1
|
Eyolfson E, Suesser KRB, Henry H, Bonilla-Del Río I, Grandes P, Mychasiuk R, Christie BR. The effect of traumatic brain injury on learning and memory: A synaptic focus. Neuroscientist 2025; 31:195-214. [PMID: 39316552 PMCID: PMC11909778 DOI: 10.1177/10738584241275583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Deficits in learning and memory are some of the most commonly reported symptoms following a traumatic brain injury (TBI). We will examine whether the neural basis of these deficits stems from alterations to bidirectional synaptic plasticity within the hippocampus. Although the CA1 subregion of the hippocampus has been a focus of TBI research, the dentate gyrus should also be given attention as it exhibits a unique ability for adult neurogenesis, a process highly susceptible to TBI-induced damage. This review examines our current understanding of how TBI results in deficits in synaptic plasticity, as well as how TBI-induced changes in endocannabinoid (eCB) systems may drive these changes. Through the synthesis and amalgamation of existing data, we propose a possible mechanism for eCB-mediated recovery in synaptic plasticity deficits. This hypothesis is based on the plausible roles of CB1 receptors in regulating inhibitory tone, influencing astrocytes and microglia, and modulating glutamate release. Dysregulation of the eCBs may be responsible for deficits in synaptic plasticity and learning following TBI. Taken together, the existing evidence indicates eCBs may contribute to TBI manifestation, pathogenesis, and recovery, but it also suggests there may be a therapeutic role for the eCB system in TBI.
Collapse
Affiliation(s)
- Eric Eyolfson
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Kirsten R. B. Suesser
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Holly Henry
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Itziar Bonilla-Del Río
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country, Leioa, Spain
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country, Leioa, Spain
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Brian R. Christie
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, San Diego State University, San Diego, CA, USA
| |
Collapse
|
2
|
Sadino A, Saptarini NM, Levita J, Ramadhan DSF, Fristiohady A, Jiranusornkul S. Identifying Potential Human Monoacylglycerol Lipase Inhibitors from the Phytoconstituents of Morinda Citrifolia L. Fruits by in silico Pharmacology and in vitro Study. J Exp Pharmacol 2024; 16:295-309. [PMID: 39345798 PMCID: PMC11436673 DOI: 10.2147/jep.s477956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
Background Human monoacylglycerol lipase (MGL) is accountable for the hydrolysis of 2-arachidonoylglycerol (2-AG), thus contributing pivotally to neuroprotection because 2-AG is the main source of arachidonic acid, the precursor of prostaglandins production. Inhibiting MGL reduces inflammatory damage in the ischemic brain and enhances cerebral blood flow. Plants have been reported for their neuroprotective effect, such as Morinda citrifolia on pentylenetetrazol (PTZ)-induced kindling seizures in mice, by reducing the seizures and restoring behavioral and biochemical changes, although the mechanism is not described. Purpose To evaluate the binding affinity and stability of phytoconstituents in M. citrifolia fruits toward human MGL (PDB ID 3PE6), compared to the known MGL inhibitors (JZL195 and ZYH). The in silico pharmacology study was validated by an in vitro study of the phytosterols and the ethanol extract of M. citrifolia fruits (EEMC) towards MGL. Methods Initially, nine phytoconstituents of M. citrifolia fruits were docked to the catalytic pocket of human MGL (PDB ID: 3PE6), and compounds with the best affinity were subjected to a molecular dynamic (MD) simulation. The in vitro study was performed using the MGL inhibitor screening assay kit. Results The best binding affinity and stability toward human MGL were shown by stigmasterol and beta-sitosterol, and the MM-PBSA total binding energy of stigmasterol and beta-sitosterol to MGL is stronger than that of JZL195 and ZYH. Moreover, beta-sitosterol and EEMC inhibit MGL with an IC50 value of, respectively, 8.10 μg/mL and 196.20 μg/mL, while JZL195 shows an IC50 of 0.028 μg/mL. Conclusion Beta-sitosterol of Morinda citrifolia fruits may have the potential to protect human neurons by occupying the catalytic site of human MGL, thus competitively inhibiting the substrate of the enzyme. However, the inhibitory activity towards human MGL is lower than JZL195.
Collapse
Affiliation(s)
- Asman Sadino
- Doctoral Program in Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Mathematics and Natural Sciences, Garut University, Garut, West Java, 44151, Indonesia
| | - Nyi Mekar Saptarini
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Jutti Levita
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, West Java, 45363, West Java, Indonesia
| | - Dwi Syah Fitra Ramadhan
- Department of Pharmacy, Poltekkes Kemenkes Makassar, Makassar, South Sulawesi, 90222, Indonesia
| | - Adryan Fristiohady
- Faculty of Pharmacy, Halu Oleo University, Kendari, Southeast Sulawesi, 93132, Indonesia
| | - Supat Jiranusornkul
- Department of Pharmaceutical Science, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
3
|
Zhu D, Zhang J, Ma X, Hu M, Gao F, Hashem JB, Lyu J, Wei J, Cui Y, Qiu S, Chen C. Overabundant endocannabinoids in neurons are detrimental to cognitive function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613513. [PMID: 39345517 PMCID: PMC11430108 DOI: 10.1101/2024.09.17.613513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
2-Arachidonoylglycerol (2-AG) is the most prevalent endocannabinoid involved in maintaining brain homeostasis. Previous studies have demonstrated that inactivating monoacylglycerol lipase (MAGL), the primary enzyme responsible for degrading 2-AG in the brain, alleviates neuropathology and prevents synaptic and cognitive decline in animal models of neurodegenerative diseases. However, we show that selectively inhibiting 2-AG metabolism in neurons impairs cognitive function in mice. This cognitive impairment appears to result from decreased expression of synaptic proteins and synapse numbers, impaired long-term synaptic plasticity and cortical circuit functional connectivity, and diminished neurogenesis. Interestingly, the synaptic and cognitive deficits induced by neuronal MAGL inactivation can be counterbalanced by inhibiting astrocytic 2-AG metabolism. Transcriptomic analyses reveal that inhibiting neuronal 2-AG degradation leads to widespread changes in expression of genes associated with synaptic function. These findings suggest that crosstalk in 2-AG signaling between astrocytes and neurons is crucial for maintaining synaptic and cognitive functions and that excessive 2-AG in neurons alone is detrimental to cognitive function.
Collapse
Affiliation(s)
- Dexiao Zhu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jian Zhang
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Xiaokuang Ma
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Mei Hu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Fei Gao
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jack B. Hashem
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jianlu Lyu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jing Wei
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Yuehua Cui
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Shenfeng Qiu
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Chu Chen
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| |
Collapse
|
4
|
Chen C. TDP-43 is a key molecule accelerating development of Alzheimer's disease following traumatic brain injury. Neural Regen Res 2024; 19:955-956. [PMID: 37862186 PMCID: PMC10749594 DOI: 10.4103/1673-5374.385301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 10/22/2023] Open
Affiliation(s)
- Chu Chen
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
5
|
Sunny A, James RR, Menon SR, Rayaroth S, Daniel A, Thompson NA, Tharakan B. Matrix Metalloproteinase-9 inhibitors as therapeutic drugs for traumatic brain injury. Neurochem Int 2024; 172:105642. [PMID: 38008261 DOI: 10.1016/j.neuint.2023.105642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality among young adults and the elderly. In the United States, TBI is responsible for around 30 percent of all injuries brought on by injuries in general. Vasogenic cerebral edema due to blood-brain barrier (BBB) dysfunction and the associated elevation of intracranial pressure (ICP) are some of the major causes of secondary injuries following traumatic brain injury. Matrix metalloproteinase-9 (MMP-9) is a therapeutic target for being an enzyme that degrades the proteins that make up a part of the microvascular basal lamina as well as inter-endothelial tight junctions of the blood-brain barrier. MMP-9-mediated BBB dysfunctions and the compromise of the BBB is a major pathway that leads the development of vasogenic cerebral edema, elevation of ICP, poor cerebral perfusion and brain herniation following traumatic brain injury. That makes MMP-9 an effective therapeutic target and endogenous or exogenous MMP-9 inhibitors as therapeutic drugs for preventing secondary brain damage after traumatic brain injury. Although our understanding of the mechanisms that underlie the primary and secondary stages of damage following a TBI has significantly improved in recent years, such information has not yet resulted in the successful development of novel pharmacological treatment options for traumatic brain injury. Recent pre-clinical and/or clinical studies have demonstrated that there are several compounds with specific or non-specific MMP-9 inhibitory properties either directly binding and inhibiting MMP-9 or by indirectly inhibiting MMP-9, with potential as therapeutic agents for traumatic brain injury. This article reviews the efficacy of several such medications and potential agents that include endogenous and exogeneous compounds that are at various levels of research and development. MMP-9-based therapeutic drug development has enormous potential in the pharmacological treatment of cerebral edema and/or neuronal injury resulting from traumatic brain injury.
Collapse
Affiliation(s)
- Angel Sunny
- Icahn School of Medicine at Mount Sinai, Elmhurst, NY, USA
| | | | | | | | - Abhijith Daniel
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Namita Ann Thompson
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
6
|
Jacotte-Simancas A, Molina PE, Gilpin NW. Repeated Mild Traumatic Brain Injury and JZL184 Produce Sex-Specific Increases in Anxiety-Like Behavior and Alcohol Consumption in Wistar Rats. J Neurotrauma 2023; 40:2427-2441. [PMID: 37503666 PMCID: PMC10649186 DOI: 10.1089/neu.2023.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Alcohol use disorder (AUD) is highly comorbid with traumatic brain injury (TBI). Previously, using a lateral fluid percussion model (LFP) (an open-head injury model) to generate a single mild to moderate traumatic brain injury (TBI) we showed that TBI produces escalation in alcohol drinking, that alcohol exposure negatively impacts TBI outcomes, and that the endocannabinoid degradation inhibitor (JZL184) confers significant protection from behavioral and neuropathological outcomes in male rodents. In the present study, we used a weight drop model (a closed-head injury model) to produce repeated mild TBI (rmTBI; three TBIs separated by 24 hours) in male and female rats to examine the sex-specific effects on anxiety-like behavior and alcohol consumption, and whether systemic treatment with JZL184 would reverse TBI effects on those behaviors. In two separate studies, adult male and female Wistar rats were subjected to rmTBI or sham procedure using the weight drop model. Physiological measures of injury severity were collected from all animals. Animals in both studies were allowed to consume alcohol using an intermittent 2-bottle choice procedure (12 pre-TBI sessions and 12 post-TBI sessions). Neurological severity and neurobehavioral scores (NSS and NBS, respectively) were tested 24 hours after the final injury. Anxiety-like behavior was tested at 37-38 days post-injury in Study 1-, and 6-8-days post-injury in Study 2. Our results show that females exhibited reduced respiratory rates relative to males with no significant differences between Sham and rmTBI, no effect of rmTBI or sex on righting reflex, and increased neurological deficits in rmTBI groups in both studies. In Study 1, rmTBI increased alcohol consumption in female but not male rats. Male rats consistently exhibited higher levels of anxiety-like behavior than females. The rmTBI did not affect anxiety-like behavior 37-38 days post-injury. In Study 2, rmTBI once again increased alcohol consumption in female but not male rats, and repeated systemic treatment with JZL184 did not affect alcohol consumption. Also in Study 2, rmTBI increased anxiety-like behavior in males but not females and repeated systemic treatment with JZL184 produced an unexpected increase in anxiety-like behavior 6-8 days post-injury. In summary, rmTBI increased alcohol consumption in female rats, systemic JZL184 treatment did not alter alcohol consumption, and both rmTBI and systemic JZL184 treatment increased anxiety-like behavior 6-8 days post-injury in males but not females, highlighting robust sex differences in rmTBI effects.
Collapse
Affiliation(s)
- Alejandra Jacotte-Simancas
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Patricia E. Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Nicholas W. Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Alcohol and Drug of Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Southeast Louisiana VA Healthcare System, New Orleans, Louisiana, USA
| |
Collapse
|
7
|
Janković T, Pilipović K. Single Versus Repetitive Traumatic Brain Injury: Current Knowledge on the Chronic Outcomes, Neuropathology and the Role of TDP-43 Proteinopathy. Exp Neurobiol 2023; 32:195-215. [PMID: 37749924 PMCID: PMC10569144 DOI: 10.5607/en23008] [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: 02/16/2023] [Revised: 07/18/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most important causes of death and disability in adults and thus an important public health problem. Following TBI, secondary pathophysiological processes develop over time and condition the development of different neurodegenerative entities. Previous studies suggest that neurobehavioral changes occurring after a single TBI are the basis for the development of Alzheimer's disease, while repetitive TBI is considered to be a contributing factor for chronic traumatic encephalopathy development. However, pathophysiological processes that determine the evolvement of a particular chronic entity are still unclear. Human post-mortem studies have found combinations of amyloid, tau, Lewi bodies, and TAR DNA-binding protein 43 (TDP-43) pathologies after both single and repetitive TBI. This review focuses on the pathological changes of TDP-43 after single and repetitive brain traumas. Numerous studies have shown that TDP-43 proteinopathy noticeably occurs after repetitive head trauma. A relatively small number of available preclinical research on single brain injury are not in complete agreement with the results from the human samples, which makes it difficult to draw specific conclusions. Also, as TBI is considered a heterogeneous type of injury, different experimental trauma models and injury intensities may cause differences in the cascade of secondary injury, which should be considered in future studies. Experimental and post-mortem studies of TDP-43 pathobiology should be carried out, preferably in the same laboratories, to determine its involvement in the development of neurodegenerative conditions after one and repetitive TBI, especially in the context of the development of new therapeutic options.
Collapse
Affiliation(s)
- Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| |
Collapse
|
8
|
Bjorklund GR, Wong J, Brafman D, Bowser R, Stabenfeldt SE. Traumatic brain injury induces TDP-43 mislocalization and neurodegenerative effects in tissue distal to the primary injury site in a non-transgenic mouse. Acta Neuropathol Commun 2023; 11:137. [PMID: 37608352 PMCID: PMC10463884 DOI: 10.1186/s40478-023-01625-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/18/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury (TBI) initiates tissue and cellular damage to the brain that is immediately followed by secondary injury sequalae with delayed and continual damage. This secondary damage includes pathological processes that may contribute to chronic neurodegeneration and permanent functional and cognitive deficits. TBI is also associated with an increased risk of developing neurodegenerative diseases such as Alzheimer's disease (AD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS) as indicated by shared pathological features. For example, abnormalities in the TAR DNA-binding Protein 43 (TDP-43) that includes cytoplasmic mislocalization, cytosolic aggregation, and an increase in phosphorylation and ubiquitination are seen in up to 50% of FTD cases, up to 70% of AD cases, and is considered a hallmark pathology of ALS occurring in > 97% of cases. Yet the prevalence of TDP-43 pathology post-TBI has yet to be fully characterized. Here, we employed a non-transgenic murine controlled cortical injury model of TBI and observed injury-induced hallmark TDP-43 pathologies in brain and spinal cord tissue distal to the primary injury site and did not include the focally damaged tissue within the primary cortical injury site. Analysis revealed a temporal-dependent and significant increase in neuronal TDP-43 mislocalization in the cortical forebrain rostral to and distant from the primary injury site up to 180 days post injury (DPI). TDP-43 mislocalization was also detected in neurons located in the ventral horns of the cervical spinal cord following a TBI. Moreover, a cortical layer-dependent affect was identified, increasing from superficial to deeper cortical layers over time from 7 DPI up to 180 DPI. Lastly, RNAseq analysis confirmed an injury-induced misregulation of several key biological processes implicated in neurons that increased over time. Collectively, this study demonstrates a connection between a single moderate TBI event and chronic neurodegenerative processes that are not limited to the primary injury site and broadly distributed throughout the cortex and corticospinal tract.
Collapse
Affiliation(s)
- George R Bjorklund
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jennifer Wong
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - David Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Robert Bowser
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
9
|
Kalbfell RM, Rettke DJ, Mackie K, Ejima K, Harezlak J, Alexander IL, Wager-Miller J, Johnson BD, Newman SD, Kawata K. The modulatory role of cannabis use in subconcussive neural injury. iScience 2023; 26:106948. [PMID: 37332596 PMCID: PMC10275955 DOI: 10.1016/j.isci.2023.106948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 01/31/2023] [Accepted: 05/20/2023] [Indexed: 06/20/2023] Open
Abstract
Cannabis use has become popular among athletes, many of whom are exposed to repetitive subconcussive head impacts. We aimed to test whether chronic cannabis use would be neuroprotective or exacerbating against acute subconcussive head impacts. This trial included 43 adult soccer players (Cannabis group using cannabis at least once a week for the past 6 months, n = 24; non-cannabis control group, n = 19). Twenty soccer headings, induced by our controlled heading model, significantly impaired ocular-motor function, but the degrees of impairments were less in the cannabis group compared to controls. The control group significantly increased its serum S100B level after heading, whereas no change was observed in the cannabis group. There was no group difference in serum neurofilament light levels at any time point. Our data suggest that chronic cannabis use may be associated with an enhancement of oculomotor functional resiliency and suppression of the neuroinflammatory response following 20 soccer headings.
Collapse
Affiliation(s)
- Rachel M. Kalbfell
- Department of Kinesiology, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
- Bioethics Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Devin J. Rettke
- Department of Kinesiology, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Ken Mackie
- Department of Psychological and Brain Sciences and Gill Center for Molecular Bioscience, The College of Arts and Sciences, Indiana University, Bloomington, IN, USA
- Program in Neuroscience, The College of Arts and Sciences, Indiana University, Bloomington, IN, USA
| | - Keisuke Ejima
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jaroslaw Harezlak
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Isabella L. Alexander
- Department of Kinesiology, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Jim Wager-Miller
- Department of Psychological and Brain Sciences and Gill Center for Molecular Bioscience, The College of Arts and Sciences, Indiana University, Bloomington, IN, USA
| | - Blair D. Johnson
- Department of Kinesiology, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Sharlene D. Newman
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL, USA
| | - Keisuke Kawata
- Department of Kinesiology, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
- Program in Neuroscience, The College of Arts and Sciences, Indiana University, Bloomington, IN, USA
| |
Collapse
|
10
|
Jacotte-Simancas A, Molina P, Gilpin N. JZL184 increases anxiety-like behavior and does not reduce alcohol consumption in female rats after repeated mild traumatic brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542943. [PMID: 37398130 PMCID: PMC10312513 DOI: 10.1101/2023.05.30.542943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Alcohol use disorder (AUD) is highly comorbid with traumatic brain injury (TBI). Previously, using a lateral fluid percussion model (LFP) (an open model of head injury) to generate a single mild to moderate traumatic brain injury (TBI), we showed that TBI produces escalation in alcohol drinking, that alcohol exposure negatively impacts TBI outcomes, and that the endocannabinoid degradation inhibitor (JZL184) confers significant protection from behavioral and neuropathological outcomes in male rodents. In the present study, we used a weight drop model (a closed model of head injury) to produce a repeated mild TBI (rmTBI, 3 TBIs, spaced by 24 hours) to examine the sex-specific effects on alcohol consumption and anxiety-like behavior in rats, and whether systemic treatment with JZL184 would reverse TBI effects on those behaviors in both sexes. In two separate studies, adult male and female Wistar rats were subjected to rmTBI or sham using the weight drop model. Physiological measures of injury severity were collected from all animals. Animals in both studies were allowed to consume alcohol using an intermittent 2-bottle choice procedure (12 pre-TBI sessions and 12 post-TBI sessions). Neurological severity and neurobehavioral scores (NSS and NBS, respectively) were tested 24 hours after the final injury. Anxiety-like behavior was tested at 37-38 days post-injury in Study 1, and 6-8 days post-injury in Study 2. Our results show that females exhibited reduced respiratory rates relative to males with no significant differences between Sham and rmTBI, no effect of rmTBI or sex on righting reflex, and increased neurological deficits in rmTBI groups in both studies. In Study 1, rmTBI increased alcohol consumption in female but not male rats. Male rats consistently exhibited higher levels of anxiety-like behavior than females. rmTBI did not affect anxiety-like behavior 37-38 days post-injury. In Study 2, rmTBI once again increased alcohol consumption in female but not male rats, and repeated systemic treatment with JZL184 did not affect alcohol consumption. Also in Study 2, rmTBI increased anxiety-like behavior in males but not females and repeated systemic treatment with JZL184 produced an unexpected increase in anxiety-like behavior 6-8 days post-injury. In summary, rmTBI increased alcohol consumption in female rats, systemic JZL184 treatment did not alter alcohol consumption, and both rmTBI and sub-chronic systemic JZL184 treatment increased anxiety-like behavior 6-8 days post-injury in males but not females, highlighting robust sex differences in rmTBI effects.
Collapse
Affiliation(s)
- Alejandra Jacotte-Simancas
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA
- Alcohol and Drug of Abuse Center of Excellence, LSUHSC, New Orleans, LA
| | - Patricia Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA
- Alcohol and Drug of Abuse Center of Excellence, LSUHSC, New Orleans, LA
| | - Nicholas Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA
- Alcohol and Drug of Abuse Center of Excellence, LSUHSC, New Orleans, LA
- Southeast Louisiana VA Healthcare System, New Orleans, LA
| |
Collapse
|
11
|
Chen C. Inhibiting degradation of 2-arachidonoylglycerol as a therapeutic strategy for neurodegenerative diseases. Pharmacol Ther 2023; 244:108394. [PMID: 36966972 PMCID: PMC10123871 DOI: 10.1016/j.pharmthera.2023.108394] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Endocannabinoids are endogenous lipid signaling mediators that participate in a variety of physiological and pathological processes. 2-Arachidonoylglycerol (2-AG) is the most abundant endocannabinoid and is a full agonist of G-protein-coupled cannabinoid receptors (CB1R and CB2R), which are targets of Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive ingredient in cannabis. While 2-AG has been well recognized as a retrograde messenger modulating synaptic transmission and plasticity at both inhibitory GABAergic and excitatory glutamatergic synapses in the brain, growing evidence suggests that 2-AG also functions as an endogenous terminator of neuroinflammation in response to harmful insults, thus maintaining brain homeostasis. Monoacylglycerol lipase (MAGL) is the key enzyme that degrades 2-AG in the brain. The immediate metabolite of 2-AG is arachidonic acid (AA), a precursor of prostaglandins (PGs) and leukotrienes. Several lines of evidence indicate that pharmacological or genetic inactivation of MAGL, which boosts 2-AG levels and reduces its hydrolytic metabolites, resolves neuroinflammation, mitigates neuropathology, and improves synaptic and cognitive functions in animal models of neurodegenerative diseases, including Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and traumatic brain injury (TBI)-induced neurodegenerative disease. Thus, it has been proposed that MAGL is a potential therapeutic target for treatment of neurodegenerative diseases. As the main enzyme hydrolyzing 2-AG, several MAGL inhibitors have been identified and developed. However, our understanding of the mechanisms by which inactivation of MAGL produces neuroprotective effects in neurodegenerative diseases remains limited. A recent finding that inhibition of 2-AG metabolism in astrocytes, but not in neurons, protects the brain from TBI-induced neuropathology might shed some light on this unsolved issue. This review provides an overview of MAGL as a potential therapeutic target for neurodegenerative diseases and discusses possible mechanisms underlying the neuroprotective effects of restraining degradation of 2-AG in the brain.
Collapse
|
12
|
Zhu D, Zhang J, Gao F, Hu M, Hashem J, Chen C. Augmentation of 2-arachidonoylglycerol signaling in astrocytes maintains synaptic functionality by regulation of miRNA-30b. Exp Neurol 2023; 361:114292. [PMID: 36481187 PMCID: PMC9892245 DOI: 10.1016/j.expneurol.2022.114292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
2-Arachidonoylglycerol (2-AG), the most abundant endocannabinoid, displays anti-inflammatory and neuroprotective properties. Inhibition of 2-AG degradation by inactivation of monoacylglycerol lipase (MAGL), a key enzyme degrading 2-AG in the brain, alleviates neuropathology and improves synaptic and cognitive functions in animal models of neurodegenerative diseases. In particular, global inactivation of MAGL by genetic deletion of mgll enhances hippocampal long-term potentiation (LTP) and hippocampus-dependent learning and memory. However, our understanding of the molecular mechanisms by which chronic inactivation of MAGL enhances synaptic activity is still limited. Here, we provide evidence that pharmacological inactivation of MAGL suppresses hippocampal expression of miR-30b, a small non-coding microRNA, and upregulates expression of its targets, including ephrin type-B receptor 2 (ephB2), sirtuin1 (sirt1), and glutamate ionotropic receptor AMPA type subunit 2 (GluA2). Importantly, suppression of miR-30b and increase of its targets by inactivation of MAGL result primarily from inhibition of 2-AG metabolism in astrocytes, rather than in neurons. Inactivation of MAGL in astrocytes prevents miR-30b overexpression-induced impairments in synaptic transmission and long-term potentiation (LTP) in the hippocampus. Suppression of miR-30b expression by inactivation of MAGL is apparently associated with augmentation of 2-AG signaling, as 2-AG induces a dose-dependent decrease in expression of miR-30b. 2-AG- or MAGL inactivation-suppressed expression of miR-30b is not mediated via CB1R, but by peroxisome proliferator-activated receptor γ (PPARγ). This is further supported by the results showing that MAGL inactivation-induced downregulation of miR-30b and upregulation of its targets are attenuated by antagonism of PPARγ, but mimicked by PPARγ agonists. In addition, we observed that 2-AG-induced reduction of miR-30b expression is mediated via NF-kB signaling. Our study provides evidence that 2-AG signaling in astrocytes plays an important role in maintaining the functional integrity of synapses in the hippocampus by regulation of miR-30b expression.
Collapse
Affiliation(s)
- Dexiao Zhu
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Jian Zhang
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Fei Gao
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Mei Hu
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Jack Hashem
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Chu Chen
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
13
|
Aychman MM, Goldman DL, Kaplan JS. Cannabidiol's neuroprotective properties and potential treatment of traumatic brain injuries. Front Neurol 2023; 14:1087011. [PMID: 36816569 PMCID: PMC9932048 DOI: 10.3389/fneur.2023.1087011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Cannabidiol (CBD) has numerous pharmacological targets that initiate anti-inflammatory, antioxidative, and antiepileptic properties. These neuroprotective benefits have generated interest in CBD's therapeutic potential against the secondary injury cascade from traumatic brain injury (TBI). There are currently no effective broad treatment strategies for combating the damaging mechanisms that follow the primary injury and lead to lasting neurological consequences or death. However, CBD's effects on different neurotransmitter systems, the blood brain barrier, oxidative stress mechanisms, and the inflammatory response provides mechanistic support for CBD's clinical utility in TBI. This review describes the cascades of damage caused by TBI and CBD's neuroprotective mechanisms to counter them. We also present challenges in the clinical treatment of TBI and discuss important future clinical research directions for integrating CBD in treatment protocols. The mechanistic evidence provided by pre-clinical research shows great potential for CBD as a much-needed improvement in the clinical treatment of TBI. Upcoming clinical trials sponsored by major professional sport leagues are the first attempts to test the efficacy of CBD in head injury treatment protocols and highlight the need for further clinical research.
Collapse
|
14
|
Zhu D, Zhang J, Hashem J, Gao F, Chen C. Inhibition of 2-arachidonoylglycerol degradation enhances glial immunity by single-cell transcriptomic analysis. J Neuroinflammation 2023; 20:17. [PMID: 36717883 PMCID: PMC9885699 DOI: 10.1186/s12974-023-02701-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND 2-Arachidonoylglycerol (2-AG) is the most abundant endogenous cannabinoid. Inhibition of 2-AG metabolism by inactivation of monoacylglycerol lipase (MAGL), the primary enzyme that degrades 2-AG in the brain, produces anti-inflammatory and neuroprotective effects in neurodegenerative diseases. However, the molecular mechanisms underlying these beneficial effects are largely unclear. METHODS Hippocampal and cortical cells were isolated from cell type-specific MAGL knockout (KO) mice. Single-cell RNA sequencing was performed by 10 × Genomics platform. Cell Ranger, Seurat (v3.2) and CellChat (1.1.3) packages were used to carry out data analysis. RESULTS Using single-cell RNA sequencing analysis, we show here that cell type-specific MAGL KO mice display distinct gene expression profiles in the brain. Inactivation of MAGL results in robust changes in expression of immune- and inflammation-related genes in microglia and astrocytes. Remarkably, upregulated expression of chemokines in microglia is more pronounced in mice lacking MAGL in astrocytes. In addition, expression of genes that regulate other cellular functions and Wnt signaling in astrocytes is altered in MAGL KO mice. CONCLUSIONS Our results provide transcriptomic evidence that cell type-specific inactivation of MAGL induces differential expression of immune-related genes and other fundamental cellular pathways in microglia and astrocytes. Upregulation of the immune/inflammatory genes suggests that tonic levels of immune/inflammatory vigilance are enhanced in microglia and astrocytes, particularly in microglia, by inhibition of 2-AG metabolism, which likely contribute to anti-inflammatory and neuroprotective effects produced by inactivation of MAGL in neurodegenerative diseases.
Collapse
Affiliation(s)
- Dexiao Zhu
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Jian Zhang
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Jack Hashem
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Fei Gao
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA
| | - Chu Chen
- grid.267309.90000 0001 0629 5880Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229 USA ,grid.267309.90000 0001 0629 5880Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| |
Collapse
|
15
|
Tanaka M, Zhang Y. Preclinical Studies of Posttraumatic Headache and the Potential Therapeutics. Cells 2022; 12:cells12010155. [PMID: 36611947 PMCID: PMC9818317 DOI: 10.3390/cells12010155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/13/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Posttraumatic headache (PTH) attributed to traumatic brain injury (TBI) is a secondary headache developed within 7 days after head injury, and in a substantial number of patients PTH becomes chronic and lasts for more than 3 months. Current medications are almost entirely relied on the treatment of primary headache such as migraine, due to its migraine-like phenotype and the limited understanding on the PTH pathogenic mechanisms. To this end, increasing preclinical studies have been conducted in the last decade. We focus in this review on the trigeminovascular system from the animal studies since it provides the primary nociceptive sensory afferents innervating the head and face region, and the pathological changes in the trigeminal pathway are thought to play a key role in the development of PTH. In addition to the pathologies, PTH-like behaviors induced by TBI and further exacerbated by nitroglycerin, a general headache inducer through vasodilation are reviewed. We will overview the current pharmacotherapies including calcitonin gene-related peptide (CGRP) monoclonal antibody and sumatriptan in the PTH animal models. Given that modulation of the endocannabinoid (eCB) system has been well-documented in the treatment of migraine and TBI, the therapeutic potential of eCB in PTH will also be discussed.
Collapse
|
16
|
Chen C. Endocannabinoid control of neuroinflammation in traumatic brain injury by monoacylglycerol lipase in astrocytes. Neural Regen Res 2022; 18:1023-1024. [PMID: 36254984 PMCID: PMC9827788 DOI: 10.4103/1673-5374.355755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Chu Chen
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA,Correspondence to: Chu Chen, or .
| |
Collapse
|
17
|
Jiang S, Zheng C, Wen G, Bu B, Zhao S, Xu X. Down-regulation of NR2B receptors contributes to the analgesic and antianxiety effects of enriched environment mediated by endocannabinoid system in the inflammatory pain mice. Behav Brain Res 2022; 435:114062. [PMID: 35985400 DOI: 10.1016/j.bbr.2022.114062] [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: 01/12/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022]
Abstract
Chronic pain states are highly prevalent and yet poorly controlled by currently available analgesics. It has been reported that enriched environment (EE), as a new way of endogenous pharmacotherapy, is effective in attenuating chronic inflammatory pain. However, the underlying molecular mechanisms are still not fully understood. NMDA NR2B receptor plays a critical role in pain transmission and modulation. Thus, in this study, we aimed at the effect of EE on the NR2B receptors expression in the prefrontal cortex, hippocampus and thalamus in the inflammatory pain mice. The results showed a significant increase of NR2B receptors in the thalamus of mice at 7 d following injection of CFA in the subcutaneous of the bottom of the left hind paw. EE significantly reduced the duration of mechanical hypersensitivity and anxiety-related behavior and the expression of NR2B receptors as compared to the standard condition. Furthermore, EE significantly increased 2-arachidonoylglycero (2-AG) levels at 7 d in the inflammatory pain mice as compared to the standard condition, and the effect of EE on the behavior and the expression of NR2B receptors was abolished by intraperitoneal injection of AM281 (a selective antagonist of CB1 receptor). Elevated 2-AG levels by intraperitoneal injection of JZL184 (a selective inhibitor of MAGL, the enzyme responsible for 2-AG hydrolysis) produced the same effect as EE. Results from this study provide the evidence that EE mimics endocannabinoids to take analgesic and anti-anxiety activities by decreasing the expression of the NR2B receptors via the CB1 receptor in the thalamus, pending further studies.
Collapse
Affiliation(s)
- Shukun Jiang
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China; Key Laboratory of Forensic Bio-evidence Sciences, Liaoning Province, PR China; China Medical University Center of Forensic Investigation, Shenyang, PR China
| | - Chuanfei Zheng
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China; Key Laboratory of Forensic Bio-evidence Sciences, Liaoning Province, PR China; China Medical University Center of Forensic Investigation, Shenyang, PR China
| | - Gehua Wen
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China; Key Laboratory of Forensic Bio-evidence Sciences, Liaoning Province, PR China; China Medical University Center of Forensic Investigation, Shenyang, PR China
| | - Bin Bu
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China; Key Laboratory of Forensic Bio-evidence Sciences, Liaoning Province, PR China; China Medical University Center of Forensic Investigation, Shenyang, PR China
| | - Shuang Zhao
- China Medical University Center of Forensic Investigation, Shenyang, PR China
| | - Xiaoming Xu
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China; Key Laboratory of Forensic Bio-evidence Sciences, Liaoning Province, PR China; China Medical University Center of Forensic Investigation, Shenyang, PR China.
| |
Collapse
|
18
|
TDP-43 drives synaptic and cognitive deterioration following traumatic brain injury. Acta Neuropathol 2022; 144:187-210. [PMID: 35713704 PMCID: PMC9945325 DOI: 10.1007/s00401-022-02449-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 11/01/2022]
Abstract
Traumatic brain injury (TBI) has been recognized as an important risk factor for Alzheimer's disease (AD). However, the molecular mechanisms by which TBI contributes to developing AD remain unclear. Here, we provide evidence that aberrant production of TDP-43 is a key factor in promoting AD neuropathology and synaptic and cognitive deterioration in mouse models of mild closed head injury (CHI). We observed that a single mild CHI is sufficient to exacerbate AD neuropathology and accelerate synaptic and cognitive deterioration in APP transgenic mice but repeated mild CHI are required to induce neuropathological changes and impairments in synaptic plasticity, spatial learning, and memory retention in wild-type animals. Importantly, these changes in animals exposed to a single or repeated mild CHI are alleviated by silencing of TDP-43 but reverted by rescue of the TDP-43 knockdown. Moreover, overexpression of TDP-43 in the hippocampus aggravates AD neuropathology and provokes cognitive impairment in APP transgenic mice, mimicking single mild CHI-induced changes. We further discovered that neuroinflammation triggered by TBI promotes NF-κB-mediated transcription and expression of TDP-43, which in turn stimulates tau phosphorylation and Aβ formation. Our findings suggest that excessive production of TDP-43 plays an important role in exacerbating AD neuropathology and in driving synaptic and cognitive declines following TBI.
Collapse
|
19
|
Lim SH, Jung H, Youn DH, Kim TY, Han SW, Kim BJ, Lee JJ, Jeon JP. Mild Traumatic Brain Injury and Subsequent Acute Pulmonary Inflammatory Response. J Korean Neurosurg Soc 2022; 65:680-687. [PMID: 35574585 PMCID: PMC9452391 DOI: 10.3340/jkns.2021.0310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/21/2022] [Indexed: 11/27/2022] Open
Abstract
Objective The influence of moderate-to-severe traumatic brain injury (TBI) on acute pulmonary injury is well established, but the association between acute pulmonary injury and mild TBI has not been well studied. Here, we evaluated the histological changes and fluctuations in inflammatory markers in the lungs to determine whether an acute pulmonary inflammatory response occurred after mild TBI.
Methods Mouse models of mild TBI (n=24) were induced via open-head injuries using a stereotaxic impactor. The brain and lungs were examined 6, 24, and 72 hours after injury and compared to sham-operated controls (n=24). Fluoro-Jade B staining and Astra blue and hematoxylin staining were performed to assess cerebral neuronal degeneration and pulmonary histological architecture. Quantitative real-time polymerase chain reaction analysis was done to measure inflammatory cytokines.
Results Increased neuronal degeneration and the mRNA expression of interleukin (IL)-6, tumor necrosis factor (TNF)-α, IL-10, and transforming growth factor (TGF)-β were observed after mild TBI. The IL-6, TNF-α, and TGF-β levels in mice with mild TBI were significantly different compared to those of sham-operated mice 24 hours after injury, and this was more pronounced at 72 hours. Mild TBI induced acute pulmonary interstitial edema with cell infiltration and alveolar morphological changes. In particular, a significant infiltration of mast cells was observed. Among the inflammatory cytokines, TNF-α was significantly increased in the lungs at 6 hours, but there was no significant difference 24 and 72 hours after injury.
Conclusion Mild TBI induced acute pulmonary interstitial inflammation and alveolar structural changes, which are likely to worsen the patient’s prognosis.
Collapse
|
20
|
Hu M, Zhu D, Zhang J, Gao F, Hashem J, Kingsley P, Marnett LJ, Mackie K, Chen C. Enhancing endocannabinoid signalling in astrocytes promotes recovery from traumatic brain injury. Brain 2022; 145:179-193. [PMID: 35136958 PMCID: PMC8967103 DOI: 10.1093/brain/awab310] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/24/2021] [Accepted: 07/26/2021] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury is an important risk factor for development of Alzheimer's disease and dementia. Unfortunately, no effective therapies are currently available for prevention and treatment of the traumatic brain injury-induced Alzheimer's disease-like neurodegenerative disease. This is largely due to our limited understanding of the mechanisms underlying traumatic brain injury-induced neuropathology. Previous studies showed that pharmacological inhibition of monoacylglycerol lipase, a key enzyme degrading the endocannabinoid 2-arachidonoylglycerol, attenuates traumatic brain injury-induced neuropathology. However, the mechanism responsible for the neuroprotective effects produced by inhibition of monoacylglycerol lipase in traumatic brain injury remains unclear. Here we first show that genetic deletion of monoacylglycerol lipase reduces neuropathology and averts synaptic and cognitive declines in mice exposed to repeated mild closed head injury. Surprisingly, these neuroprotective effects result primarily from inhibition of 2-arachidonoylglycerol metabolism in astrocytes, rather than in neurons. Single-cell RNA-sequencing data reveal that astrocytic monoacylglycerol lipase knockout mice display greater resilience to traumatic brain injury-induced changes in expression of genes associated with inflammation or maintenance of brain homeostasis in astrocytes and microglia. The monoacylglycerol lipase inactivation-produced neuroprotection is abrogated by deletion of the cannabinoid receptor-1 or by adeno-associated virus vector-mediated silencing of astrocytic peroxisome proliferator-activated receptor-γ. This is further supported by the fact that overexpression of peroxisome proliferator-activated receptor-γ in astrocytes prevents traumatic brain injury-induced neuropathology and impairments in spatial learning and memory. Our results reveal a previously undefined cell type-specific role of 2-arachidonoylglycerol metabolism and signalling pathways in traumatic brain injury-induced neuropathology, suggesting that enhanced 2-arachidonoylglycerol signalling in astrocytes is responsible for the monoacylglycerol lipase inactivation-produced alleviation of neuropathology and deficits in synaptic and cognitive functions in traumatic brain injury.
Collapse
Affiliation(s)
- Mei Hu
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Dexiao Zhu
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jian Zhang
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Fei Gao
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jack Hashem
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Philip Kingsley
- Departments of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lawrence J Marnett
- Departments of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Chu Chen
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA,Correspondence to: Chu Chen, PhD Department of Cellular and Integrative Physiology, School of Medicine University of Texas Health Science Center at San Antonio 7703 Floyd Curl Drive, San Antonio, TX 78229, USA E-mail: or
| |
Collapse
|
21
|
OUP accepted manuscript. Brain 2022; 145:7-10. [DOI: 10.1093/brain/awac014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 11/12/2022] Open
|
22
|
Selvaraj P, Tanaka M, Wen J, Zhang Y. The Novel Monoacylglycerol Lipase Inhibitor MJN110 Suppresses Neuroinflammation, Normalizes Synaptic Composition and Improves Behavioral Performance in the Repetitive Traumatic Brain Injury Mouse Model. Cells 2021; 10:cells10123454. [PMID: 34943962 PMCID: PMC8700188 DOI: 10.3390/cells10123454] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
Modulation of the endocannabinoid system has emerged as an effective approach for the treatment of many neurodegenerative and neuropsychological diseases. However, the underlying mechanisms are still uncertain. Using a repetitive mild traumatic brain injury (mTBI) mouse model, we found that there was an impairment in locomotor function and working memory within two weeks post-injury, and that treatment with MJN110, a novel inhibitor of the principal 2-arachidononyl glycerol (2-AG) hydrolytic enzyme monoacylglycerol lipase dose-dependently ameliorated those behavioral changes. Spatial learning and memory deficits examined by Morris water maze between three and four weeks post-TBI were also reversed in the drug treated animals. Administration of MJN110 selectively elevated the levels of 2-AG and reduced the production of arachidonic acid (AA) and prostaglandin E2 (PGE2) in the TBI mouse brain. The increased production of proinflammatory cytokines, accumulation of astrocytes and microglia in the TBI mouse ipsilateral cerebral cortex and hippocampus were significantly reduced by MJN110 treatment. Neuronal cell death was also attenuated in the drug treated animals. MJN110 treatment normalized the expression of the NMDA receptor subunits NR2A and NR2B, the AMPA receptor subunits GluR1 and GluR2, and the GABAA receptor subunits α1, β2,3 and γ2, which were all reduced at 1, 2 and 4 weeks post-injury. The reduced inflammatory response and restored glutamate and GABA receptor expression likely contribute to the improved motor function, learning and memory in the MJN110 treated animals. The therapeutic effects of MJN110 were partially mediated by activation of CB1 and CB2 cannabinoid receptors and were eliminated when it was co-administered with DO34, a novel inhibitor of the 2-AG biosynthetic enzymes. Our results suggest that augmentation of the endogenous levels of 2-AG can be therapeutically useful in the treatment of TBI by suppressing neuroinflammation and maintaining the balance between excitatory and inhibitory neurotransmission.
Collapse
Affiliation(s)
- Prabhuanand Selvaraj
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
| | - Jie Wen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +1-301-295-3212
| |
Collapse
|
23
|
Zhu D, Gao F, Chen C. Endocannabinoid Metabolism and Traumatic Brain Injury. Cells 2021; 10:cells10112979. [PMID: 34831202 PMCID: PMC8616221 DOI: 10.3390/cells10112979] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Traumatic brain injury (TBI) represents a major cause of morbidity and disability and is a risk factor for developing neurodegenerative diseases, including Alzheimer’s disease (AD). However, no effective therapies are currently available for TBI-induced AD-like disease. Endocannabinoids are endogenous lipid mediators involved in a variety of physiological and pathological processes. The compound 2-arachidonoylglycerol (2-AG) is the most abundant endocannabinoid with profound anti-inflammatory and neuroprotective properties. This molecule is predominantly metabolized by monoacylglycerol lipase (MAGL), a key enzyme degrading about 85% of 2-AG in the brain. Studies using animal models of inflammation, AD, and TBI provide evidence that inactivation of MAGL, which augments 2-AG signaling and reduces its metabolites, exerts neuroprotective effects, suggesting that MAGL is a promising therapeutic target for neurodegenerative diseases. In this short review, we provide an overview of the inhibition of 2-AG metabolism for the alleviation of neuropathology and the improvement of synaptic and cognitive functions after TBI.
Collapse
|
24
|
Saulino PA, Greenwald BD, Gordon DJ. The changing landscape of the use of medical marijuana after traumatic brain injury: a narrative review. Brain Inj 2021; 35:1510-1520. [PMID: 34632896 DOI: 10.1080/02699052.2021.1978548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To summarize the potential therapeutic benefits of medical marijuana for patients with traumatic brain injury (TBI). METHODS A systematic search was conducted using PubMed and Cochran's library for information regard the safety and efficacy of medical marijuana as a therapeutic agent. We investigated, in depth, articles specifically evaluating medical marijuana's use in TBI, as well as articles that summarized the effects of marijuana in general. Articles from the year 2000-2020 were included. RESULTS A total of 37 articles met our inclusion criteria. An additional 3 articles were obtained from reference lists. CONCLUSION Studies have shown that medical marijuana can potentially aid the recovery from TBI by modulating the endocannabinoid system, reducing inflammation and secondary injury. Adverse cognitive and physiological effects have been observed in the acute setting as well as chronically, though more research is necessitated. There is also the concern of significant drug-drug interactions that have not been thoroughly studied. Thus, while there is evidence that medical marijuana can be beneficial in the treatment of TBI, more research is necessitated to fully explore the long-term efficacy and adverse effects.
Collapse
Affiliation(s)
- Patrick A Saulino
- Rutgers Robert Wood Johnson Medical School, Ringgold Standard Institution, Piscataway, New Jersey, USA
| | - Brian D Greenwald
- Center for Brain Injuries, JFK Johnson Rehabilitation Institute, Ringgold Standard Institution - Physical Medicine and Rehabilitation, Edison, New Jersey, USA.,Rutgers Robert Wood Johnson Medical School New Brunswick, - Physical Medicine and Rehabilitation, Edison, New Jersey, USA
| | - Dustin J Gordon
- Rehabilitation Specialists, Ringgold Standard Institution, Fairleigh Dickinson University, Fair Lawn, New Jersey, USA.,Fairleigh Dickinson University in Teaneck, New Jersey, USA
| |
Collapse
|
25
|
Zanfirescu A, Ungurianu A, Mihai DP, Radulescu D, Nitulescu GM. Targeting Monoacylglycerol Lipase in Pursuit of Therapies for Neurological and Neurodegenerative Diseases. Molecules 2021; 26:5668. [PMID: 34577139 PMCID: PMC8468992 DOI: 10.3390/molecules26185668] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Neurological and neurodegenerative diseases are debilitating conditions, and frequently lack an effective treatment. Monoacylglycerol lipase (MAGL) is a key enzyme involved in the metabolism of 2-AG (2-arachidonoylglycerol), a neuroprotective endocannabinoid intimately linked to the generation of pro- and anti-inflammatory molecules. Consequently, synthesizing selective MAGL inhibitors has become a focus point in drug design and development. The purpose of this review was to summarize the diverse synthetic scaffolds of MAGL inhibitors concerning their potency, mechanisms of action and potential therapeutic applications, focusing on the results of studies published in the past five years. The main irreversible inhibitors identified were derivatives of hexafluoroisopropyl alcohol carbamates, glycol carbamates, azetidone triazole ureas and benzisothiazolinone, whereas the most promising reversible inhibitors were derivatives of salicylketoxime, piperidine, pyrrolidone and azetidinyl amides. We reviewed the results of in-depth chemical, mechanistic and computational studies on MAGL inhibitors, in addition to the results of in vitro findings concerning selectivity and potency of inhibitors, using the half maximal inhibitory concentration (IC50) as an indicator of their effect on MAGL. Further, for highlighting the potential usefulness of highly selective and effective inhibitors, we examined the preclinical in vivo reports regarding the promising therapeutic applications of MAGL pharmacological inhibition.
Collapse
Affiliation(s)
| | - Anca Ungurianu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (A.Z.); (D.P.M.); (D.R.); (G.M.N.)
| | | | | | | |
Collapse
|
26
|
Youn DH, Tran NM, Kim BJ, Kim Y, Jeon JP, Yoo H. Shape effect of cerium oxide nanoparticles on mild traumatic brain injury. Sci Rep 2021; 11:15571. [PMID: 34330990 PMCID: PMC8324865 DOI: 10.1038/s41598-021-95057-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
The catalytic performance and therapeutic effect of nanoparticles varies with shape. Here, we investigated and compared the therapeutic outcomes of ceria nanospheres (Ceria NSs) and ceria nanorods (Ceria NRs) in an in vivo study of mild traumatic brain injury (mTBI). In vivo TBI was induced in a mouse model of open head injury using a stereotaxic impactor. Outcomes including cytoprotective effects, cognitive function, and cerebral edema were investigated after retro-orbital injection of 11.6 mM of ceria nanoparticles. Ceria nanoparticles significantly reduced fluoro-jade B (FJB)-positive cells and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL)-positive cells, and restored mRNA levels of superoxide dismutase 1 (SOD1) and SOD2. They also decreased the cyclooxygenase-2 (COX-2) expression compared with the untreated control group. Comparing the two nanomaterials, Ceria NRs showed less stable and high-energy (100) and (110) planes, which increased the number of active sites. The Ce3+/Ce4+ molar ratio of Ceria NRs (0.40) was greater than that of Ceria NSs (0.27). Ceria NRs (0.059 ± 0.021) appeared to exhibit better anti-inflammatory effect than Ceria NSs (0.133 ± 0.024), but the effect was statistically insignificant (p = 0.190). Ceria nanoparticles also improved cognitive impairment following mTBI compared with the control group, but the effect did not differ significantly according to the nanoshape. However, Ceria NRs (70.1 ± 0.5%) significantly decreased brain water content compared with Ceria NSs (73.7 ± 0.4%; p = 0.0015), indicating a more effective reduction in brain edema (p = 0.0015). Compared with Ceria NSs, the Ceria NRs are more effective in alleviating cerebral edema following in vivo mTBI.
Collapse
Affiliation(s)
- Dong Hyuk Youn
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Ngoc Minh Tran
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Bong Jun Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Youngmi Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Jin Pyeong Jeon
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Republic of Korea.
- Genetic and Research Inc., Chuncheon, Republic of Korea.
- Department of Neurosurgery, Hallym University College of Medicine, 77 Sakju-ro, Chuncheon, 24253, Republic of Korea.
| | - Hyojong Yoo
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea.
| |
Collapse
|
27
|
Rajič Bumber J, Pilipović K, Janković T, Dolenec P, Gržeta N, Križ J, Župan G. Repetitive Traumatic Brain Injury Is Associated With TDP-43 Alterations, Neurodegeneration, and Glial Activation in Mice. J Neuropathol Exp Neurol 2021; 80:2-14. [PMID: 33212475 DOI: 10.1093/jnen/nlaa130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence points to a relationship between repetitive mild traumatic brain injury (mTBI), the Tar DNA binding protein 43 (TDP-43) pathology and some neurodegenerative diseases, but the underlying pathophysiological mechanisms are still unknown. We examined TDP-43 regulation, neurodegeneration, and glial responses following repetitive mTBI in nontransgenic mice and in animals with overexpression of human mutant TDP-43 protein (TDP-43G348C). In the frontal cortices of the injured nontransgenic animals, early TDP-43 cytoplasmatic translocation and overexpression of the protein and its pathological forms were detected. In the injured animals of both genotypes, neurodegeneration and pronounced glial activity were detected in the optic tract. In TDP-43G348C mice, these changes were significantly higher at day 7 after the last mTBI compared with the values in the nontransgenic animals. Results of this study suggest that the changes in the TDP-43 regulation in the frontal cortices of the nontransgenic animals were a transient stress response to the brain injury. Repetitive mTBI did not produce additional TDP-43 dysregulation or neurodegeneration or pronounced gliosis in the frontal cortex of TDP-43G348C mice. Our research also suggests that overexpression of mutated human TDP-43 possibly predisposes the brain to more intense neurodegeneration and glial activation in the optic tract after repetitive mTBI.
Collapse
Affiliation(s)
- Jelena Rajič Bumber
- From the Department of Pharmacology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Kristina Pilipović
- From the Department of Pharmacology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Tamara Janković
- From the Department of Pharmacology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Petra Dolenec
- From the Department of Pharmacology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Nika Gržeta
- From the Department of Pharmacology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jasna Križ
- Department of Psychiatry and Neuroscience, Faculty of Medicine, University of Laval, Quebec, QC, Canada
| | - Gordana Župan
- From the Department of Pharmacology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
28
|
Pilipović K, Rajič Bumber J, Dolenec P, Gržeta N, Janković T, Križ J, Župan G. Long-Term Effects of Repetitive Mild Traumatic Injury on the Visual System in Wild-Type and TDP-43 Transgenic Mice. Int J Mol Sci 2021; 22:ijms22126584. [PMID: 34205342 PMCID: PMC8235442 DOI: 10.3390/ijms22126584] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 01/29/2023] Open
Abstract
Little is known about the impairments and pathological changes in the visual system in mild brain trauma, especially repetitive mild traumatic brain injury (mTBI). The goal of this study was to examine and compare the effects of repeated head impacts on the neurodegeneration, axonal integrity, and glial activity in the optic tract (OT), as well as on neuronal preservation, glial responses, and synaptic organization in the lateral geniculate nucleus (LGN) and superior colliculus (SC), in wild-type mice and transgenic animals with overexpression of human TDP-43 mutant protein (TDP-43G348C) at 6 months after repeated closed head traumas. Animals were also assessed in the Barnes maze (BM) task. Neurodegeneration, axonal injury, and gliosis were detected in the OT of the injured animals of both genotypes. In the traumatized mice, myelination of surviving axons was mostly preserved, and the expression of neurofilament light chain was unaffected. Repetitive mTBI did not induce changes in the LGN and the SC, nor did it affect the performance of the BM task in the traumatized wild-type and TDP-43 transgenic mice. Differences in neuropathological and behavioral assessments between the injured wild-type and TDP-43G348C mice were not revealed. Results of the current study suggest that repetitive mTBI was associated with chronic damage and inflammation in the OT in wild-type and TDP-43G348C mice, which were not accompanied with behavioral problems and were not affected by the TDP-43 genotype, while the LGN and the SC remained preserved in the used experimental conditions.
Collapse
Affiliation(s)
- Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000 Rijeka, Croatia; (K.P.); (J.R.B.); (P.D.); (N.G.); (T.J.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000 Rijeka, Croatia; (K.P.); (J.R.B.); (P.D.); (N.G.); (T.J.)
| | - Petra Dolenec
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000 Rijeka, Croatia; (K.P.); (J.R.B.); (P.D.); (N.G.); (T.J.)
| | - Nika Gržeta
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000 Rijeka, Croatia; (K.P.); (J.R.B.); (P.D.); (N.G.); (T.J.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000 Rijeka, Croatia; (K.P.); (J.R.B.); (P.D.); (N.G.); (T.J.)
| | - Jasna Križ
- Department of Psychiatry and Neuroscience, Faculty of Medicine, University Laval, Québec City, QC G1V 0A6, Canada;
| | - Gordana Župan
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51 000 Rijeka, Croatia; (K.P.); (J.R.B.); (P.D.); (N.G.); (T.J.)
- Correspondence:
| |
Collapse
|
29
|
O'Brien LD, Smith TL, Donvito G, Cravatt BF, Newton J, Spiegel S, Reeves TM, Phillips LL, Lichtman AH. Diacylglycerol Lipase-β Knockout Mice Display a Sex-Dependent Attenuation of Traumatic Brain Injury-Induced Mortality with No Impact on Memory or Other Functional Consequences. Cannabis Cannabinoid Res 2021; 6:508-521. [PMID: 34142866 DOI: 10.1089/can.2020.0175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Background: The endogenous cannabinoid system modulates inflammatory signaling in a variety of pathological states, including traumatic brain injury (TBI). The selective expression of diacylglycerol lipase-β (DAGL-β), the 2-arachidonylglycerol biosynthetic enzyme, on resident immune cells of the brain (microglia) and the role of this pathway in neuroinflammation, suggest that this enzyme may contribute to TBI-induced neuroinflammation. Accordingly, we tested whether DAGL-β-/- mice would show a protective phenotype from the deleterious consequences of TBI on cognitive and neurological motor functions. Materials and Methods: DAGL-β-/- and -β+/+ mice were subjected to the lateral fluid percussion model of TBI and assessed for learning and memory in the Morris water maze (MWM) Fixed Platform (reference memory) and Reversal (cognitive flexibility) tasks, as well as in a cued MWM task to infer potential sensorimotor/motivational deficits. In addition, subjects were assessed for motor behavior (Rotarod and the Neurological Severity Score assays) and in the light/dark box and the elevated plus maze to infer whether these manipulations affected anxiety-like behavior. Finally, we also examined whether brain injury disrupts the ceramide/sphingolipid lipid signaling system and if DAGL-β deletion offers protection. Results: TBI disrupted all measures of neurological motor function and reduced body weight, but did not affect body temperature or performance in common assays used to infer anxiety. TBI also impaired performance in MWM Fixed Platform and Reversal tasks, but did not affect cued MWM performance. Although no differences were found between DAGL-β-/- and -β+/+ mice in any of these measures, male DAGL-β-/- mice displayed an unexpected survival-protective phenotype, which persisted at increased injury severities. In contrast, TBI did not elicit mortality in female mice regardless of genotype. TBI also produced significant changes in sphingolipid profiles (a family of lipids, members of which have been linked to both apoptotic and antiapoptotic pathways), in which DAGL-β deletion modestly altered levels of select species. Conclusions: These findings indicate that although DAGL-β does not play a necessary role in TBI-induced cognitive and neurological function, it appears to contribute to the increased vulnerability of male mice to TBI-induced mortality, whereas female mice show high survival rates irrespective of DAGL-β expression.
Collapse
Affiliation(s)
- Lesley D O'Brien
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terry L Smith
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Giulia Donvito
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Benjamin F Cravatt
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Jason Newton
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Thomas M Reeves
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
30
|
Inhibition of 2-Arachidonoylglycerol Metabolism Alleviates Neuropathology and Improves Cognitive Function in a Tau Mouse Model of Alzheimer's Disease. Mol Neurobiol 2021; 58:4122-4133. [PMID: 33939165 DOI: 10.1007/s12035-021-02400-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, which affects more than 5 million individuals in the USA. Unfortunately, no effective therapies are currently available to prevent development of AD or to halt progression of the disease. It has been proposed that monoacylglycerol lipase (MAGL), the key enzyme degrading the endocannabinoid 2-arachidonoylglycerol (2-AG) in the brain, is a therapeutic target for AD based on the studies using the APP transgenic models of AD. While inhibition of 2-AG metabolism mitigates β-amyloid (Aβ) neuropathology, it is still not clear whether inactivation of MAGL alleviates tauopathies as accumulation and deposition of intracellular hyperphosphorylated tau protein are the neuropathological hallmark of AD. Here we show that JZL184, a potent MAGL inhibitor, significantly reduced proinflammatory cytokines, astrogliosis, phosphorylated GSK3β and tau, cleaved caspase-3, and phosphorylated NF-kB while it elevated PPARγ in P301S/PS19 mice, a tau mouse model of AD. Importantly, tau transgenic mice treated with JZL184 displayed improvements in spatial learning and memory retention. In addition, inactivation of MAGL ameliorates deteriorations in expression of synaptic proteins in P301S/PS19 mice. Our results provide further evidence that MAGL is a promising therapeutic target for AD.
Collapse
|
31
|
Pierre K, Dyson K, Dagra A, Williams E, Porche K, Lucke-Wold B. Chronic Traumatic Encephalopathy: Update on Current Clinical Diagnosis and Management. Biomedicines 2021; 9:415. [PMID: 33921385 PMCID: PMC8069746 DOI: 10.3390/biomedicines9040415] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 02/05/2023] Open
Abstract
Chronic traumatic encephalopathy is a disease afflicting individuals exposed to repetitive neurotrauma. Unfortunately, diagnosis is made by postmortem pathologic analysis, and treatment options are primarily symptomatic. In this clinical update, we review clinical and pathologic diagnostic criteria and recommended symptomatic treatments. We also review animal models and recent discoveries from pre-clinical studies. Furthermore, we highlight the recent advances in diagnosis using diffusor tensor imaging, functional magnetic resonance imaging, positron emission tomography, and the fluid biomarkers t-tau, sTREM2, CCL11, NFL, and GFAP. We also provide an update on emerging pharmaceutical treatments, including immunotherapies and those that target tau acetylation, tau phosphorylation, and inflammation. Lastly, we highlight the current literature gaps and guide future directions to further improve clinical diagnosis and management of patients suffering from this condition.
Collapse
Affiliation(s)
- Kevin Pierre
- College of Medicine, University of Florida, Gainesville, FL 32611, USA; (K.P.); (K.D.); (A.D.); (E.W.)
| | - Kyle Dyson
- College of Medicine, University of Florida, Gainesville, FL 32611, USA; (K.P.); (K.D.); (A.D.); (E.W.)
| | - Abeer Dagra
- College of Medicine, University of Florida, Gainesville, FL 32611, USA; (K.P.); (K.D.); (A.D.); (E.W.)
| | - Eric Williams
- College of Medicine, University of Florida, Gainesville, FL 32611, USA; (K.P.); (K.D.); (A.D.); (E.W.)
| | - Ken Porche
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA;
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA;
| |
Collapse
|
32
|
Ojo J, Eisenbaum M, Shackleton B, Lynch C, Joshi U, Saltiel N, Pearson A, Ringland C, Paris D, Mouzon B, Mullan M, Crawford F, Bachmeier C. Mural cell dysfunction leads to altered cerebrovascular tau uptake following repetitive head trauma. Neurobiol Dis 2020; 150:105237. [PMID: 33383188 PMCID: PMC8170787 DOI: 10.1016/j.nbd.2020.105237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/07/2020] [Accepted: 12/26/2020] [Indexed: 12/29/2022] Open
Abstract
A pathological characteristic of repetitive traumatic brain injury (TBI) is the deposition of hyperphosphorylated and aggregated tau species in the brain and increased levels of extracellular monomeric tau are believed to play a role in the pathogenesis of neurodegenerative tauopathies. The pathways by which extracellular tau is eliminated from the brain, however, remains elusive. The purpose of this study was to examine tau uptake by cerebrovascular cells and the effect of TBI on these processes. We found monomeric tau interacts with brain vascular mural cells (pericytes and smooth muscle cells) to a greater extent than other cerebrovascular cells, indicating mural cells may contribute to the elimination of extracellular tau, as previously described for other solutes such as beta-amyloid. Consistent with other neurodegenerative disorders, we observed a progressive decline in cerebrovascular mural cell markers up to 12 months post-injury in a mouse model of repetitive mild TBI (r-mTBI) and human TBI brain specimens, when compared to control. These changes appear to reflect mural cell degeneration and not cellular loss as no difference in the mural cell population was observed between r-mTBI and r-sham animals as determined through flow cytometry. Moreover, freshly isolated r-mTBI cerebrovessels showed reduced tau uptake at 6 and 12 months post-injury compared to r-sham animals, which may be the result of diminished cerebrovascular endocytosis, as caveolin-1 levels were significantly decreased in mouse r-mTBI and human TBI cerebrovessels compared to their respective controls. Further emphasizing the interaction between mural cells and tau, similar reductions in mural cell markers, tau uptake, and caveolin-1 were observed in cerebrovessels from transgenic mural cell-depleted animals. In conclusion, our studies indicate repeated injuries to the brain causes chronic mural cell degeneration, reducing the caveolar-mediated uptake of tau by these cells. Alterations in tau uptake by vascular mural cells may contribute to tau deposition in the brain following head trauma and could represent a novel therapeutic target for TBI or other neurodegenerative disorders.
Collapse
Affiliation(s)
- Joseph Ojo
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Max Eisenbaum
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Ben Shackleton
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Cillian Lynch
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Utsav Joshi
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | | | - Andrew Pearson
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Charis Ringland
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Daniel Paris
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Benoit Mouzon
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Michael Mullan
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; Bay Pines VA Healthcare System, Bay Pines, FL, USA.
| |
Collapse
|
33
|
Chiasseu M, Fesharaki-Zadeh A, Saito T, Saido TC, Strittmatter SM. Gene-environment interaction promotes Alzheimer's risk as revealed by synergy of repeated mild traumatic brain injury and mouse App knock-in. Neurobiol Dis 2020; 145:105059. [PMID: 32858147 PMCID: PMC7572902 DOI: 10.1016/j.nbd.2020.105059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/17/2020] [Accepted: 08/20/2020] [Indexed: 11/20/2022] Open
Abstract
There is a strong unmet need for translational progress towards Alzheimer's disease (AD) modifying therapy. Unfortunately, preclinical modeling of the disease has been disappointing, relying primarily on transgenic mouse overexpression of rare dominant mutations. Clinical manifestation of AD symptoms is known to reflect interaction between environmental and genetic risks. Mild traumatic brain injury (mTBI) is an environmental risk for dementia, including Alzheimer's, but there has been limited mechanistic analysis of mTBI contribution to AD. Here, we investigate the interplay between mTBI and Aβ precursor protein gene mutation in AD pathogenesis. We employed a knock-in (KI) model of AD that expresses the Aß-containing exons from human APP bearing the Swedish and Iberian mutations, namely AppNL-F/NL-F mice. Without environmental risk, this genetic variation yields minimal mouse symptomatology. Anesthetized 4-month-old KI mice and their age-matched wild type (WT) controls were subjected to repeated mild closed head injury (rmCHI), once daily for 14 days. Anesthetized, uninjured genotype- and age-matched mice were used as sham controls. At 3- and 8-months post-injury, amyloid-β, phospho-tau and Iba1 expression in the injured KI cortices were assessed. Our data reveal that rmCHI enhances accumulation of amyloid-β and hyperphosphorylated tau inclusions, as well as neuroinflammation in AppNL-F/NL-F mice. Furthermore, novel object recognition and Morris water maze tests demonstrated that rmCHI greatly exacerbates persistent cognitive deficits in APPNL-F/NL-F mice. Therefore, study of gene-environment interaction demonstrates that combining risk factors provides a more robust model for AD, and that repeated mTBI substantially accelerates AD pathology in a genetically susceptible situation.
Collapse
Affiliation(s)
- Marius Chiasseu
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA; Departments of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Arman Fesharaki-Zadeh
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA; Departments of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-ku, Mizuho-cho, Nagoya, Aichi 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA; Departments of Neurology, Yale University School of Medicine, New Haven, CT, USA; Departments of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
34
|
Tyukhtenko S, Ma X, Rajarshi G, Karageorgos I, Anderson KW, Hudgens JW, Guo JJ, Nasr ML, Zvonok N, Vemuri K, Wagner G, Makriyannis A. Conformational gating, dynamics and allostery in human monoacylglycerol lipase. Sci Rep 2020; 10:18531. [PMID: 33116203 PMCID: PMC7595040 DOI: 10.1038/s41598-020-75497-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 10/08/2020] [Indexed: 11/09/2022] Open
Abstract
Inhibition of human Monoacylglycerol Lipase (hMGL) offers a novel approach for treating neurological diseases. The design of inhibitors, targeting active-inactive conformational transitions of the enzyme, can be aided by understanding the interplay between structure and dynamics. Here, we report the effects of mutations within the catalytic triad on structure, conformational gating and dynamics of hMGL by combining kinetics, NMR, and HDX-MS data with metadynamics simulations. We found that point mutations alter delicate conformational equilibria between active and inactive states. HDX-MS reveals regions of the hMGL that become substantially more dynamic upon substitution of catalytic acid Asp-239 by alanine. These regions, located far from the catalytic triad, include not only loops but also rigid α-helixes and β-strands, suggesting their involvement in allosteric regulation as channels for long-range signal transmission. The results identify the existence of a preorganized global communication network comprising of tertiary (residue-residue contacts) and quaternary (rigid-body contacts) networks that mediate robust, rapid intraprotein signal transmission. Catalytic Asp-239 controls hMGL allosteric communications and may be considered as an essential residue for the integration and transmission of information to enzymes' remote regions, in addition to its well-known role to facilitate Ser-122 activation. Our findings may assist in the identification of new druggable sites in hMGL.
Collapse
Affiliation(s)
- Sergiy Tyukhtenko
- Center for Drug Discovery and Departments of Pharmaceutical Sciences and Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115-5000, USA.
| | - Xiaoyu Ma
- Center for Drug Discovery and Departments of Pharmaceutical Sciences and Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115-5000, USA
| | - Girija Rajarshi
- Center for Drug Discovery and Departments of Pharmaceutical Sciences and Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115-5000, USA
| | - Ioannis Karageorgos
- BioProcess Measurements Group, Biomolecular Measurement Division, National Institute of Standards & Technology, Rockville, MD, 20850, USA.,Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Kyle W Anderson
- BioProcess Measurements Group, Biomolecular Measurement Division, National Institute of Standards & Technology, Rockville, MD, 20850, USA.,Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Jeffrey W Hudgens
- BioProcess Measurements Group, Biomolecular Measurement Division, National Institute of Standards & Technology, Rockville, MD, 20850, USA.,Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Jason J Guo
- Center for Drug Discovery and Departments of Pharmaceutical Sciences and Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115-5000, USA.,Barnett Institute for Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115-5000, USA
| | - Mahmoud L Nasr
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.,Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nikolai Zvonok
- Center for Drug Discovery and Departments of Pharmaceutical Sciences and Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115-5000, USA
| | - Kiran Vemuri
- Center for Drug Discovery and Departments of Pharmaceutical Sciences and Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115-5000, USA
| | - Gerhard Wagner
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery and Departments of Pharmaceutical Sciences and Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115-5000, USA.
| |
Collapse
|
35
|
Potential application of endocannabinoid system agents in neuropsychiatric and neurodegenerative diseases-focusing on FAAH/MAGL inhibitors. Acta Pharmacol Sin 2020; 41:1263-1271. [PMID: 32203086 PMCID: PMC7608191 DOI: 10.1038/s41401-020-0385-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/15/2020] [Accepted: 02/19/2020] [Indexed: 01/01/2023]
Abstract
The endocannabinoid system (ECS) has received extensive attention for its neuroprotective effect on the brain. This system comprises endocannabinoids, endocannabinoid receptors, and the corresponding ligands and proteins. The molecular players involved in their regulation and metabolism are potential therapeutic targets for neuropsychiatric diseases including anxiety, depression and neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). The inhibitors of two endocannabinoid hydrolases, i.e., fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), have the capacity to increase the level of endocannabinoids indirectly, causing fewer side effects than those associated with direct supplementation of cannabinoids. Their antidepressant and anxiolytic mechanisms are considered to modulate the hypothalamic-pituitary-adrenal axis and regulate synaptic and neural plasticity. In terms of AD/PD, treatment with FAAH/MAGL inhibitors leads to reduction in amyloid β-protein deposition and inhibition of the death of dopamine neurons, which are commonly accepted to underlie the pathogenesis of AD and PD, respectively. Inflammation as the cause of depression/anxiety and PD/AD is also the target of FAAH/MAGL inhibitors. In this review, we summarize the application and involvement of FAAH/MAGL inhibitors in related neurological diseases. Focus on the latest research progress using FAAH/MAGL inhibitors is expected to facilitate the development of novel approaches with therapeutic potential.
Collapse
|
36
|
Wang ZM, Liu C, Wang YY, Deng YS, He XC, Du HZ, Liu CM, Teng ZQ. SerpinA3N deficiency deteriorates impairments of learning and memory in mice following hippocampal stab injury. Cell Death Discov 2020; 6:88. [PMID: 33014432 PMCID: PMC7501238 DOI: 10.1038/s41420-020-00325-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/22/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury is a global leading cause of disability and death, which puts patients at high risk for developing dementia. Early intervention is believed as the key to minimize the development of brain damages that could aggravate the symptoms. Here, we report that the serine protease inhibitor SerpinA3N is upregulated in hippocampal neurons in the early stage of hippocampal stab injury (HSI), while its deficiency causes a greater degree of neuronal apoptosis and severer impairments of spatial learning and memory in mice after HSI. We further show that MMP2 is a key substrate of SerpinA3N, and MMP2 specific inhibitor (ARP100) can protect against neuronal apoptosis and cognitive dysfunction in mice after HSI. These findings demonstrate a critical role for SerpinA3N in neuroprotection, suggesting that SerpinA3N and MMP2 inhibitors might be a novel therapeutic agents for neurotrauma.
Collapse
Affiliation(s)
- Zhi-Meng Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Cong Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Ying-Ying Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Yu-Sen Deng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Xuan-Cheng He
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| | - Hong-Zhen Du
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| |
Collapse
|
37
|
Breen PW, Krishnan V. Recent Preclinical Insights Into the Treatment of Chronic Traumatic Encephalopathy. Front Neurosci 2020; 14:616. [PMID: 32774238 PMCID: PMC7381336 DOI: 10.3389/fnins.2020.00616] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/18/2020] [Indexed: 12/29/2022] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative condition associated with significant mortality and morbidity. The central pathophysiological mechanisms by which repetitive cranial injury results in the neurodegeneration of CTE are poorly understood. Current well-established working models emphasize a central role for trauma-induced excessive phosphorylation and accumulation of insoluble tangles of Tau protein. In this review, we summarize recent data from preclinical animal models of CTE where a series of candidate treatments have been carefully evaluated, including kinase inhibitors, antibody therapy, and anti-inflammatory therapies. We discuss the overall translational potential of these approaches and provide recommendations for future bench-to-bedside treatment strategies.
Collapse
Affiliation(s)
- Patrick W Breen
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Vaishnav Krishnan
- Department of Neurology, Baylor College of Medicine, Houston, TX United States
| |
Collapse
|
38
|
Reddy V, Grogan D, Ahluwalia M, Salles ÉL, Ahluwalia P, Khodadadi H, Alverson K, Nguyen A, Raju SP, Gaur P, Braun M, Vale FL, Costigliola V, Dhandapani K, Baban B, Vaibhav K. Targeting the endocannabinoid system: a predictive, preventive, and personalized medicine-directed approach to the management of brain pathologies. EPMA J 2020; 11:217-250. [PMID: 32549916 PMCID: PMC7272537 DOI: 10.1007/s13167-020-00203-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Cannabis-inspired medical products are garnering increasing attention from the scientific community, general public, and health policy makers. A plethora of scientific literature demonstrates intricate engagement of the endocannabinoid system with human immunology, psychology, developmental processes, neuronal plasticity, signal transduction, and metabolic regulation. Despite the therapeutic potential, the adverse psychoactive effects and historical stigma, cannabinoids have limited widespread clinical application. Therefore, it is plausible to weigh carefully the beneficial effects of cannabinoids against the potential adverse impacts for every individual. This is where the concept of "personalized medicine" as a promising approach for disease prediction and prevention may take into the account. The goal of this review is to provide an outline of the endocannabinoid system, including endocannabinoid metabolizing pathways, and will progress to a more in-depth discussion of the therapeutic interventions by endocannabinoids in various neurological disorders.
Collapse
Affiliation(s)
- Vamsi Reddy
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Dayton Grogan
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Meenakshi Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Évila Lopes Salles
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Katelyn Alverson
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Andy Nguyen
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Srikrishnan P. Raju
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
- Brown University, Providence, RI USA
| | - Pankaj Gaur
- Georgia Cancer Center, Augusta University, Augusta, GA USA
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, USA
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, USA
| | - Fernando L. Vale
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | | | - Krishnan Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| |
Collapse
|
39
|
Bachstetter AD, Morganti JM, Bodnar CN, Webster SJ, Higgins EK, Roberts KN, Snider H, Meier SE, Nation GK, Goulding DS, Hamm M, Powell DK, Vandsburger M, Van Eldik LJ, Abisambra JF. The effects of mild closed head injuries on tauopathy and cognitive deficits in rodents: Primary results in wild type and rTg4510 mice, and a systematic review. Exp Neurol 2020; 326:113180. [PMID: 31930992 PMCID: PMC7373372 DOI: 10.1016/j.expneurol.2020.113180] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/02/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022]
Abstract
In humans, the majority of sustained traumatic brain injuries (TBIs) are classified as 'mild' and most often a result of a closed head injury (CHI). The effects of a non-penetrating CHI are not benign and may lead to chronic pathology and behavioral dysfunction, which could be worsened by repeated head injury. Clinical-neuropathological correlation studies provide evidence that conversion of tau into abnormally phosphorylated proteotoxic intermediates (p-tau) could be part of the pathophysiology triggered by a single TBI and enhanced by repeated TBIs. However, the link between p-tau and CHI in rodents remains controversial. To address this question experimentally, we induced a single CHI or two CHIs to WT or rTg4510 mice. We found that 2× CHI increased tau phosphorylation in WT mice and rTg4510 mice. Behavioral characterization in WT mice found chronic deficits in the radial arm water maze in 2× CHI mice that had partially resolved in the 1× CHI mice. Moreover, using Manganese-Enhanced Magnetic Resonance Imaging with R1 mapping - a novel functional neuroimaging technique - we found greater deficits in the rTg4510 mice following 2× CHI compared to 1× CHI. To integrate our findings with prior work in the field, we conducted a systematic review of rodent mild repetitive CHI studies. Following Prisma guidelines, we identified 25 original peer-reviewed papers. Results from our experiments, as well as our systematic review, provide compelling evidence that tau phosphorylation is modified by experimental mild TBI studies; however, changes in p-tau levels are not universally reported. Together, our results provide evidence that repetitive TBIs can result in worse and more persistent neurological deficits compared to a single TBI, but the direct link between the worsened outcome and elevated p-tau could not be established.
Collapse
Affiliation(s)
- Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America.
| | - Josh M Morganti
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Colleen N Bodnar
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Scott J Webster
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Emma K Higgins
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Kelly N Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Henry Snider
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Shelby E Meier
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Department of Physiology, University of Kentucky, Lexington, KY, United States of America
| | - Grant K Nation
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Department of Physiology, University of Kentucky, Lexington, KY, United States of America
| | - Danielle S Goulding
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Matthew Hamm
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States of America
| | - David K Powell
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Department of Biomedical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Moriel Vandsburger
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, United States of America
| | - Linda J Van Eldik
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Jose F Abisambra
- Department of Physiology, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
40
|
Sacramento CB, Sondhi D, Rosenberg JB, Chen A, Giordano S, Pey E, Lee V, Stiles KM, Havlicek DF, Leopold PL, Kaminsky SM, Crystal RG. Anti-Phospho-Tau Gene Therapy for Chronic Traumatic Encephalopathy. Hum Gene Ther 2019; 31:57-69. [PMID: 31608704 DOI: 10.1089/hum.2019.174] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a progressive neurodegenerative disorder caused by repetitive trauma to the central nervous system (CNS) suffered by soldiers, contact sport athletes, and civilians following accident-related trauma. CTE is a CNS tauopathy, with trauma-induced inflammation leading to accumulation of hyperphosphorylated forms of the microtubule-binding protein Tau (pTau), resulting in neurofibrillary tangles and progressive loss of neurons. At present, there are no therapies to treat CTE. We hypothesized that direct CNS administration of an adeno-associated virus (AAV) vector coding for an anti-pTau antibody would generate sufficient levels of anti-pTau in the CNS to suppress pTau accumulation thus interrupting the pathogenic process. Using a serotype AAVrh.10 gene transfer vector coding for a monoclonal antibody directed against pTau, we demonstrate the feasibility of this strategy in a murine CTE model in which pTau accumulation was elicited by repeated traumatic brain injury (TBI) using a closed cortical impact procedure over 5 days. Direct delivery of AAVrh.10 expression vectors coding for either of the two different anti-pTau antibodies to the hippocampus of these TBI mice significantly reduced pTau levels across the CNS. Using doses that can be safely scaled to humans, the data demonstrate that CNS administration of AAVrh.10anti-pTau is effective, providing a new strategy to interrupt the CTE consequences of TBI.
Collapse
Affiliation(s)
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Alvin Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephanie Giordano
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Eduard Pey
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Vladlena Lee
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Katie M Stiles
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - David F Havlicek
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Philip L Leopold
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
41
|
Grimsey NL, Savinainen JR, Attili B, Ahamed M. Regulating membrane lipid levels at the synapse by small-molecule inhibitors of monoacylglycerol lipase: new developments in therapeutic and PET imaging applications. Drug Discov Today 2019; 25:330-343. [PMID: 31622747 DOI: 10.1016/j.drudis.2019.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/17/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022]
Abstract
Monoacylglycerol lipase (MAGL) is a major endocannabinoid hydrolyzing enzyme and can be regulated to control endogenous lipid levels in the brain. This review highlights the pharmacological roles and in vivo PET imaging of MAGL in brain.
Collapse
Affiliation(s)
- Natasha L Grimsey
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Juha R Savinainen
- Institute of Biomedicine, Faculty of Health Sciences, The University of Eastern Finland, Finland
| | - Bala Attili
- Department of Radiology, The University of Cambridge, UK
| | - Muneer Ahamed
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Australia.
| |
Collapse
|
42
|
The Administration of the New Pyrimidine Derivative-4-{2-[2-(3,4-Dimethoxyphenyl)-Vinyl]-6-Ethyl-4-Oxo-5-Phenyl-4H-Pyrimidine-1-Il}Benzsulfamide Restores the Activity of Brain Cells in Experimental Chronic Traumatic Encephalopathy by Maintaining Mitochondrial Function. ACTA ACUST UNITED AC 2019; 55:medicina55070386. [PMID: 31319603 PMCID: PMC6681389 DOI: 10.3390/medicina55070386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
Background and objectives: To evaluate the effect of a new pyrimidine derivative on the change of mitochondrial function in experimental chronic traumatic encephalopathy. Materials and methods: The study was performed on male mice of the BALB/c line (acute toxicity was assessed) and male rats of the Wistar line, which were modeled chronic traumatic encephalopathy by the method of free fall of the load (weight 150 g from a 50 cm height). The injury to rats was reproduced once a day for 7 days. Further, cognitive functions, changes in sensorimotor deficiency, cerebral blood flow, neuron-specific enolase(NSE), S100β, glial fibrillary acidic protein (GFAP) (in blood serum) and β-amyloid, adenosine triphosphate (ATP) (in brain tissue supernatant) were evaluated. Mitochondrial respiration was also measured. Choline alfoscerate (100 mg/kg) was used as a reference drug. Results: The study found that the use of a new pyrimidine derivative contributed to the preservation of the mitochondrial respirometric function and cognitive functions in rats. In addition, against the administration of test-object marked increase in the concentration of ATP, the velocity of cerebral blood flow was 4.2 times (p < 0.05) and 35.6% (p < 0.05), respectively, as well as reduced concentration and GFAP, NSE, S100β, β-amyloid and sensorimotor deficit at 2.7 (p < 0.05) times; 2 times (p < 0.05); 2.4 times (p < 0.05); of 30.4% (p < 0.05 and 46.5% (p < 0.05), respectively. The LD50 (per os) for the test-object was 4973.56 ± 573.72 mg/kg. Conclusion: Based on the obtained data, high therapeutic efficacy and low systemic toxicity of the application are assumed 4-{2-[2-(3,4-dimethoxyphenyl)-vinyl]-6-ethyl-4-oxo-5-phenyl-4H-pyrimidine-1-Il}benzsulfamide in chronic traumatic encephalopathy.
Collapse
|
43
|
Heyburn L, Sajja VSSS, Long JB. The Role of TDP-43 in Military-Relevant TBI and Chronic Neurodegeneration. Front Neurol 2019; 10:680. [PMID: 31316455 PMCID: PMC6610302 DOI: 10.3389/fneur.2019.00680] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022] Open
Abstract
Due largely to the use of improvised explosive devices (IEDs) and other explosives in recent military conflicts, blast-related TBI has emerged as a prominent injury sustained by warfighters. In the recent wars in Iraq and Afghanistan, traumatic brain injury (TBI) has been one of the most common types of injury sustained by soldiers and military personnel; of the ~380,000 TBIs reported in service members from 2000 to 2017, 82.3% were classified as mild (mTBI). While mTBI is associated with normal structural imaging, brief or no loss of consciousness, and rapid recovery of mental state, mTBI can nevertheless lead to persistent behavioral and cognitive effects. As in other cases of mTBI, exposure to low-level blast often does not cause immediate overt neurological effects, but may similarly lead to persistent behavioral and cognitive deficits. These effects are likely to be compounded when multiple exposures to blast and/or impact are sustained, since there is increasing evidence that multiple mTBIs can lead to chronic neurodegeneration. One common form of this deleterious outcome is frontotemporal lobar degeneration (FTLD), which is a progressive neurodegenerative process marked by atrophy of the frontal and temporal lobes, leading to frontotemporal dementia, a common form of dementia affecting behavior, cognition and language. About half of all cases of FTLD are marked by TAR-DNA binding protein (TDP-43)-positive protein inclusions. TDP-43, a DNA/RNA binding protein, controls the expression of thousands of genes and is associated with several neurodegenerative diseases including amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease, and chronic traumatic encephalopathy. TDP-43 abnormalities have also been associated with traumatic brain injury in both pre-clinical and clinical studies. The role of TDP-43 in the manifestation of FTLD pathology in military TBI cases is currently unclear, and to date there has been only a limited number of pre-clinical studies addressing the effects of repeated blast-related mild TBI (rbTBI) in relation to FTLD and TDP-43. This review will summarize some of these findings and address the concerns and critical knowledge gaps associated with FTLD manifestation with military populations, as well as clinical findings on other forms of mTBI.
Collapse
Affiliation(s)
- Lanier Heyburn
- Blast Induced Neurotrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Venkata S S S Sajja
- Blast Induced Neurotrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Joseph B Long
- Blast Induced Neurotrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
44
|
Selvaraj P, Wen J, Tanaka M, Zhang Y. Therapeutic Effect of a Novel Fatty Acid Amide Hydrolase Inhibitor PF04457845 in the Repetitive Closed Head Injury Mouse Model. J Neurotrauma 2019; 36:1655-1669. [PMID: 30526351 DOI: 10.1089/neu.2018.6226] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Concussive traumatic brain injury (TBI) is the predominant type of brain injury in young adults and is a risk factor for the development of chronic traumatic encephalopathy and other neurodegenerative diseases late in life. Using a repetitive closed head injury mouse model, we found that treatment with PF04457845, a novel fatty acid amide hydrolase (FAAH) inhibitor that selectively elevated the brain levels of anandamide, improved locomotor function, learning, and memory in TBI mice examined by beam walk, Y-maze, and Morris water maze tests. The accumulation of microglia and astrocytes and the expression of proinflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha (TNF-α), in the ipsilateral TBI mouse cortex and hippocampus were significantly reduced by drug treatment. The increased expression of amyloid precursor protein (APP), phosphorylated Tau (p-Tau), phosphorylated glycogen synthase kinase 3 beta (pGSK3β) and p35/p25 subunits and the decreased expression of the pre-synaptic proteins, synaptophysin, synaptosome-associated protein of 25 kDa (SNAP25), and cysteine string protein alpha (α-CSP), in TBI mouse brain were also normalized by PF04458745 treatment. The improved locomotor function and working memory were partially mediated by activation of both cannabinoid (CB)1 and CB2 receptors, whereas the improvement on spatial learning and memory seemed to be CB1 receptor dependent. Interestingly, the blockage of PF04457845 on the reduced expression of synaptophysin, but not SNAP25 and α-CSP, was reversed by coadministration of the CB1 receptor antagonist. These results suggest that the therapeutic effect of PF04457845 is mediated by both cannabinoid receptor dependent and independent mechanisms, and selective inhibition of FAAH possesses a great potential for the treatment of TBI.
Collapse
Affiliation(s)
- Prabhuanand Selvaraj
- 1 Department of Anatomy, Physiology, and Genetics and Uniformed Services University of the Health Sciences, Bethesda, Maryland.,2 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Jie Wen
- 1 Department of Anatomy, Physiology, and Genetics and Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Mikiei Tanaka
- 1 Department of Anatomy, Physiology, and Genetics and Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Yumin Zhang
- 1 Department of Anatomy, Physiology, and Genetics and Uniformed Services University of the Health Sciences, Bethesda, Maryland.,2 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
45
|
Song Y, Hu M, Zhang J, Teng ZQ, Chen C. A novel mechanism of synaptic and cognitive impairments mediated via microRNA-30b in Alzheimer's disease. EBioMedicine 2019; 39:409-421. [PMID: 30522932 PMCID: PMC6354659 DOI: 10.1016/j.ebiom.2018.11.059] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND It is widely accepted that cognitive and memory deficits in Alzheimer's disease (AD) primarily result from synaptic failure. However, the mechanisms that underlie synaptic and cognitive dysfunction remain unclear. METHODS We utilized molecular biology techniques, electrophysiological recordings, fluorescence in situ hybridization (FISH), immuno- and Golgi-staining, chromatin immunoprecipitation (CHIP); lentivirus (LV)-based microRNA overexpression and 'sponging', and behavioral tests to assess upregulated miR-30b causing synaptic and cognitive declines in APP transgenic (TG) mice. FINDINGS We provide evidence that expression of miR-30b, which targets molecules important for maintaining synaptic integrity, including ephrin type-B receptor 2 (ephB2), sirtuin1 (sirt1), and glutamate ionotropic receptor AMPA type subunit 2 (GluA2), is robustly upregulated in the brains of both AD patients and APP transgenic (TG) mice, an animal model of AD, while expression of its targets is significantly downregulated. Overexpression of miR-30b in the hippocampus of normal wild-type (WT) mice impairs synaptic and cognitive functions, mimicking those seen in TG mice. Conversely, knockdown of endogenous miR-30b in TG mice prevents synaptic and cognitive decline. We further observed that expression of miR-30b is upregulated by proinflammatory cytokines and Aβ42 through NF-κB signaling. INTERPRETATION Our results provide a previously undefined mechanism by which unregulated miR-30b causes synaptic and cognitive dysfunction in AD, suggesting that reversal of dysregulated miR-30b in the brain may prevent or slow cognitive declines in AD. FUND: This work was supported by National Institutes of Health grants R01NS076815, R01MH113535, R01AG058621, P30GM103340 Pilot Project, and by the LSUHSC School of Medicine Research Enhancement Program grant (to C.C.).
Collapse
Affiliation(s)
- Yunping Song
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Mei Hu
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Jian Zhang
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Zhao-Qian Teng
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Chu Chen
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Department of Otorhinolaryngology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
46
|
Clapper JR, Henry CL, Niphakis MJ, Knize AM, Coppola AR, Simon GM, Ngo N, Herbst RA, Herbst DM, Reed AW, Cisar JS, Weber OD, Viader A, Alexander JP, Cunningham ML, Jones TK, Fraser IP, Grice CA, Ezekowitz RAB, O'Neill GP, Blankman JL. Monoacylglycerol Lipase Inhibition in Human and Rodent Systems Supports Clinical Evaluation of Endocannabinoid Modulators. J Pharmacol Exp Ther 2018; 367:494-508. [PMID: 30305428 DOI: 10.1124/jpet.118.252296] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/05/2018] [Indexed: 03/08/2025] Open
Abstract
Monoacylglycerol lipase (MGLL) is the primary degradative enzyme for the endocannabinoid 2-arachidonoylglycerol (2-AG). The first MGLL inhibitors have recently entered clinical development for the treatment of neurologic disorders. To support this clinical path, we report the pharmacological characterization of the highly potent and selective MGLL inhibitor ABD-1970 [1,1,1,3,3,3-hexafluoropropan-2-yl 4-(2-(8-oxa-3-azabicyclo[3.2.1]octan-3-yl)-4-chlorobenzyl)piperazine-1-carboxylate]. We used ABD-1970 to confirm the role of MGLL in human systems and to define the relationship between MGLL target engagement, brain 2-AG concentrations, and efficacy. Because MGLL contributes to arachidonic acid metabolism in a subset of rodent tissues, we further used ABD-1970 to evaluate whether selective MGLL inhibition would affect prostanoid production in several human assays known to be sensitive to cyclooxygenase inhibitors. ABD-1970 robustly elevated brain 2-AG content and displayed antinociceptive and antipruritic activity in a battery of rodent models (ED50 values of 1-2 mg/kg). The antinociceptive effects of ABD-1970 were potentiated when combined with analgesic standards of care and occurred without overt cannabimimetic effects. ABD-1970 also blocked 2-AG hydrolysis in human brain tissue and elevated 2-AG content in human blood without affecting stimulated prostanoid production. These findings support the clinical development of MGLL inhibitors as a differentiated mechanism to treat pain and other neurologic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nhi Ngo
- Abide Therapeutics, San Diego, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Morin A, Mouzon B, Ferguson S, Paris D, Saltiel N, Lungmus C, Mullan M, Crawford F. Treatment With Nilvadipine Mitigates Inflammatory Pathology and Improves Spatial Memory in Aged hTau Mice After Repetitive Mild TBI. Front Aging Neurosci 2018; 10:292. [PMID: 30364309 PMCID: PMC6193195 DOI: 10.3389/fnagi.2018.00292] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/04/2018] [Indexed: 12/14/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is the most common form of brain trauma worldwide. The effects of mTBI are not well-studied within the elderly population, yet older adults constitute a significant portion of all mTBI patients. Few preclinical studies have focused on the effects of mTBI, or mTBI treatments, in the aged brain, and none have explored repetitive mTBI (r-mTBI). In this study, we have administered our well-characterized 5-injury model (5 r-mTBI) to hTau mice aged 24 months to explore the neurobehavioral and neuropathological outcomes, and the effects of treatment with the dihydropyridine, Nilvadipine. Our previous studies have shown that Nilvadipine inhibits spleen tyrosine kinase (Syk), is effective at reducing inflammation, tau hyperphosphorylation, and amyloid production, and it has recently been investigated in a European Phase III clinical trial for Alzheimer’s disease (AD). In our 24-month-old r-mTBI mice, we observed increased neuroinflammation and a trend toward impaired cognitive performance compared to sham controls. Treatment with Nilvadipine mitigated the TBI-induced inflammatory response in aged r-mTBI animals and significantly improved spatial memory. To our knowledge, this is the only preclinical study focusing on the treatment of r-mTBI in aged, and these results suggest a therapeutic potential of Nilvadipine for consequences of mTBI.
Collapse
Affiliation(s)
- Alexander Morin
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Benoit Mouzon
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Scott Ferguson
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Daniel Paris
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Nicole Saltiel
- The Roskamp Institute, Sarasota, FL, United States.,James A Haley Veterans Administration, Tampa, FL, United States
| | | | - Mike Mullan
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| |
Collapse
|
48
|
Elliott MB, Ward SJ, Abood ME, Tuma RF, Jallo JI. Understanding the endocannabinoid system as a modulator of the trigeminal pain response to concussion. Concussion 2018; 2:CNC49. [PMID: 30202590 PMCID: PMC6122691 DOI: 10.2217/cnc-2017-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Post-traumatic headache is the most common symptom of postconcussion syndrome and becomes a chronic neurological disorder in a substantial proportion of patients. This review provides a brief overview of the epidemiology of postconcussion headache, research models used to study this disorder, as well as the proposed mechanisms. An objective of this review is to enhance the understanding of how the endogenous cannabinoid system is essential for maintaining the balance of the CNS and regulating inflammation after injury, and in turn making the endocannabinoid system a potential modulator of the trigeminal response to concussion. The review describes the role of endocannabinoid modulation of pain and the potential for use of phytocannabinoids to treat pain, migraine and concussion.
Collapse
Affiliation(s)
- Melanie B Elliott
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| | - Sara J Ward
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Mary E Abood
- Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Ronald F Tuma
- Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Jack I Jallo
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| |
Collapse
|
49
|
Cisar JS, Weber OD, Clapper JR, Blankman JL, Henry CL, Simon GM, Alexander JP, Jones TK, Ezekowitz RAB, O’Neill GP, Grice CA. Identification of ABX-1431, a Selective Inhibitor of Monoacylglycerol Lipase and Clinical Candidate for Treatment of Neurological Disorders. J Med Chem 2018; 61:9062-9084. [DOI: 10.1021/acs.jmedchem.8b00951] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Justin S. Cisar
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Olivia D. Weber
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Jason R. Clapper
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Jacqueline L. Blankman
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Cassandra L. Henry
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Gabriel M. Simon
- Vividion Therapeutics, 3565 General Atomics Court, Suite 100, San Diego, California 92121, United States
| | - Jessica P. Alexander
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Todd K. Jones
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - R. Alan B. Ezekowitz
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Gary P. O’Neill
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| | - Cheryl A. Grice
- Abide Therapeutics, 10835 Road to the Cure, Suite 250, San Diego, California 92121, United States
| |
Collapse
|
50
|
Paloczi J, Varga ZV, Hasko G, Pacher P. Neuroprotection in Oxidative Stress-Related Neurodegenerative Diseases: Role of Endocannabinoid System Modulation. Antioxid Redox Signal 2018; 29:75-108. [PMID: 28497982 PMCID: PMC5984569 DOI: 10.1089/ars.2017.7144] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/11/2017] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Redox imbalance may lead to overproduction of reactive oxygen and nitrogen species (ROS/RNS) and subsequent oxidative tissue damage, which is a critical event in the course of neurodegenerative diseases. It is still not fully elucidated, however, whether oxidative stress is the primary trigger or a consequence in the process of neurodegeneration. Recent Advances: Increasing evidence suggests that oxidative stress is involved in the propagation of neuronal injury and consequent inflammatory response, which in concert promote development of pathological alterations characteristic of most common neurodegenerative diseases. CRITICAL ISSUES Accumulating recent evidence also suggests that there is an important interplay between the lipid endocannabinoid system [ECS; comprising the main cannabinoid 1 and 2 receptors (CB1 and CB2), endocannabinoids, and their synthetic and metabolizing enzymes] and various key inflammatory and redox-dependent processes. FUTURE DIRECTIONS Targeting the ECS to modulate redox state-dependent cell death and to decrease consequent or preceding inflammatory response holds therapeutic potential in a multitude of oxidative stress-related acute or chronic neurodegenerative disorders from stroke and traumatic brain injury to Alzheimer's and Parkinson's diseases and multiple sclerosis, just to name a few, which will be discussed in this overview. Antioxid. Redox Signal. 29, 75-108.
Collapse
Affiliation(s)
- Janos Paloczi
- Laboratory of Cardiovascular Physiology and Tissue Injury (LCPTI), National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland
| | - Zoltan V. Varga
- Laboratory of Cardiovascular Physiology and Tissue Injury (LCPTI), National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland
| | - George Hasko
- Department of Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury (LCPTI), National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland
| |
Collapse
|