatic delirium are discussed, including primary injury to structures mediating arousal and attention and secondary injury due to progressive inflammatory destruction of the brain parenchyma. Other potential etiologic contributors include dysregulation of neurotransmission due to intravenous sedatives, seizures, organ failure, sleep cycle disruption or other delirium risk factors. Delirium screening can be accomplished in TBI patients and the presence of delirium portends worse outcomes. There is evidence that multi-component care bundles including an analgesia-prioritized sedation algorithm, regular spontaneous awakening and breathing trials, protocolized delirium assessment, early mobility and family engagement can reduce the burden of ICU delirium. The aim of this review is to summarize the approach to delirium in TBI patients with an emphasis on pathogenesis and management. Emerging CNS-active drug therapies that show promise in preclinical studies are highlighted.
Collapse
Affiliation(s)
| | | | | | | | | | - Katarzyna Kotfis
- Address correspondence to this author at the Department of Anesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University in Szczecin, Poland; E-mail:
| |
Collapse
|
21
|
Combination Therapy of Gabapentin and N-Acetylcysteine Against Posttraumatic Epilepsy in Rats. Neurochem Res 2020; 45:1802-1812. [DOI: 10.1007/s11064-020-03042-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/30/2020] [Accepted: 04/28/2020] [Indexed: 01/14/2023]
|
22
|
Chakraborty S, Tripathi SJ, Srikumar B, Raju T, Shankaranarayana Rao B. N-acetyl cysteine ameliorates depression-induced cognitive deficits by restoring the volumes of hippocampal subfields and associated neurochemical changes. Neurochem Int 2020; 132:104605. [DOI: 10.1016/j.neuint.2019.104605] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/11/2019] [Accepted: 11/15/2019] [Indexed: 12/14/2022]
|
23
|
Jha KA, Pentecost M, Lenin R, Gentry J, Klaic L, Del Mar N, Reiner A, Yang CH, Pfeffer LM, Sohl N, Gangaraju R. TSG-6 in conditioned media from adipose mesenchymal stem cells protects against visual deficits in mild traumatic brain injury model through neurovascular modulation. Stem Cell Res Ther 2019; 10:318. [PMID: 31690344 PMCID: PMC6833275 DOI: 10.1186/s13287-019-1436-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/21/2019] [Accepted: 09/30/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Retinal inflammation affecting the neurovascular unit may play a role in the development of visual deficits following mild traumatic brain injury (mTBI). We have shown that concentrated conditioned media from adipose tissue-derived mesenchymal stem cells (ASC-CCM) can limit retinal damage from blast injury and improve visual function. In this study, we addressed the hypothesis that TNFα-stimulated gene-6 (TSG-6), an anti-inflammatory protein released by mesenchymal cells, mediates the observed therapeutic potential of ASCs via neurovascular modulation. METHODS About 12-week-old C57Bl/6 mice were subjected to 50-psi air pulse on the left side of the head overlying the forebrain resulting in an mTBI. Age-matched sham blast mice served as control. About 1 μl of ASC-CCM (siControl-ASC-CCM) or TSG-6 knockdown ASC-CCM (siTSG-6-ASC-CCM) was delivered intravitreally into both eyes. One month following injection, the ocular function was assessed followed by molecular and immunohistological analysis. In vitro, mouse microglial cells were used to evaluate the anti-inflammatory effect of ASC-CCM. Efficacy of ASC-CCM in normalizing retinal vascular permeability was assessed using trans-endothelial resistance (TER) and VE-cadherin expression in the presence of TNFα (1 ng/ml). RESULTS We show that intravitreal injection of ASC-CCM (siControl-ASC-CCM) but not the TSG-6 knockdown ASC-CCM (siTSG-6-ASC-CCM) mitigates the loss of visual acuity and contrast sensitivity, retinal expression of genes associated with microglial and endothelial activation, and retinal GFAP immunoreactivity at 4 weeks after blast injury. In vitro, siControl-ASC-CCM but not the siTSG-6-ASC-CCM not only suppressed microglial activation and STAT3 phosphorylation but also protected against TNFα-induced endothelial permeability as measured by transendothelial electrical resistance and decreased STAT3 phosphorylation. CONCLUSIONS Our findings suggest that ASCs respond to an inflammatory milieu by secreting higher levels of TSG-6 that mediates the resolution of the inflammatory cascade on multiple cell types and correlates with the therapeutic potency of the ASC-CCM. These results expand our understanding of innate mesenchymal cell function and confirm the importance of considering methods to increase the production of key analytes such as TSG-6 if mesenchymal stem cell secretome-derived biologics are to be developed as a treatment solution against the traumatic effects of blast injuries and other neurovascular inflammatory conditions of the retina.
Collapse
Affiliation(s)
- Kumar Abhiram Jha
- Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, 930 Madison Ave, Suite#768, Memphis, TN, 38163, USA
| | - Mickey Pentecost
- Cell Care Therapeutics, Inc., Los Angeles, CA, USA.,Present Address: Pathways to Stem Cell Science, Monrovia, CA, USA
| | - Raji Lenin
- Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, 930 Madison Ave, Suite#768, Memphis, TN, 38163, USA
| | - Jordy Gentry
- Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, 930 Madison Ave, Suite#768, Memphis, TN, 38163, USA
| | - Lada Klaic
- Cell Care Therapeutics, Inc., Los Angeles, CA, USA
| | - Nobel Del Mar
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, 855 Monroe Avenue, Suite#515, Memphis, TN, 38163, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, 855 Monroe Avenue, Suite#515, Memphis, TN, 38163, USA
| | - Chuan He Yang
- Department of Pathology, University of Tennessee Health Science Center, College of Medicine, 19 South Manassas Street, Suite#214, Memphis, TN, 38163, USA
| | - Lawrence M Pfeffer
- Department of Pathology, University of Tennessee Health Science Center, College of Medicine, 19 South Manassas Street, Suite#214, Memphis, TN, 38163, USA
| | - Nicolas Sohl
- Cell Care Therapeutics, Inc., Los Angeles, CA, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, 930 Madison Ave, Suite#768, Memphis, TN, 38163, USA. .,Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, 855 Monroe Avenue, Suite#515, Memphis, TN, 38163, USA.
| |
Collapse
|
24
|
Positive allosteric modulation of the α7 nicotinic acetylcholine receptor as a treatment for cognitive deficits after traumatic brain injury. PLoS One 2019; 14:e0223180. [PMID: 31581202 PMCID: PMC6776323 DOI: 10.1371/journal.pone.0223180] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/16/2019] [Indexed: 11/19/2022] Open
Abstract
Cognitive impairments are a common consequence of traumatic brain injury (TBI). The hippocampus is a subcortical structure that plays a key role in the formation of declarative memories and is highly vulnerable to TBI. The α7 nicotinic acetylcholine receptor (nAChR) is highly expressed in the hippocampus and reduced expression and function of this receptor are linked with cognitive impairments in Alzheimer's disease and schizophrenia. Positive allosteric modulation of α7 nAChRs with AVL-3288 enhances receptor currents and improves cognitive functioning in naïve animals and healthy human subjects. Therefore, we hypothesized that targeting the α7 nAChR with the positive allosteric modulator AVL-3288 would enhance cognitive functioning in the chronic recovery period of TBI. To test this hypothesis, adult male Sprague Dawley rats received moderate parasagittal fluid-percussion brain injury or sham surgery. At 3 months after recovery, animals were treated with vehicle or AVL-3288 at 30 min prior to cue and contextual fear conditioning and the water maze task. Treatment of TBI animals with AVL-3288 rescued learning and memory deficits in water maze retention and working memory. AVL-3288 treatment also improved cue and contextual fear memory when tested at 24 hr and 1 month after training, when TBI animals were treated acutely just during fear conditioning at 3 months post-TBI. Hippocampal atrophy but not cortical atrophy was reduced with AVL-3288 treatment in the chronic recovery phase of TBI. AVL-3288 application to acute hippocampal slices from animals at 3 months after TBI rescued basal synaptic transmission deficits and long-term potentiation (LTP) in area CA1. Our results demonstrate that AVL-3288 improves hippocampal synaptic plasticity, and learning and memory performance after TBI in the chronic recovery period. Enhancing cholinergic transmission through positive allosteric modulation of the α7 nAChR may be a novel therapeutic to improve cognition after TBI.
Collapse
|
25
|
Machado CA, Silva ACSE, de Miranda AS, Cordeiro TME, Ferreira RN, de Souza LC, Teixeira AL, de Miranda AS. Immune-Based Therapies for Traumatic Brain Injury: Insights from Pre-Clinical Studies. Curr Med Chem 2019; 27:5374-5402. [PMID: 31291871 DOI: 10.2174/0929867326666190710173234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/24/2019] [Accepted: 05/22/2019] [Indexed: 12/18/2022]
Abstract
Traumatic Brain Injury (TBI) is a major public health problem. It is the leading cause of death and disability, especially among children and young adults. The neurobiology basis underlying TBI pathophysiology remains to be fully revealed. Over the past years, emerging evidence has supported the hypothesis that TBI is an inflammatory based condition, paving the way for the development of potential therapeutic targets. There is no treatment capable to prevent or minimize TBIassociated outcomes. Therefore, the search for effective therapies is a priority goal. In this context, animal models have become valuable tools to study molecular and cellular mechanisms involved in TBI pathogenesis as well as novel treatments. Herein, we discuss therapeutic strategies to treat TBI focused on immunomodulatory and/or anti-inflammatory approaches in the pre-clinical setting.
Collapse
Affiliation(s)
- Caroline Amaral Machado
- Laboratorio de Neurobiologia, Departamento de Morfologia, Instituto de Ciencias Biologicas, UFMG, Brazil
| | - Ana Cristina Simões E Silva
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Amanda Silva de Miranda
- Departamento de Quimica, Instituto de Ciencias Exatas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Thiago Macedo E Cordeiro
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Rodrigo Novaes Ferreira
- Laboratorio de Neurobiologia, Departamento de Morfologia, Instituto de Ciencias Biologicas, UFMG, Brazil
| | - Leonardo Cruz de Souza
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center, Houston, United States
| | - Aline Silva de Miranda
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
26
|
Anastasio TJ. Exploring the Correlation between the Cognitive Benefits of Drug Combinations in a Clinical Database and the Efficacies of the Same Drug Combinations Predicted from a Computational Model. J Alzheimers Dis 2019; 70:287-302. [PMID: 31177222 PMCID: PMC6700640 DOI: 10.3233/jad-190144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Identification of drug combinations that could be effective in Alzheimer’s disease treatment is made difficult by the sheer number of possible combinations. This analysis identifies as potentially therapeutic those drug combinations that rank highest when their efficacy is determined jointly from two independent data sources. Estimates of the efficacy of the same drug combinations were derived from a clinical dataset on cognitively impaired elderly participants and from pre-clinical data, in the form of a computational model of neuroinflammation. Linear regression was used to show that the two sets of estimates were correlated, and to rule out confounds. The ten highest ranking, jointly determined drug combinations most frequently consisted of COX2 inhibitors and aspirin, along with various antihypertensive medications. Ten combinations of from five to nine drugs, and the three-drug combination of a COX2 inhibitor, aspirin, and a calcium-channel blocker, are discussed as candidates for consideration in future pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Thomas J Anastasio
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The objective of this article is to highlight the potential role of the galantamine-memantine combination as a novel antioxidant treatment for schizophrenia. RECENT FINDINGS In addition to the well-known mechanisms of action of galantamine and memantine, these medications also have antioxidant activity. Furthermore, an interplay exists between oxidative stress, inflammation (redox-inflammatory hypothesis), and kynurenine pathway metabolites. Also, there is an interaction between brain-derived neurotrophic factor and oxidative stress in schizophrenia. Oxidative stress may be associated with positive, cognitive, and negative symptoms and impairments in white matter integrity in schizophrenia. The antipsychotic-galantamine-memantine combination may provide a novel strategy in schizophrenia to treat positive, cognitive, and negative symptoms. SUMMARY A "single antioxidant" may be inadequate to counteract the complex cascade of oxidative stress. The galantamine-memantine combination as "double antioxidants" is promising. Hence, randomized controlled trials are warranted with the antipsychotic-galantamine-memantine combination with oxidative stress and antioxidant biomarkers in schizophrenia.
Collapse
|
28
|
Rubenstein R, Sharma DR, Chang B, Oumata N, Cam M, Vaucelle L, Lindberg MF, Chiu A, Wisniewski T, Wang KKW, Meijer L. Novel Mouse Tauopathy Model for Repetitive Mild Traumatic Brain Injury: Evaluation of Long-Term Effects on Cognition and Biomarker Levels After Therapeutic Inhibition of Tau Phosphorylation. Front Neurol 2019; 10:124. [PMID: 30915013 PMCID: PMC6421297 DOI: 10.3389/fneur.2019.00124] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is a risk factor for a group of neurodegenerative diseases termed tauopathies, which includes Alzheimer's disease and chronic traumatic encephalopathy (CTE). Although TBI is stratified by impact severity as either mild (m), moderate or severe, mTBI is the most common and the most difficult to diagnose. Tauopathies are pathologically related by the accumulation of hyperphosphorylated tau (P-tau) and increased total tau (T-tau). Here we describe: (i) a novel human tau-expressing transgenic mouse model, TghTau/PS1, to study repetitive mild closed head injury (rmCHI), (ii) quantitative comparison of T-tau and P-tau from brain and plasma in TghTau/PS1 mice over a 12 month period following rmCHI (and sham), (iii) the usefulness of P-tau as an early- and late-stage blood-based biochemical biomarker for rmCHI, (iii) the influence of kinase-targeted therapeutic intervention on rmCHI-associated cognitive deficits using a combination of lithium chloride (LiCl) and R-roscovitine (ros), and (iv) correlation of behavioral and cognitive changes with concentrations of the brain and blood-based T-tau and P-tau. Compared to sham-treated mice, behavior changes and cognitive deficits of rmCHI-treated TghTau/PS1 mice correlated with increases in both cortex and plasma T-tau and P-tau levels over 12 months. In addition, T-tau, but more predominantly P-tau, levels were significantly reduced in the cortex and plasma by LiCl + ros approaching the biomarker levels in sham and drug-treated sham mice (the drugs had only modest effects on the T-tau and P-tau levels in sham mice) throughout the 12 month study period. Furthermore, although we also observed a reversal of the abnormal behavior and cognitive deficits in the drug-treated rmCHI mice (compared to the untreated rmCHI mice) throughout the time course, these drug-treated effects were most pronounced up until 10 and 12 months where the abnormal behavior and cognition deficits began to gradually increase. These studies describe: (a) a translational relevant animal model for TBI-linked tauopathies, and (b) utilization of T-tau and P-tau as rmCHI biomarkers in plasma to monitor novel therapeutic strategies and treatment regimens for these neurodegenerative diseases.
Collapse
Affiliation(s)
- Richard Rubenstein
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Deep R Sharma
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Binggong Chang
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Nassima Oumata
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | - Morgane Cam
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | - Lise Vaucelle
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | | | - Allen Chiu
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Departments of Neurology, Pathology and Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, Gainesville, FL, United States
| | - Laurent Meijer
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| |
Collapse
|
29
|
Koola MM. Antipsychotic-minocycline-acetylcysteine combination for positive, cognitive, and negative symptoms of schizophrenia. Asian J Psychiatr 2019; 40:100-102. [PMID: 30776665 DOI: 10.1016/j.ajp.2019.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/26/2022]
Abstract
Preclinical evidence shows that the minocycline and N-acetylcysteine (NAC) combination synergistically improved cognition. Meta-analyses of randomized controlled trials (RCTs) with minocycline and NAC have shown some efficacy signal for positive, cognitive, and negative symptoms of schizophrenia. Hence, the combination may be more effective than either medication alone. The objective of this article is to highlight the potential role of the minocycline-NAC combination for the treatment of schizophrenia. The antipsychotic-minocycline-NAC combination is promising and has the potential to concurrently treat positive, cognitive, and primary negative symptoms. RCTs are warranted with the minocycline-NAC combination to address the unmet clinical need in schizophrenia.
Collapse
Affiliation(s)
- Maju Mathew Koola
- Department of Psychiatry and Behavioral Sciences, George Washington University School of Medicine and Health Sciences, 2300 I St NW, 20037, Washington, DC, USA.
| |
Collapse
|
30
|
Park H, You N, Lee J, Suh M. Longitudinal study of hemodynamics and dendritic membrane potential changes in the mouse cortex following a soft cranial window installation. NEUROPHOTONICS 2019; 6:015006. [PMID: 30820438 PMCID: PMC6387987 DOI: 10.1117/1.nph.6.1.015006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/25/2019] [Indexed: 05/21/2023]
Abstract
The soft cranial window using polydimethylsiloxane allows direct multiple access to neural tissue during long-term monitoring. However, the chronic effects of soft window installation on the brain have not been fully studied. Here, we investigate the long-term effects of soft window installation on sensory-evoked cerebral hemodynamics and neuronal activity. We monitored the brain tissue immunocytohistology for 6 weeks postinstallation. Heightened reactive astrocytic and microglia levels were found at 2 weeks postinstallation. By 6 weeks postinstallation, mice had expression levels similar to those of normal animals. We recorded sensory-evoked hemodynamics of the barrel cortex and LFP during whisker stimulation at these time points. Animals at 6 weeks postinstallation showed stronger hemodynamic responses and focalized barrel mapping than 2-week postoperative mice. LFP recordings of 6-week postoperative mice also showed higher neural activity at the barrel column corresponding to the stimulated whisker. Furthermore, the expression level of interleukin- 1 β was highly upregulated at 2 weeks postinstallation. When we treated animals postoperatively with minocycline plus N-acetylcystein, a drug-suppressing inflammatory cytokine, these animals did not show declined hemodynamic responses and neuronal activities. This result suggests that neuroinflammation following soft window installation may alter hemodynamic and neuronal responses upon sensory stimulation.
Collapse
Affiliation(s)
- Hyejin Park
- Institute for Basic Science, Center for Neuroscience Imaging Research, Suwon, Republic of Korea
- Sungkyunkwan University, Department of Biological Sciences, Suwon, Republic of Korea
- Sungkyunkwan University, Biomedical Institute for Convergence, Suwon, Republic of Korea
| | - Nayeon You
- Institute for Basic Science, Center for Neuroscience Imaging Research, Suwon, Republic of Korea
- Sungkyunkwan University, Department of Biomedical Engineering, Suwon, Republic of Korea
| | - Juheon Lee
- Institute for Basic Science, Center for Neuroscience Imaging Research, Suwon, Republic of Korea
- Sungkyunkwan University, Department of Biomedical Engineering, Suwon, Republic of Korea
| | - Minah Suh
- Institute for Basic Science, Center for Neuroscience Imaging Research, Suwon, Republic of Korea
- Sungkyunkwan University, Biomedical Institute for Convergence, Suwon, Republic of Korea
- Sungkyunkwan University, Department of Biomedical Engineering, Suwon, Republic of Korea
- Sungkyunkwan University, SAHIST, Suwon, Republic of Korea
- Address all correspondence to Minah Suh, E-mail:
| |
Collapse
|
31
|
More J, Galusso N, Veloso P, Montecinos L, Finkelstein JP, Sanchez G, Bull R, Valdés JL, Hidalgo C, Paula-Lima A. N-Acetylcysteine Prevents the Spatial Memory Deficits and the Redox-Dependent RyR2 Decrease Displayed by an Alzheimer's Disease Rat Model. Front Aging Neurosci 2018; 10:399. [PMID: 30574085 PMCID: PMC6291746 DOI: 10.3389/fnagi.2018.00399] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/19/2018] [Indexed: 12/28/2022] Open
Abstract
We have previously reported that primary hippocampal neurons exposed to synaptotoxic amyloid beta oligomers (AβOs), which are likely causative agents of Alzheimer’s disease (AD), exhibit abnormal Ca2+ signals, mitochondrial dysfunction and defective structural plasticity. Additionally, AβOs-exposed neurons exhibit a decrease in the protein content of type-2 ryanodine receptor (RyR2) Ca2+ channels, which exert critical roles in hippocampal synaptic plasticity and spatial memory processes. The antioxidant N-acetylcysteine (NAC) prevents these deleterious effects of AβOs in vitro. The main contribution of the present work is to show that AβOs injections directly into the hippocampus, by engaging oxidation-mediated reversible pathways significantly decreased RyR2 protein content but increased single RyR2 channel activation by Ca2+ and caused considerable spatial memory deficits. AβOs injections into the CA3 hippocampal region impaired rat performance in the Oasis maze spatial memory task, decreased hippocampal glutathione levels and overall content of plasticity-related proteins (c-Fos, Arc, and RyR2) and increased ERK1/2 phosphorylation. In contrast, in hippocampus-derived mitochondria-associated membranes (MAM) AβOs injections increased RyR2 levels. Rats fed with NAC for 3-weeks prior to AβOs injections displayed comparable redox potential, RyR2 and Arc protein contents, similar ERK1/2 phosphorylation and RyR2 single channel activation by Ca2+ as saline-injected (control) rats. NAC-fed rats subsequently injected with AβOs displayed the same behavior in the spatial memory task as control rats. Based on the present in vivo results, we propose that redox-sensitive neuronal RyR2 channels partake in the mechanism underlying AβOs-induced memory disruption in rodents.
Collapse
Affiliation(s)
- Jamileth More
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Nadia Galusso
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
| | - Pablo Veloso
- Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile
| | - Luis Montecinos
- CEMC, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Gina Sanchez
- CEMC, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Pathophysiology Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Ricardo Bull
- Physiology and Biophysics Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - José Luis Valdés
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile.,CEMC, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Physiology and Biophysics Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrea Paula-Lima
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile.,Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile
| |
Collapse
|
32
|
Cory-Slechta DA, Allen JL, Conrad K, Marvin E, Sobolewski M. Developmental exposure to low level ambient ultrafine particle air pollution and cognitive dysfunction. Neurotoxicology 2018; 69:217-231. [PMID: 29247674 PMCID: PMC5999548 DOI: 10.1016/j.neuro.2017.12.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/08/2017] [Accepted: 12/10/2017] [Indexed: 12/27/2022]
Abstract
Developmental exposures to ambient ultrafine particles (UFPs) can produce multiple neuropathological and neurochemical changes that might contribute to persistent alterations in cognitive-type functions. The objective of the current study was to test the hypothesis that developmental UFP exposure produced impairments in learning, memory and impulsive-like behaviors and to determine whether these were selective and thus independent of deficits in other behavioral domains such as motor activity or motivation. Performance on measures of learning (repeated learning), memory (novel object recognition, NOR), impulsive-like behavior (differential reinforcement of low rate (DRL), schedule of reward and delay of reward (DOR)), motor activity (locomotor behavior) and motivation (progressive ratio schedule) were examined in adult mice that had been exposed to concentrated (10-20x) ambient ultrafine particles (CAPS) averaging approximately 45 ug/m3 particle mass concentrations from postnatal day (PND) 4-7 and 10-13 for 4 h/day. Given the number of behavioral tests, animals were tested in different groups. Results showed male-specific alterations in learning and memory functions (repeated learning, NOR and DRL) specifically during transitions in reinforcement contingencies (changes in rules governing behavior) that did not appear to be related to alterations in locomotor function or motivation. Females did not exhibit cognitive-like deficits at these exposure concentrations, but displayed behaviors consistent with altered motivation, including increases in response rates during repeated learning, significantly increased latencies to respond on the delay of reward paradigm, and reductions in the progressive ratio break point. Consistent with our prior findings, male-specific learning and memory-related deficits were seen and occurred even at relatively low level developmental UFP exposures, while females show alterations in motivational behaviors but not final performance. These findings add to the evidence suggesting the need to regulate UFP levels.
Collapse
Affiliation(s)
- D A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States.
| | - J L Allen
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - K Conrad
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - E Marvin
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - M Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| |
Collapse
|
33
|
Vonder Haar C, Ferland JMN, Kaur S, Riparip LK, Rosi S, Winstanley CA. Cocaine self-administration is increased after frontal traumatic brain injury and associated with neuroinflammation. Eur J Neurosci 2018; 50:2134-2145. [PMID: 30118561 DOI: 10.1111/ejn.14123] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) has been linked to the development of numerous psychiatric diseases, including substance use disorder. However, it can be difficult to ascertain from clinical data whether the TBI is cause or consequence of increased addiction vulnerability. Surprisingly few studies have taken advantage of animal models to investigate the causal nature of this relationship. In terms of a plausible neurobiological mechanism through which TBI could magnify the risk of substance dependence, numerous studies indicate that TBI can cause widespread disruption to monoaminergic signaling in striatal regions, and also increases neuroinflammation. In the current study, male Long-Evans rats received either a mild or severe TBI centered over the frontal cortex via controlled cortical impact, and were subsequently trained to self-administer cocaine over 10 6-hour sessions. At the end of the study, markers of striatal dopaminergic function, and levels of inflammatory cytokine levels in the frontal lobes, were assessed via western blot and multiplex ELISA, respectively. There was significantly higher cocaine intake in a subset of animals with either mild or severe TBI. However, many animals within both TBI groups failed to acquire self-administration. Principal components analysis suggested that both dopaminergic and neuroinflammatory proteins were associated with overall cocaine intake, yet only an inflammatory component was associated with acquisition of self-administration, suggesting neuroinflammation may make a more substantial contribution to the likelihood of drug-taking. Should neuroinflammation play a causal role in mediating TBI-induced addiction risk, anti-inflammatory therapy may reduce the likelihood of substance abuse in TBI populations.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, PO Box 6040, 53 Campus Drive, Morgantown, WV, 26505, USA.,Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jacqueline-Marie N Ferland
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Sukhbir Kaur
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lara-Kirstie Riparip
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Susanna Rosi
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Catharine A Winstanley
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
34
|
Sulhan S, Lyon KA, Shapiro LA, Huang JH. Neuroinflammation and blood-brain barrier disruption following traumatic brain injury: Pathophysiology and potential therapeutic targets. J Neurosci Res 2018; 98:19-28. [PMID: 30259550 DOI: 10.1002/jnr.24331] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/11/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
Traumatic Brain Injury (TBI) is the most frequent cause of death and disability in young adults and children in the developed world, occurring in over 1.7 million persons and resulting in 50,000 deaths in the United States alone. The Centers for Disease Control and Prevention estimate that between 3.2 and 5.3 million persons in the United States live with a TBI-related disability, including several neurocognitive disorders and functional limitations. Following the primary mechanical injury in TBI, literature suggests the presence of a delayed secondary injury involving a variety of neuroinflammatory changes. In the hours to days following a TBI, several signaling molecules and metabolic derangements result in disruption of the blood-brain barrier, leading to an extravasation of immune cells and cerebral edema. The primary, sudden injury in TBI occurs as a direct result of impact and therefore cannot be treated, but the timeline and pathophysiology of the delayed, secondary injury allows for a window of possible therapeutic options. The goal of this review is to discuss the pathophysiology of the primary and delayed injury in TBI as well as present several preclinical studies that identify molecular targets in the potential treatment of TBI. Additionally, certain recent clinical trials are briefly discussed to demonstrate the current state of TBI investigation.
Collapse
Affiliation(s)
- Suraj Sulhan
- Department of Neurosurgery, Baylor Scott & White Health Neuroscience Institute, Temple, Texas.,College of Medicine, Texas A&M University, Temple, Texas
| | - Kristopher A Lyon
- Department of Neurosurgery, Baylor Scott & White Health Neuroscience Institute, Temple, Texas.,College of Medicine, Texas A&M University, Temple, Texas
| | - Lee A Shapiro
- College of Medicine, Texas A&M University, Temple, Texas
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health Neuroscience Institute, Temple, Texas.,College of Medicine, Texas A&M University, Temple, Texas
| |
Collapse
|
35
|
Haber M, James J, Kim J, Sangobowale M, Irizarry R, Ho J, Nikulina E, Grin'kina NM, Ramadani A, Hartman I, Bergold PJ. Minocycline plus N-acteylcysteine induces remyelination, synergistically protects oligodendrocytes and modifies neuroinflammation in a rat model of mild traumatic brain injury. J Cereb Blood Flow Metab 2018; 38:1312-1326. [PMID: 28685618 PMCID: PMC6092769 DOI: 10.1177/0271678x17718106] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mild traumatic brain injury afflicts over 2 million people annually and little can be done for the underlying injury. The Food and Drug Administration-approved drugs Minocycline plus N-acetylcysteine (MINO plus NAC) synergistically improved cognition and memory in a rat mild controlled cortical impact (mCCI) model of traumatic brain injury.3 The underlying cellular and molecular mechanisms of the drug combination are unknown. This study addressed the effect of the drug combination on white matter damage and neuroinflammation after mCCI. Brain tissue from mCCI rats given either sham-injury, saline, MINO alone, NAC alone, or MINO plus NAC was investigated via histology and qPCR at four time points (2, 4, 7, and 14 days post-injury) for markers of white matter damage and neuroinflammation. MINO plus NAC synergistically protected resident oligodendrocytes and decreased the number of oligodendrocyte precursor cells. Activation of microglia/macrophages (MP/MG) was synergistically increased in white matter two days post-injury after MINO plus NAC treatment. Patterns of M1 and M2 MP/MG were also altered after treatment. The modulation of neuroinflammation is a potential mechanism to promote remyelination and improve cognition and memory. These data also provide new and important insights into how drug treatments can induce repair after traumatic brain injury.
Collapse
Affiliation(s)
- Margalit Haber
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Jessica James
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Justine Kim
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Michael Sangobowale
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Rachel Irizarry
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Johnson Ho
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Elena Nikulina
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Natalia M Grin'kina
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Albana Ramadani
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Isabella Hartman
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| | - Peter J Bergold
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, NY, USA
| |
Collapse
|
36
|
Miao H, Li R, Han C, Lu X, Zhang H. Minocycline promotes posthemorrhagic neurogenesis via M2 microglia polarization via upregulation of the TrkB/BDNF pathway in rats. J Neurophysiol 2018; 120:1307-1317. [PMID: 29790836 DOI: 10.1152/jn.00234.2018] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating disease worldwide with increasing mortality. The present study investigated whether minocycline was neuroprotective and induced M2 microglial polarization via upregulation of the TrkB/BDNF pathway after ICH. ICH was induced via injection of autologous blood into 150 Sprague-Dawley rats. A selective TrkB antagonist [N2-2-2-oxoazepan-3-yl amino] carbonyl phenyl benzo (b) thiophene-2-carboxamide (ANA 12)] and agonist [ N-[2-(5-hydroxy-1H-indol-3-yl) ethyl]-2-oxopiperidine-3-carboxamide (HIOC)] were used to investigate the mechanism of minocycline-induced neuroprotection. Minocycline improved ICH-induced neurological deficits and reduced M1 microglia marker protein (CD68, CD16) expression as well as M2 microglial polarization (CD206 and arginase 1 protein). Minocycline administration enhanced microglia-neuron cross talk and promoted the proliferation of neuronal progenitor cells, such as DCX- and Tuj-1-positive cells, 24 h after ICH. Minocycline also increased M2 microglia-derived brain-derived neurotrophic factors (BDNF) and the upstream TrkB pathway. ANA 12 reversed the neuroprotective effects of minocycline. HIOC exhibited the same effects as minocycline and accelerated neurogenesis after ICH. This study demonstrated for the first time that minocycline promoted M2 microglia polarization via upregulation of the TrkB/BDNF pathway and promoted neurogenesis after ICH. This study contributes to our understanding of the therapeutic potential of minocycline in ICH. NEW & NOTEWORTHY The present study gives several novel points: 1) Minocycline promotes neurogenesis after intracerebral hemorrhage in rats. 2) Minocycline induces activated M1 microglia into M2 neurotrophic phenotype. 3) M2 microglia secreting BDNF remodel the damaged neurocircuit.
Collapse
Affiliation(s)
- Hongsheng Miao
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine
| | - Runming Li
- Department of Neurosurgery, No. 205 Hospital of People's Liberation Army of China, Jingzhou, China
| | - Cong Han
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine
| | - Xiuzhen Lu
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine
| | - Hang Zhang
- Department of Neurosurgery, No. 205 Hospital of People's Liberation Army of China, Jingzhou, China
| |
Collapse
|
37
|
Wang J, Su E, Wang H, Guo C, Lawrence DA, Eitzman DT. Traumatic Brain Injury Leads to Accelerated Atherosclerosis in Apolipoprotein E Deficient Mice. Sci Rep 2018; 8:5639. [PMID: 29618740 PMCID: PMC5884790 DOI: 10.1038/s41598-018-23959-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/21/2018] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) has been associated with atherosclerosis and cardiovascular mortality in humans. However the causal relationship between TBI and vascular disease is unclear. This study investigated the direct role of TBI on vascular disease using a murine model of atherosclerosis. Apolipoprotein E deficient mice were placed on a western diet beginning at 10 weeks of age. Induction of TBI or a sham operation was performed at 14 weeks of age and mice were sacrificed 6 weeks later at 20 weeks of age. MRI revealed evidence of uniform brain injury in all mice subjected to TBI. There were no differences in total cholesterol levels or blood pressure between the groups. Complete blood counts and flow cytometry analysis performed on peripheral blood 6 weeks following TBI revealed a higher percentage of Ly6C-high monocytes in mice subjected to TBI compared to sham-treated mice. Mice with TBI also showed elevated levels of plasma soluble E-selectin and bone marrow tyrosine hydroxylase. Analysis of atherosclerosis at the time of sacrifice revealed increased atherosclerosis with increased Ly6C/G immunostaining in TBI mice compared to sham-treated mice. In conclusion, progression of atherosclerosis is accelerated following TBI. Targeting inflammatory pathways in patients with TBI may reduce subsequent vascular complications.
Collapse
Affiliation(s)
- Jintao Wang
- University of Michigan, Department of Internal Medicine, Cardiovascular Research Center, Ann Arbor, Michigan, USA
| | - Enming Su
- University of Michigan, Department of Internal Medicine, Cardiovascular Research Center, Ann Arbor, Michigan, USA
| | - Hui Wang
- University of Michigan, Department of Internal Medicine, Cardiovascular Research Center, Ann Arbor, Michigan, USA
| | - Chiao Guo
- University of Michigan, Department of Internal Medicine, Cardiovascular Research Center, Ann Arbor, Michigan, USA
| | - Daniel A Lawrence
- University of Michigan, Department of Internal Medicine, Cardiovascular Research Center, Ann Arbor, Michigan, USA
| | - Daniel T Eitzman
- University of Michigan, Department of Internal Medicine, Cardiovascular Research Center, Ann Arbor, Michigan, USA.
| |
Collapse
|
38
|
Abstract
Accumulating research substantiates the statement that inflammation plays an important role in the development of stroke. Both proinflammatory and anti-inflammatory mediators are involved in the pathogenesis of stroke, an imbalance of which leads to inflammation. Anti-inflammation is a kind of hopeful strategy for the prevention and treatment of stroke. Substantial studies have demonstrated that minocycline, a second-generation semisynthetic antibiotic belonging to the tetracycline family, can inhibit neuroinflammation, inflammatory mediators and microglia activation, and improve neurological outcome. Experimental and clinical data have found the preclinical and clinical potential of minocycline in the treatment of stroke due to its anti-inflammation properties and anti-inflammation-induced pathogeneses, including antioxidative stress, antiapoptosis, inhibiting leukocyte migration and microglial activation, and decreasing matrix metalloproteinases activity. Hence, it suggests a great future for minocycline in the therapeutics of stroke that diminish the inflammatory progress of stroke.
Collapse
|
39
|
Yin J, Zhang H, Chen H, Lv Q, Jin X. Hypertonic Saline Alleviates Brain Edema After Traumatic Brain Injury via Downregulation of Aquaporin 4 in Rats. Med Sci Monit 2018; 24:1863-1870. [PMID: 29600800 PMCID: PMC5890824 DOI: 10.12659/msm.907212] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Hypertonic saline (HS) has been successfully used for treatment of various forms of brain edema. Decreased expression of aquaporin (AQP)4 and pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-1β have been linked to edema pathogenesis. This study examined the effect of 3% HS on brain edema in a rat model of traumatic brain injury (TBI). Material/Methods Sprague-Dawley rats were subjected to TBI induced by a controlled cortical impactor. The HS group was injected with 3% NaCl until the end of the study period. AQP4, TNF-α, IL-1β, and caspase-3 levels were measured by Western blotting, immunohistochemistry, enzyme-linked immunosorbent assay, and quantitative real-time PCR. Brain water content was also measured. Apoptotic cells in brain tissue were detected with terminal deoxynucleotidyl transferase dUTP nick-end labeling. Brain water content decreased following treatment with 3% HS relative to the TBI group. Results This was accompanied by decreases in AQP4, TNF-α, and IL-1β mRNA and protein levels. TBI resulted in increases in caspase-3 mRNA expression and the number of apoptotic cells; treatment with 3% HS suppressed apoptosis as compared to the TBI group. Conclusions Treatment with 3% HS ameliorated TBI-induced brain edema, possibly by suppressing brain edema, pro-inflammatory cytokine expression, and apoptosis.
Collapse
Affiliation(s)
- Jian Yin
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Haixiao Zhang
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Huai Chen
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Qingping Lv
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xuhong Jin
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
40
|
Sangobowale MA, Grin'kina NM, Whitney K, Nikulina E, St Laurent-Ariot K, Ho JS, Bayzan N, Bergold PJ. Minocycline plus N-Acetylcysteine Reduce Behavioral Deficits and Improve Histology with a Clinically Useful Time Window. J Neurotrauma 2018; 35:907-917. [PMID: 29187031 DOI: 10.1089/neu.2017.5348] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
There are no drugs to manage traumatic brain injury (TBI) presently. A major problem in developing therapeutics is that drugs to manage TBI lack sufficient potency when dosed within a clinically relevant time window. Previous studies have shown that minocycline (MINO, 45 mg/kg) plus N-acetylcysteine (NAC, 150 mg/kg) synergistically improved cognition and memory, modulated inflammation, and prevented loss of oligodendrocytes that remyelinated damaged white matter when first dosed 1 h after controlled cortical impact (CCI) in rats. We show that MINO (45 mg/kg) plus NAC (150 mg/kg) also prevent brain injury in a mouse closed head injury (CHI) TBI model. Using the CHI model, the concentrations of MINO and NAC were titrated to determine that MINO (22.5 mg/kg) plus NAC (75 mg/kg) was more potent than the original formulation. MINO (22.5 mg/kg) plus NAC (75 mg/kg) also limited injury in the rat CCI model. The therapeutic time window of MINO plus NAC was then tested in the CHI and CCI models. Mice and rats could acquire an active place avoidance task when MINO plus NAC was first dosed at 12 h post-injury. A first dose at 12 h also limited gray matter injury in the hippocampus and preserved myelin in multiple white matter tracts. Mice and rats acquired Barnes maze when MINO plus NAC was first dosed at 24 h post-injury. These data suggest that MINO (22.5 mg/kg) plus NAC (75 mg/kg) remain potent when dosed at clinically useful time windows. Both MINO and NAC are drugs approved by the Food and Drug Administration and have been administered safely to patients in clinical trials at the doses in the new formulation. This suggests that the drug combination of MINO plus NAC may be effective in treating patients with TBI.
Collapse
Affiliation(s)
| | - Natalia M Grin'kina
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Kristen Whitney
- School of Graduate Studies, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Elena Nikulina
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Karrah St Laurent-Ariot
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Johnson S Ho
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Narek Bayzan
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Peter J Bergold
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York.,Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
41
|
Bhatti J, Nascimento B, Akhtar U, Rhind SG, Tien H, Nathens A, da Luz LT. Systematic Review of Human and Animal Studies Examining the Efficacy and Safety of N-Acetylcysteine (NAC) and N-Acetylcysteine Amide (NACA) in Traumatic Brain Injury: Impact on Neurofunctional Outcome and Biomarkers of Oxidative Stress and Inflammation. Front Neurol 2018; 8:744. [PMID: 29387038 PMCID: PMC5776005 DOI: 10.3389/fneur.2017.00744] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Background No new therapies for traumatic brain injury (TBI) have been officially translated into current practice. At the tissue and cellular level, both inflammatory and oxidative processes may be exacerbated post-injury and contribute to further brain damage. N-acetylcysteine (NAC) has the potential to downregulate both processes. This review focuses on the potential neuroprotective utility of NAC and N-acetylcysteine amide (NACA) post-TBI. Methods Medline, Embase, Cochrane Library, and ClinicalTrials.gov were searched up to July 2017. Studies that examined clinical and laboratory effects of NAC and NACA post-TBI in human and animal studies were included. Risk of bias was assessed in human and animal studies according to the design of each study (randomized or not). The primary outcome assessed was the effect of NAC/NACA treatment on functional outcome, while secondary outcomes included the impact on biomarkers of inflammation and oxidation. Due to the clinical and methodological heterogeneity observed across studies, no meta-analyses were conducted. Results Our analyses revealed only three human trials, including two randomized controlled trials (RCTs) and 20 animal studies conducted using standardized animal models of brain injury. The two RCTs reported improvement in the functional outcome post-NAC/NACA administration. Overall, the evidence from animal studies is more robust and demonstrated substantial improvement of cognition and psychomotor performance following NAC/NACA use. Animal studies also reported significantly more cortical sparing, reduced apoptosis, and lower levels of biomarkers of inflammation and oxidative stress. No safety concerns were reported in any of the studies included in this analysis. Conclusion Evidence from the animal literature demonstrates a robust association for the prophylactic application of NAC and NACA post-TBI with improved neurofunctional outcomes and downregulation of inflammatory and oxidative stress markers at the tissue level. While a growing body of scientific literature suggests putative beneficial effects of NAC/NACA treatment for TBI, the lack of well-designed and controlled clinical investigations, evaluating therapeutic outcomes, prognostic biomarkers, and safety profiles, limits definitive interpretation and recommendations for its application in humans at this time.
Collapse
Affiliation(s)
- Junaid Bhatti
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Barto Nascimento
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Umbreen Akhtar
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Shawn G Rhind
- Defense Research and Development Canada (DRDC), Toronto Research Centre, Toronto, ON, Canada
| | - Homer Tien
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Avery Nathens
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Luis Teodoro da Luz
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
42
|
Somayaji MR, Przekwas AJ, Gupta RK. Combination Therapy for Multi-Target Manipulation of Secondary Brain Injury Mechanisms. Curr Neuropharmacol 2018; 16:484-504. [PMID: 28847295 PMCID: PMC6018188 DOI: 10.2174/1570159x15666170828165711] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a major healthcare problem that affects millions of people worldwide. Despite advances in understanding and developing preventative and treatment strategies using preclinical animal models, clinical trials to date have failed, and a 'magic bullet' for effectively treating TBI-induced damage does not exist. Thus, novel pharmacological strategies to effectively manipulate the complex and heterogeneous pathophysiology of secondary injury mechanisms are needed. Given that goal, this paper discusses the relevance and advantages of combination therapies (COMTs) for 'multi-target manipulation' of the secondary injury cascade by administering multiple drugs to achieve an optimal therapeutic window of opportunity (e.g., temporally broad window) and compares these regimens to monotherapies that manipulate a single target with a single drug at a given time. Furthermore, we posit that integrated mechanistic multiscale models that combine primary injury biomechanics, secondary injury mechanobiology/neurobiology, physiology, pharmacology and mathematical programming techniques could account for vast differences in the biological space and time scales and help to accelerate drug development, to optimize pharmacological COMT protocols and to improve treatment outcomes.
Collapse
Affiliation(s)
| | | | - Raj K. Gupta
- Department of Defense Blast Injury Research Program Coordinating Office, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| |
Collapse
|
43
|
Yang X, Zhang JD, Duan L, Xiong HG, Jiang YP, Liang HC. Microglia activation mediated by toll-like receptor-4 impairs brain white matter tracts in rats. J Biomed Res 2017; 32:136-144. [PMID: 29358565 PMCID: PMC5895568 DOI: 10.7555/jbr.32.20170033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Microglia activation and white matter injury coexist after repeated episodes of mild brain trauma and ischemic stroke. Axon degeneration and demyelination can activate microglia; however, it is unclear whether early microglia activation can impair the function of white matter tracts and lead to injury. Rat corpus callosum (CC) slices were treated with lipopolysaccharide (LPS) or LPS + Rhodobacter sphaeroides (RS)-LPS that is a toll-like receptor 4 (TLR-4) antagonist. Functional changes reflected by the change of axon compound action potentials (CAPs) and the accumulation of β-amyloid precursor protein (β-APP) in CC nerve fibers. Microglia activation was monitored by ionized calcium binding adaptor-1 immunofluorescent stain, based on well-established morphological criteria and paralleled proportional area measurement. Input-output (I/O) curves of CAPs in response to increased stimuli were significantly downshifted in a dose-dependent manner in LPS (0.2, 0.5 and 1.0 μg/mL)-treated slices, implying that axons neurophysiological function was undermined. LPS caused significant β-APP accumulation in CC tissues, reflecting the deterioration of fast axon transport. LPS-induced I/O curve downshift and β-APP accumulation were significantly reversed by the pre-treatment or co-incubation with RS-LPS. RS-LPS alone did not change the I/O curve. The degree of malfunction was correlated with microglia activation, as was shown by the measurements of proportional areas. Function of CC nerve fibers was evidently impaired by microglia activation and reversed by a TLP-4 antagonist, suggesting that the TLP-4 pathway lead to microglia activation.
Collapse
Affiliation(s)
- Xinglong Yang
- Department of Neurosurgery, Affiliated Hospital to Academy of Military Medicine Sciences, Beijing 100071, China
| | - Jing-Dong Zhang
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lian Duan
- Department of Neurosurgery, Affiliated Hospital to Academy of Military Medicine Sciences, Beijing 100071, China
| | - Huan-Gui Xiong
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yan-Ping Jiang
- Department of Otolaryngology, the 306th PLA Hospital, Beijing 100101, China
| | | |
Collapse
|
44
|
Rosenblatt J, Reitzel RA, Viola GM, Vargas-Cruz N, Selber J, Raad I. Sodium Mercaptoethane Sulfonate Reduces Collagenolytic Degradation and Synergistically Enhances Antimicrobial Durability in an Antibiotic-Loaded Biopolymer Film for Prevention of Surgical-Site Infections. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3149536. [PMID: 29238713 PMCID: PMC5697372 DOI: 10.1155/2017/3149536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/09/2017] [Indexed: 11/26/2022]
Abstract
Implant-associated surgical-site infections can have significant clinical consequences. Previously we reported a method for prophylactically disinfecting implant surfaces in surgical pockets, where an antibiotic solution containing minocycline (M) and rifampin (R) was applied as a solid film in a crosslinked biopolymer matrix that partially liquefied in situ to provide extended prophylaxis. Here we studied the effect of adding sodium 2-mercaptoethane sulfonate (MeSNA) on durability of prophylaxis in an in vitro model of implant-associated surgical-site infection. Adding MeSNA to the M/R biopolymer, antimicrobial film extended the duration for which biofilm formation by multidrug-resistant Pseudomonas aeruginosa (MDR-PA) was prevented on silicone surfaces in the model. M/R films with and without MeSNA were effective in preventing colonization by methicillin-resistant Staphylococcus aureus. Independent experiments revealed that MeSNA directly inhibited proteolytic digestion of the biopolymer film and synergistically enhanced antimicrobial potency of M/R against MDR-PA. Incubation of the MeSNA containing films with L929 fibroblasts revealed no impairment of cellular metabolic activity or viability.
Collapse
Affiliation(s)
- Joel Rosenblatt
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ruth A. Reitzel
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George M. Viola
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nylev Vargas-Cruz
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jesse Selber
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Issam Raad
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
45
|
Liu Y, Wang R, Zhao Z, Dong W, Zhang X, Chen X, Ma L. Short-term caloric restriction exerts neuroprotective effects following mild traumatic brain injury by promoting autophagy and inhibiting astrocyte activation. Behav Brain Res 2017; 331:135-142. [PMID: 28495608 DOI: 10.1016/j.bbr.2017.04.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/08/2017] [Accepted: 04/11/2017] [Indexed: 11/26/2022]
Abstract
Cognitive deficits may occur after mild traumatic brain injury (mTBI), but effective treatment modalities are presently unavailable. Caloric restriction (CR) has beneficial effects on neurodegenerative diseases and brain injury. However, the underlying mechanisms have not yet been clearly defined. Therefore, the aim of the present study was to investigate the short-term effects of CR treatment on cognitive function in mice after mTBI. Forty-five 12-week-old C57/BL6 mice were subjected to closed-head mTBI using a weight drop device. The mice were then randomly divided into three groups according to their diet for 30 days: the normal calorie group (mTBI+NC group, n=15), the caloric restriction group (mTBI+CR group, n=15), and the high energy group (mTBI+HE group, n=15). After 30 days, the Morris water maze test was performed to evaluate learning abilities. Nissl staining, immunohistochemistry, and western blotting were used to monitor pathological changes and changes in autophagy-associated proteins in the hippocampus. The average escape latency was significantly shorter in the mTBI+CR group than in the mTBI+NC and mTBI+HE groups, and the number of target platform crossings in the mTBI+CR group was significantly higher than in the other two groups. In the hippocampus, the expression of GFAP and mTOR was increased in the mTBI+HE group and decreased in the mTBI+CR group. Conversely, the expression of LC3B was decreased in the mTBI+HE group and increased in the mTBI+CR group. Our findings suggest that short-term CR after mTBI may ameliorate cognitive dysfunction induced by mTBI by increasing the level of autophagy and suppressing astrocyte activation.
Collapse
Affiliation(s)
- Yuan Liu
- Central Laboratory, Xuan Wu Hospital, Capital Medical University, Beijing Institute for Brain Disorders, Center of Alzheimer's Disease, Beijing 100053, China
| | - Rong Wang
- Central Laboratory, Xuan Wu Hospital, Capital Medical University, Beijing Institute for Brain Disorders, Center of Alzheimer's Disease, Beijing 100053, China; Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing 100053, China.
| | - Zhiwei Zhao
- Central Laboratory, Xuan Wu Hospital, Capital Medical University, Beijing Institute for Brain Disorders, Center of Alzheimer's Disease, Beijing 100053, China
| | - Wen Dong
- Central Laboratory, Xuan Wu Hospital, Capital Medical University, Beijing Institute for Brain Disorders, Center of Alzheimer's Disease, Beijing 100053, China
| | - Xu Zhang
- Central Laboratory, Xuan Wu Hospital, Capital Medical University, Beijing Institute for Brain Disorders, Center of Alzheimer's Disease, Beijing 100053, China
| | - Xi Chen
- Department of Infectious Diseases, Chifeng City Hospital, Chifeng Clinical Medicine College of Inner Mongolia Medical University, Chifeng, Inner Mongolia Autonomous Region, 024000, China
| | - Lina Ma
- Department of Geriatrics, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
46
|
SanMartín CD, Veloso P, Adasme T, Lobos P, Bruna B, Galaz J, García A, Hartel S, Hidalgo C, Paula-Lima AC. RyR2-Mediated Ca 2+ Release and Mitochondrial ROS Generation Partake in the Synaptic Dysfunction Caused by Amyloid β Peptide Oligomers. Front Mol Neurosci 2017; 10:115. [PMID: 28487634 PMCID: PMC5403897 DOI: 10.3389/fnmol.2017.00115] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/05/2017] [Indexed: 11/13/2022] Open
Abstract
Amyloid β peptide oligomers (AβOs), toxic aggregates with pivotal roles in Alzheimer's disease, trigger persistent and low magnitude Ca2+ signals in neurons. We reported previously that these Ca2+ signals, which arise from Ca2+ entry and subsequent amplification by Ca2+ release through ryanodine receptor (RyR) channels, promote mitochondrial network fragmentation and reduce RyR2 expression. Here, we examined if AβOs, by inducing redox sensitive RyR-mediated Ca2+ release, stimulate mitochondrial Ca2+-uptake, ROS generation and mitochondrial fragmentation, and also investigated the effects of the antioxidant N-acetyl cysteine (NAC) and the mitochondrial antioxidant EUK-134 on AβOs-induced mitochondrial dysfunction. In addition, we studied the contribution of the RyR2 isoform to AβOs-induced Ca2+ release, mitochondrial Ca2+ uptake and fragmentation. We show here that inhibition of NADPH oxidase type-2 prevented the emergence of RyR-mediated cytoplasmic Ca2+ signals induced by AβOs in primary hippocampal neurons. Treatment with AβOs promoted mitochondrial Ca2+ uptake and increased mitochondrial superoxide and hydrogen peroxide levels; ryanodine, at concentrations that suppress RyR activity, prevented these responses. The antioxidants NAC and EUK-134 impeded the mitochondrial ROS increase induced by AβOs. Additionally, EUK-134 prevented the mitochondrial fragmentation induced by AβOs, as previously reported for NAC and ryanodine. These findings show that both antioxidants, NAC and EUK-134, prevented the Ca2+-mediated noxious effects of AβOs on mitochondrial function. Our results also indicate that Ca2+ release mediated by the RyR2 isoform causes the deleterious effects of AβOs on mitochondrial function. Knockdown of RyR2 with antisense oligonucleotides reduced by about 50% RyR2 mRNA and protein levels in primary hippocampal neurons, decreased by 40% Ca2+ release induced by the RyR agonist 4-chloro-m-cresol, and significantly reduced the cytoplasmic and mitochondrial Ca2+ signals and the mitochondrial fragmentation induced by AβOs. Based on our results, we propose that AβOs-induced Ca2+ entry and ROS generation jointly stimulate RyR2 activity, causing mitochondrial Ca2+ overload and fragmentation in a feed forward injurious cycle. The present novel findings highlight the specific participation of RyR2-mediated Ca2+ release on AβOs-induced mitochondrial malfunction.
Collapse
Affiliation(s)
- Carol D SanMartín
- Department of de Neurology and Neurosurgery, Clinical Hospital Universidad de ChileSantiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile
| | - Pablo Veloso
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile.,Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de ChileSantiago, Chile
| | - Tatiana Adasme
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile.,Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O HigginsSantiago, Chile
| | - Pedro Lobos
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile
| | - Barbara Bruna
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile
| | - Jose Galaz
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile
| | - Alejandra García
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile.,Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Center of Medical Informatics and Telemedicine and National Center for Health Information Systems, Faculty of Medicine, Universidad de ChileSantiago, Chile
| | - Steffen Hartel
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile.,Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Center of Medical Informatics and Telemedicine and National Center for Health Information Systems, Faculty of Medicine, Universidad de ChileSantiago, Chile
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile.,Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de ChileSantiago, Chile
| | - Andrea C Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de ChileSantiago, Chile.,Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de ChileSantiago, Chile
| |
Collapse
|
47
|
The Polarization States of Microglia in TBI: A New Paradigm for Pharmacological Intervention. Neural Plast 2017; 2017:5405104. [PMID: 28255460 PMCID: PMC5309408 DOI: 10.1155/2017/5405104] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/17/2016] [Accepted: 01/11/2017] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) is a serious medical and social problem worldwide. Because of the complex pathophysiological mechanisms of TBI, effective pharmacotherapy is still lacking. The microglial cells are resident tissue macrophages located in the brain and have two major polarization states, M1 phenotype and M2 phenotype, when activated. The M1 phenotype is related to the release of proinflammatory cytokines and secondary brain injury, while the M2 phenotype has been proved to be responsible for the release of anti-inflammation cytokines and for central nervous system (CNS) repair. In animal models, pharmacological strategies inhibiting the M1 phenotype and promoting the M2 phenotype of microglial cells could alleviate cerebral damage and improve neurological function recovery after TBI. In this review, we aimed to summarize the current knowledge about the pathological significance of microglial M1/M2 polarization in the pathophysiology of TBI. In addition, we reviewed several drugs that have provided neuroprotective effects against brain injury following TBI by altering the polarization states of the microglia. We emphasized that future investigation of the regulation mechanisms of microglial M1/M2 polarization in TBI is anticipated, which could contribute to the development of new targets of pharmacological intervention in TBI.
Collapse
|
48
|
Trojian TH, Wang DH, Leddy JJ. Nutritional Supplements for the Treatment and Prevention of Sports-Related Concussion—Evidence Still Lacking. Curr Sports Med Rep 2017; 16:247-255. [DOI: 10.1249/jsr.0000000000000387] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
Kochanek PM, Bramlett HM, Shear DA, Dixon CE, Mondello S, Dietrich WD, Hayes RL, Wang KKW, Poloyac SM, Empey PE, Povlishock JT, Mountney A, Browning M, Deng-Bryant Y, Yan HQ, Jackson TC, Catania M, Glushakova O, Richieri SP, Tortella FC. Synthesis of Findings, Current Investigations, and Future Directions: Operation Brain Trauma Therapy. J Neurotrauma 2016; 33:606-14. [PMID: 26671284 DOI: 10.1089/neu.2015.4133] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Operation Brain Trauma Therapy (OBTT) is a fully operational, rigorous, and productive multicenter, pre-clinical drug and circulating biomarker screening consortium for the field of traumatic brain injury (TBI). In this article, we synthesize the findings from the first five therapies tested by OBTT and discuss both the current work that is ongoing and potential future directions. Based on the results generated from the first five therapies tested within the exacting approach used by OBTT, four (nicotinamide, erythropoietin, cyclosporine A, and simvastatin) performed below or well below what was expected based on the published literature. OBTT has identified, however, the early post-TBI administration of levetiracetam as a promising agent and has advanced it to a gyrencephalic large animal model--fluid percussion injury in micropigs. The sixth and seventh therapies have just completed testing (glibenclamide and Kollidon VA 64), and an eighth drug (AER 271) is in testing. Incorporation of circulating brain injury biomarker assessments into these pre-clinical studies suggests considerable potential for diagnostic and theranostic utility of glial fibrillary acidic protein in pre-clinical studies. Given the failures in clinical translation of therapies in TBI, rigorous multicenter, pre-clinical approaches to therapeutic screening such as OBTT may be important for the ultimate translation of therapies to the human condition.
Collapse
Affiliation(s)
- Patrick M Kochanek
- 1 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Helen M Bramlett
- 2 Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami , Miami, Florida.,3 Bruce W. Carter Department of Veterans Affairs Medical Center , Miami, Florida
| | - Deborah A Shear
- 4 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - C Edward Dixon
- 5 Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Stefania Mondello
- 6 Department of Neurosciences, University of Messina , Messina, Italy
| | - W Dalton Dietrich
- 2 Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami , Miami, Florida
| | - Ronald L Hayes
- 7 Center for Innovative Research, Center for Neuroproteomics and Biomarkers Research, Banyan Biomarkers, Inc. , Alachua, Florida
| | - Kevin K W Wang
- 8 Center of Neuroproteomics and Biomarkers Research, Department of Psychiatry and Neuroscience, University of Florida. Gainesville, Florida
| | - Samuel M Poloyac
- 9 Center for Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy , Pittsburgh, Pennsylvania
| | - Philip E Empey
- 9 Center for Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy , Pittsburgh, Pennsylvania
| | - John T Povlishock
- 10 Department of Anatomy and Neurobiology, Virginia Commonwealth University , Richmond, Virginia
| | - Andrea Mountney
- 4 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Megan Browning
- 1 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Ying Deng-Bryant
- 4 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Hong Q Yan
- 5 Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Travis C Jackson
- 1 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | | | | | | | - Frank C Tortella
- 4 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| |
Collapse
|
50
|
Shultz SR, McDonald SJ, Vonder Haar C, Meconi A, Vink R, van Donkelaar P, Taneja C, Iverson GL, Christie BR. The potential for animal models to provide insight into mild traumatic brain injury: Translational challenges and strategies. Neurosci Biobehav Rev 2016; 76:396-414. [PMID: 27659125 DOI: 10.1016/j.neubiorev.2016.09.014] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 09/07/2016] [Accepted: 09/16/2016] [Indexed: 12/14/2022]
Abstract
Mild traumatic brain injury (mTBI) is a common health problem. There is tremendous variability and heterogeneity in human mTBI, including mechanisms of injury, biomechanical forces, injury severity, spatial and temporal pathophysiology, genetic factors, pre-injury vulnerability and resilience factors, and clinical outcomes. Animal models greatly reduce this variability and heterogeneity, and provide a means to study mTBI in a rigorous, controlled, and efficient manner. Rodent models, in particular, are time- and cost-efficient, and they allow researchers to measure morphological, cellular, molecular, and behavioral variables in a single study. However, inter-species differences in anatomy, morphology, metabolism, neurobiology, and lifespan create translational challenges. Although the term "mild" TBI is used often in the pre-clinical literature, clearly defined criteria for mild, moderate, and severe TBI in animal models have not been agreed upon. In this review, we introduce current issues facing the mTBI field, summarize the available research methodologies and previous studies in mTBI animal models, and discuss how a translational research approach may be useful in advancing our understanding and management of mTBI.
Collapse
Affiliation(s)
- Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Cole Vonder Haar
- Department of Psychology, The University of British Columbia, Vancouver, BC, Canada
| | - Alicia Meconi
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Robert Vink
- Division of Health Sciences, The University of South Australia, Adelaide, SA, Australia
| | - Paul van Donkelaar
- School of Health and Exercise Sciences, The University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Chand Taneja
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, and MassGeneral Hospital for Children™ Sports Concussion Program, Boston, MA, USA
| | - Brian R Christie
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| |
Collapse
|