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 2024:10738584241275583. [PMID: 39316552 DOI: 10.1177/10738584241275583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
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
|
3
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. HISTOLOGICAL COMPARISON OF REPEATED MILD WEIGHT DROP AND LATERAL FLUID PERCUSSION INJURY MODELS OF TRAUMATIC BRAIN INJURY IN FEMALE AND MALE RATS. Shock 2024; 62:398-409. [PMID: 38813916 DOI: 10.1097/shk.0000000000002395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
ABSTRACT In preclinical traumatic brain injury (TBI) research, the animal model should be selected based on the research question and outcome measures of interest. Direct side-by-side comparisons of different injury models are essential for informing such decisions. Here, we used immunohistochemistry to compare the outcomes from two common models of TBI, lateral fluid percussion (LFP) and repeated mild weight drop (rmWD) in adult female and male Wistar rats. Specifically, we measured the effects of LFP and rmWD on markers of cerebrovascular and tight junction disruption, neuroinflammation, mature neurons, and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA 2/3 area of the hippocampus. Animals were randomized into the LFP or rmWD group. On day 1, the LFP group received a craniotomy, and on day 4, injury (or sham procedure; randomly assigned). The rmWD animals underwent either injury or isoflurane only (randomly assigned) on each of those 4 days. Seven days after injury, brains were harvested for analysis. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy only, whereas rmWD animals showed the least residual changes compared with isoflurane-only controls, supporting consideration of rmWD as a mild injury. LFP led to longer-lasting disruptions, perhaps more representative of moderate TBI. We also report that craniotomy and LFP produced greater disruptions in females relative to males. These findings will assist the field in the selection of animal models based on target severity of postinjury outcomes and support the inclusion of both sexes and appropriate control groups.
Collapse
Affiliation(s)
| | - Shealan C Cruise
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | | | | |
Collapse
|
4
|
Martínez-Torres AM, Morán J. Aquaporin 4 and the endocannabinoid system: a potential therapeutic target in brain injury. Exp Brain Res 2024; 242:2041-2058. [PMID: 39043897 PMCID: PMC11306651 DOI: 10.1007/s00221-024-06896-7] [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: 02/20/2024] [Accepted: 07/14/2024] [Indexed: 07/25/2024]
Abstract
Brain edema is a critical complication arising from stroke and traumatic brain injury (TBI) with an important impact on patient recovery and can lead to long-term consequences. Therapeutic options to reduce edema progression are limited with variable patient outcomes. Aquaporin 4 (AQP4) is a water channel that allows bidirectional water diffusion across the astrocyte membrane and participates in the distinct phases of cerebral edema. The absence or inhibition of this channel has been demonstrated to ameliorate edema and brain damage. The endocannabinoid system (ECS) is a neuromodulator system with a wide expression in the brain and its activation has shown neuroprotective properties in diverse models of neuronal damage. This review describes and discusses the major features of ECS and AQP4 and their role during brain damage, observing that ECS stimulation reduces edema and injury size in diverse models of brain damage, however, the relationship between AQP4 expression and dynamics and ECS activation remains unclear. The research on these topics holds promising therapeutic implications for the treatment of brain edema following stroke and TBI.
Collapse
Affiliation(s)
- Ari Misael Martínez-Torres
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacán, Apartado Postal 70-253, 04510, Ciudad de Mexico, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacán, Apartado Postal 70-253, 04510, Ciudad de Mexico, México.
| |
Collapse
|
5
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. Histological comparison of repeated mild weight drop and lateral fluid percussion injury models of traumatic brain injury (TBI) in female and male rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578177. [PMID: 38352449 PMCID: PMC10862833 DOI: 10.1101/2024.01.31.578177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Traumatic brain injury (TBI) heterogeneity has led to the development of several preclinical models, each modeling a distinct subset of outcomes. Selection of an injury model should be guided by the research question and the specific outcome measures of interest. Consequently, there is a need for conducting direct comparisons of different TBI models. Here, we used immunohistochemistry to directly compare the outcomes from two common models, lateral fluid percussion (LFP) and repeat mild weight drop (rmWD), on neuropathology in adult female and male Wistar rats. Specifically, we used immunohistochemistry to measure the effects of LFP and rmWD on cerebrovascular and tight junction disruption, inflammatory markers, mature neurons and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA2/3 area of the hippocampus. Animals were randomized into either LFP or rmWD groups. The LFP group received a craniotomy prior to LFP (or corresponding sham procedure) three days later, while rmWD animals underwent either weight drop or sham (isoflurane only) on each of those four days. After a recovery period of 7 days, animals were euthanized, and brains were harvested for analysis of RECA-1, claudin-5, GFAP, Iba-1, CD-68, NeuN, and wisteria floribunda lectin. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy-only, while rmWD animals showed the least residual changes compared to isoflurane-only controls. These findings support consideration of rmWD as a mild, transient injury. LFP leads to longer-lasting disruptions that are more closely associated with a moderate TBI. We further show that both craniotomy and LFP produced greater disruptions in females relative to males at 7 days post-injury. These findings support the inclusion of a time-matched experimentally-naïve or anesthesia-only control group in preclinical TBI research to enhance the validity of data interpretation and conclusions.
Collapse
|
6
|
Weera MM, Webster DA, Shackett RS, Benvenuti F, Middleton JW, Gilpin NW. Traumatic Stress-Induced Increases in Anxiety-like Behavior and Alcohol Self-Administration Are Mediated by Central Amygdala CRF1 Neurons That Project to the Lateral Hypothalamus. J Neurosci 2023; 43:8690-8699. [PMID: 37932105 PMCID: PMC10727175 DOI: 10.1523/jneurosci.1414-23.2023] [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: 07/26/2023] [Revised: 09/21/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023] Open
Abstract
Avoidance stress coping, defined as persistent internal and/or external avoidance of stress-related stimuli, is a key feature of anxiety- and stress-related disorders, and contributes to increases in alcohol misuse after stress exposure. Previous work using a rat model of predator odor stress avoidance identified corticotropin-releasing factor (CRF) signaling via CRF Type 1 receptors (CRF1) in the CeA, as well as CeA projections to the lateral hypothalamus (LH) as key mediators of conditioned avoidance of stress-paired contexts and/or increased alcohol drinking after stress. Here, we report that CRF1-expressing CeA cells that project to the LH are preferentially activated in male and female rats that show persistent avoidance of predator odor stress-paired contexts (termed Avoider rats), and that chemogenetic inhibition of these cells rescues stress-induced increases in anxiety-like behavior and alcohol self-administration in male and female Avoider rats. Using slice electrophysiology, we found that prior predator odor stress exposure blunts inhibitory synaptic transmission and increases synaptic drive in CRF1 CeA-LH cells. In addition, we found that CRF bath application reduces synaptic drive in CRF1 CeA-LH cells in Non-Avoiders only. Collectively, these data show that CRF1 CeA-LH cells contribute to stress-induced increases in anxiety-like behavior and alcohol self-administration in male and female Avoider rats.SIGNIFICANCE STATEMENT Stress may lead to a variety of behavioral and physiological negative consequences, and better understanding of the neurobiological mechanisms that contribute to negative stress effects may lead to improved prevention and treatment strategies. This study, performed in laboratory rats, shows that animals that exhibit avoidance stress coping go on to develop heightened anxiety-like behavior and alcohol self-administration, and that these behaviors can be rescued by inhibiting the activity of a specific population of neurons in the central amygdala. This study also describes stress-induced physiological changes in these neurons that may contribute to their role in promoting increased anxiety and alcohol self-administration.
Collapse
Affiliation(s)
- Marcus M Weera
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Daniel A Webster
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Rosetta S Shackett
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Federica Benvenuti
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Jason W Middleton
- Department of Cell Biology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Nicholas W Gilpin
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Southeast Louisiana VA Healthcare System, New Orleans, Louisiana 70119
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
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: 15] [Impact Index Per Article: 15.0] [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
|
11
|
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: 4.0] [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
|
12
|
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: 3.0] [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
|
13
|
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: 1] [Impact Index Per Article: 0.5] [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
|
14
|
Jacotte-Simancas A, Middleton JW, Stielper ZF, Edwards S, Molina PE, Gilpin NW. Brain Injury Effects on Neuronal Activation and Synaptic Transmission in the Basolateral Amygdala of Adult Male and Female Wistar Rats. J Neurotrauma 2022; 39:544-559. [PMID: 35081744 PMCID: PMC8978566 DOI: 10.1089/neu.2021.0270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Traumatic brain injury (TBI) is defined as brain damage produced by an external mechanical force that leads to behavioral, cognitive, and psychiatric sequelae. The basolateral amygdala (BLA) is involved in emotional regulation, and its function and morphology are altered following TBI. Little is known about potential sex-specific effects of TBI on BLA neuronal function, but it is critical for the field to identify potential sex differences in TBI effects on brain and behavior. Here, we hypothesized that TBI would produce sex-specific acute (1 h) effects on BLA neuronal activation, excitability, and synaptic transmission in adult male and female rats. Forty-nine Wistar rats (n = 23 males and 26 females) were randomized to TBI (using lateral fluid percussion) or Sham groups in two separate studies. Study 1 used in situ hybridization (i.e., RNAscope) to measure BLA expression of c-fos (a marker of cell activation), vGlut, and vGat (markers of glutamatergic and GABAergic neurons, respectively) messenger RNA (mRNA). Study 2 used slice electrophysiology to measure intrinsic excitability and excitatory/inhibitory synaptic transmission in putative pyramidal neurons in the BLA. Physiological measures of injury severity were collected from all animals. Our results show that females exhibit increased apnea duration and reduced respiratory rate post-TBI relative to males. In male and female rats, TBI increased c-fos expression in BLA glutamatergic cells but not in BLA GABAergic cells, and TBI increased firing rate in BLA pyramidal neurons. Further, TBI increased spontaneous excitatory and inhibitory postsynaptic current (sEPSC and sIPSC) amplitude in BLA neurons of females relative to all other groups. TBI increased sEPSC frequency in BLA neurons of females relative to males but did not alter sIPSC frequency. In summary, lateral fluid percussion produced different physiological responses in male and female rats, as well as sex-specific alterations in BLA neuronal activation, excitability, and synaptic transmission 1 h after injury.
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
| | - Jason W. Middleton
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zachary F. Stielper
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Scott Edwards
- 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
|
15
|
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: 19] [Impact Index Per Article: 9.5] [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
|
16
|
Chen S, Siedhoff HR, Zhang H, Liu P, Balderrama A, Li R, Johnson C, Greenlief CM, Koopmans B, Hoffman T, DePalma RG, Li DP, Cui J, Gu Z. Low-intensity blast induces acute glutamatergic hyperexcitability in mouse hippocampus leading to long-term learning deficits and altered expression of proteins involved in synaptic plasticity and serine protease inhibitors. Neurobiol Dis 2022; 165:105634. [PMID: 35077822 DOI: 10.1016/j.nbd.2022.105634] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 11/26/2022] Open
Abstract
Neurocognitive consequences of blast-induced traumatic brain injury (bTBI) pose significant concerns for military service members and veterans with the majority of "invisible injury." However, the underlying mechanism of such mild bTBI by low-intensity blast (LIB) exposure for long-term cognitive and mental deficits remains elusive. Our previous studies have shown that mice exposed to LIB result in nanoscale ultrastructural abnormalities in the absence of gross or apparent cellular damage in the brain. Here we tested the hypothesis that glutamatergic hyperexcitability may contribute to long-term learning deficits. Using brain slice electrophysiological recordings, we found an increase in averaged frequencies with a burst pattern of miniature excitatory postsynaptic currents (mEPSCs) in hippocampal CA3 neurons in LIB-exposed mice at 1- and 7-days post injury, which was blocked by a specific NMDA receptor antagonist AP5. In addition, cognitive function assessed at 3-months post LIB exposure by automated home-cage monitoring showed deficits in dynamic patterns of discrimination learning and cognitive flexibility in LIB-exposed mice. Collected hippocampal tissue was further processed for quantitative global-proteomic analysis. Advanced data-independent acquisition for quantitative tandem mass spectrometry analysis identified altered expression of proteins involved in synaptic plasticity and serine protease inhibitors in LIB-exposed mice. Some were correlated with the ability of discrimination learning and cognitive flexibility. These findings show that acute glutamatergic hyperexcitability in the hippocampus induced by LIB may contribute to long-term cognitive dysfunction and protein alterations. Studies using this military-relevant mouse model of mild bTBI provide valuable insights into developing a potential therapeutic strategy to ameliorate hyperexcitability-modulated LIB injuries.
Collapse
Affiliation(s)
- Shanyan Chen
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Heather R Siedhoff
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Hua Zhang
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Pei Liu
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, MO 65211, USA
| | - Ashley Balderrama
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Runting Li
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Catherine Johnson
- Department of Mining and Nuclear Engineering, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | - C Michael Greenlief
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, MO 65211, USA
| | | | - Timothy Hoffman
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Ralph G DePalma
- Office of Research and Development, Department of Veterans Affairs, Washington DC 20420, USA; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - De-Pei Li
- Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Jiankun Cui
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - Zezong Gu
- Truman VA Hospital Research Service, Columbia, MO 65201, USA; Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
17
|
Ponomarenko A, Tyrtyshnaia A, Ivashkevich D, Manzhulo I. Mild Traumatic Brain Injury Contributes to the Development of Delayed Neuroinflammation. Neuroimmunomodulation 2022; 29:135-142. [PMID: 34583355 DOI: 10.1159/000519011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/10/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION In recent years, according to the literature, the problem of mild traumatic brain injury (mTBI) has become more and more urgent. Compared to moderate to severe craniocerebral trauma, mTBI occurs in a far greater number of people. The delayed sequelae caused by a single mTBI or multiple mTBIs are a significant public health problem. METHODS A weight-drop model was used for the formation of mTBI. A metal rod weighing 337 g with a blunt tip of 3 mm diameter was uplifted at 8 cm height and held by a lever. The trauma was created by lowering the lever and the rod and free-dropping onto the rat skull. In the cerebral cortex of experimental animals, we analyzed the level of microglial activity (Iba-1-positive system) and the expression of pro-inflammatory markers (IL1β, IL6, and CD86). Also, the expression level of the endocannabinoid system receptor (cannabinoid receptor type 1 [CB1]) was assessed in brain samples. RESULTS Experiments have shown that mTBI increases (1) the amount of microglia (iba-1) activated by the pro-inflammatory pathway (CD86); (2) the level of pro-inflammatory cytokines IL1β and IL6; and (3) CB1R activity. CONCLUSION Overall, the results of this study indicate that mTBI induces a sustained neuroinflammatory response.
Collapse
Affiliation(s)
- Arina Ponomarenko
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Anna Tyrtyshnaia
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Darya Ivashkevich
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Igor Manzhulo
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| |
Collapse
|
18
|
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
|
19
|
Browne CA, Hildegard A Wulf BA, Jacobson ML, Oyola M, Wu TJ, Lucki I. Long-term increase in sensitivity to ketamine's behavioral effects in mice exposed to mild blast induced traumatic brain injury. Exp Neurol 2021; 350:113963. [PMID: 34968423 DOI: 10.1016/j.expneurol.2021.113963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/23/2021] [Accepted: 12/22/2021] [Indexed: 11/04/2022]
Abstract
Neurobehavioral deficits emerge in nearly 50% of patients following a mild traumatic brain injury (TBI) and may persist for months. Ketamine is used frequently as an anesthetic, analgesic and for management of persistent psychiatric complications. Although ketamine may produce beneficial effects in patients with a history of TBI, differential sensitivity to its impairing effects could make the therapeutic use of ketamine in TBI patients unsafe. This series of studies examined male C57BL/6 J mice exposed to a mild single blast overpressure (mbTBI) for indications of altered sensitivity to ketamine at varying times after injury. Dystaxia (altered gait), diminished sensorimotor gating (reduced prepulse inhibition) impaired working memory (step-down inhibitory avoidance) were examined in mbTBI and sham animals 15 min following intraperitoneal injections of saline or R,S-ketamine hydrochloride, from day 7-16 post injury and again from day 35-43 post injury. Behavioral performance in the forced swim test and sucrose preference test were evaluated on day 28 and day 74 post injury respectively, 24 h following drug administration. Dynamic gait stability was compromised in mbTBI mice on day 7 and 35 post injury and further exacerbated following ketamine administration. On day 14 and 42 post injury, prepulse inhibition was robustly decreased by mbTBI, which ketamine further reduced. Ketamine-associated memory impairment was apparent selectively in mbTBI animals 1 h, 24 h and day 28 post shock (tested on day 15/16/43 post injury). Ketamine selectively reduced immobility scores in the FST in mbTBI animals (day 28) and reversed mbTBI induced decreases in sucrose consumption (Day 74). These results demonstrate increased sensitivity to ketamine in mice when tested for extended periods after TBI. The results suggest that ketamine may be effective for treating neuropsychiatric complications that emerge after TBI but urge caution when used in clinical practice for enhanced sensitivity to its side effects in this patient population.
Collapse
Affiliation(s)
- Caroline A Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America.
| | - B A Hildegard A Wulf
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Moriah L Jacobson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Mario Oyola
- Department of Gynecologic Surgery & Obstetrics, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - T John Wu
- Department of Gynecologic Surgery & Obstetrics, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America
| |
Collapse
|
20
|
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: 2] [Impact Index Per Article: 0.7] [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
|
21
|
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: 2.7] [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
|
22
|
Kasatkina LA, Rittchen S, Sturm EM. Neuroprotective and Immunomodulatory Action of the Endocannabinoid System under Neuroinflammation. Int J Mol Sci 2021; 22:ijms22115431. [PMID: 34063947 PMCID: PMC8196612 DOI: 10.3390/ijms22115431] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/17/2022] Open
Abstract
Endocannabinoids (eCBs) are lipid-based retrograde messengers with a relatively short half-life that are produced endogenously and, upon binding to the primary cannabinoid receptors CB1/2, mediate multiple mechanisms of intercellular communication within the body. Endocannabinoid signaling is implicated in brain development, memory formation, learning, mood, anxiety, depression, feeding behavior, analgesia, and drug addiction. It is now recognized that the endocannabinoid system mediates not only neuronal communications but also governs the crosstalk between neurons, glia, and immune cells, and thus represents an important player within the neuroimmune interface. Generation of primary endocannabinoids is accompanied by the production of their congeners, the N-acylethanolamines (NAEs), which together with N-acylneurotransmitters, lipoamino acids and primary fatty acid amides comprise expanded endocannabinoid/endovanilloid signaling systems. Most of these compounds do not bind CB1/2, but signal via several other pathways involving the transient receptor potential cation channel subfamily V member 1 (TRPV1), peroxisome proliferator-activated receptor (PPAR)-α and non-cannabinoid G-protein coupled receptors (GPRs) to mediate anti-inflammatory, immunomodulatory and neuroprotective activities. In vivo generation of the cannabinoid compounds is triggered by physiological and pathological stimuli and, specifically in the brain, mediates fine regulation of synaptic strength, neuroprotection, and resolution of neuroinflammation. Here, we review the role of the endocannabinoid system in intrinsic neuroprotective mechanisms and its therapeutic potential for the treatment of neuroinflammation and associated synaptopathy.
Collapse
Affiliation(s)
- Ludmila A. Kasatkina
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sonja Rittchen
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
| | - Eva M. Sturm
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
- Correspondence:
| |
Collapse
|
23
|
Hergert DC, Robertson-Benta C, Sicard V, Schwotzer D, Hutchison K, Covey DP, Quinn DK, Sadek JR, McDonald J, Mayer AR. Use of Medical Cannabis to Treat Traumatic Brain Injury. J Neurotrauma 2021; 38:1904-1917. [PMID: 33256496 DOI: 10.1089/neu.2020.7148] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There is not a single pharmacological agent with demonstrated therapeutic efficacy for traumatic brain injury (TBI). With recent legalization efforts and the growing popularity of medical cannabis, patients with TBI will inevitably consider medical cannabis as a treatment option. Pre-clinical TBI research suggests that cannabinoids have neuroprotective and psychotherapeutic properties. In contrast, recreational cannabis use has consistently shown to have detrimental effects. Our review identified a paucity of high-quality studies examining the beneficial and adverse effects of medical cannabis on TBI, with only a single phase III randomized control trial. However, observational studies demonstrate that TBI patients are using medical and recreational cannabis to treat their symptoms, highlighting inconsistencies between public policy, perception of potential efficacy, and the dearth of empirical evidence. We conclude that randomized controlled trials and prospective studies with appropriate control groups are necessary to fully understand the efficacy and potential adverse effects of medical cannabis for TBI.
Collapse
Affiliation(s)
- Danielle C Hergert
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, Albuquerque, New Mexico, USA
| | - Cidney Robertson-Benta
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, Albuquerque, New Mexico, USA
| | - Veronik Sicard
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, Albuquerque, New Mexico, USA
| | - Daniela Schwotzer
- Lovelace Biomedical and Environmental Research Institute, Albuquerque, New Mexico, USA
| | - Kent Hutchison
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado, USA
| | - Dan P Covey
- Lovelace Biomedical and Environmental Research Institute, Albuquerque, New Mexico, USA
| | - Davin K Quinn
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Joseph R Sadek
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| | - Jacob McDonald
- Lovelace Biomedical and Environmental Research Institute, Albuquerque, New Mexico, USA
| | - Andrew R Mayer
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Pete & Nancy Domenici Hall, Albuquerque, New Mexico, USA.,Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Psychology Department, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
24
|
Central Amygdala Projections to Lateral Hypothalamus Mediate Avoidance Behavior in Rats. J Neurosci 2021; 41:61-72. [PMID: 33188067 DOI: 10.1523/jneurosci.0236-20.2020] [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: 01/30/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 11/21/2022] Open
Abstract
Persistent avoidance of stress-related stimuli following acute stress exposure predicts negative outcomes such as substance abuse and traumatic stress disorders. Previous work using a rat model showed that the central amygdala (CeA) plays an important role in avoidance of a predator odor stress-paired context. Here, we show that CeA projections to the lateral hypothalamus (LH) are preferentially activated in male rats that show avoidance of a predator odor-paired context (termed Avoider rats), that chemogenetic inhibition of CeA-LH projections attenuates avoidance in male Avoider rats, that chemogenetic stimulation of the CeA-LH circuit produces conditioned place avoidance (CPA) in otherwise naive male rats, and that avoidance behavior is associated with intrinsic properties of LH-projecting CeA cells. Collectively, these data show that CeA-LH projections are important for persistent avoidance of stress-related stimuli following acute stress exposure.SIGNIFICANCE STATEMENT This study in rats shows that a specific circuit in the brain [i.e., neurons that project from the central amygdala (CeA) to the lateral hypothalamus (LH)] mediates avoidance of stress-associated stimuli. In addition, this study shows that intrinsic physiological properties of cells in this brain circuit are associated with avoidance of stress-associated stimuli. Further characterization of the CeA-LH circuit may improve our understanding of the neural mechanisms underlying specific aspects of stress-related disorders in humans.
Collapse
|
25
|
Stielper ZF, Fucich EA, Middleton JW, Hillard CJ, Edwards S, Molina PE, Gilpin NW. Traumatic Brain Injury and Alcohol Drinking Alter Basolateral Amygdala Endocannabinoids in Female Rats. J Neurotrauma 2020; 38:422-434. [PMID: 32838651 DOI: 10.1089/neu.2020.7175] [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: 12/24/2022] Open
Abstract
Traumatic brain injury (TBI) affects approximately 3 million Americans yearly and increases vulnerability to developing psychiatric comorbidities. Alcohol use disorder (AUD) is the most prevalent psychiatric diagnosis preceding injury and TBI may increase subsequent alcohol use. The basolateral amygdala (BLA) is a limbic structure commonly affected by TBI that is implicated in anxiety and AUD. Endocannabinoids (eCBs) regulate synaptic activity in the BLA, and BLA eCB modulation alters anxiety-like behavior and stress reactivity. Previous work from our laboratories showed that systemic eCB degradation inhibition ameliorates TBI-induced increases in anxiety-like behavior and motivation to respond for alcohol in male rats. Here, we used a lateral fluid percussion model to test moderate TBI effects on anxiety-like behavior, alcohol drinking, and eCB levels and cell signaling in BLA, as well as the effect of alcohol drinking on anxiety-like behavior and the BLA eCB system, in female rats. Our results show that TBI does not promote escalation of operant alcohol self-administration or increase anxiety-like behavior in female rats. In the BLA, TBI and alcohol drinking alter tissue amounts of 2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamine (anandamide; AEA) 1 h post-injury, and 2-AG levels remain low 11 days post-injury. Eleven days after injury, BLA pyramidal neurons were hyperexcitable, but measures of synaptic transmission and eCB signaling were unchanged. These data show that TBI impacts BLA 2-AG tissue levels, that this effect is modified by alcohol drinking, and also that TBI increases BLA cell excitability.
Collapse
Affiliation(s)
- Zachary F Stielper
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Elizabeth A Fucich
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Neuroscience Program, Tulane University, New Orleans, Louisiana, USA
| | - Jason W Middleton
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Alcohol and Drug 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 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 Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Southeast Louisiana VA Healthcare System, New Orleans, Louisiana, USA
| |
Collapse
|
26
|
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: 5.5] [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
|
27
|
Cannabinoid agonists possibly mediate interaction between cholinergic and cannabinoid systems in regulating intestinal inflammation. Med Hypotheses 2020; 139:109613. [DOI: 10.1016/j.mehy.2020.109613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 02/07/2023]
|
28
|
Fucich EA, Stielper ZF, Cancienne HL, Edwards S, Gilpin NW, Molina PE, Middleton JW. Endocannabinoid degradation inhibitors ameliorate neuronal and synaptic alterations following traumatic brain injury. J Neurophysiol 2020; 123:707-717. [PMID: 31913777 PMCID: PMC7052644 DOI: 10.1152/jn.00570.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 12/13/2022] Open
Abstract
Our previous work showed that lateral fluid percussion injury to the sensorimotor cortex (SMC) of anesthetized rats increased neuronal synaptic hyperexcitability in layer 5 (L5) neurons in ex vivo brain slices 10 days postinjury. Furthermore, endocannabinoid (EC) degradation inhibition via intraperitoneal JZL184 injection 30 min postinjury attenuated synaptic hyperexcitability. This study tested the hypothesis that traumatic brain injury (TBI) induces synaptic and intrinsic neuronal alterations of L5 SMC pyramidal neurons and that these alterations are significantly attenuated by in vivo post-TBI treatment with EC degradation inhibitors. We tested the effects of systemically administered EC degradation enzyme inhibitors (JZL184, MJN110, URB597, or JZL195) with differential selectivity for fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) on electrophysiological parameters in SMC neurons of TBI- and sham-treated rats 10 days post-TBI. We recorded intrinsic neuronal properties, including resting membrane voltage, input resistance, spike threshold, spiking responses to current input, voltage "sag" (rebound response to hyperpolarization-activated inward current), and burst firing. We also measured the frequency and amplitude of spontaneous excitatory postsynaptic currents. We then used the aggregate parameter sets (intrinsic + synaptic properties) to apply a machine learning classification algorithm to quantitatively compare neural population responses from each experimental group. Collectively, our electrophysiological and computational results indicate that sham neurons are the most distinguishable from TBI neurons. Administration of EC degradation inhibitors post-TBI exerted varying degrees of rescue, approximating the neuronal phenotype of sham neurons, with neurons from TBI/JZL195 (a dual MAGL/FAAH inhibitor) being most similar to neurons from sham rats.NEW & NOTEWORTHY This study elucidates neuronal properties altered by traumatic brain injury (TBI) in layer 5 of sensorimotor cortex, which may be implicated in post-TBI circuit dysfunction. We compared effects of systemic administration of four different endocannabinoid degradation inhibitors within a clinically relevant window postinjury. Electrophysiological measures and using a machine learning classification algorithm collectively suggest that pharmacological inhibitors targeting both monoacylglycerol lipase and fatty acid amide hydrolase (e.g., JZL195) may be most efficacious in attenuating TBI-induced neuronal dysfunction at site of injury.
Collapse
Affiliation(s)
- Elizabeth A Fucich
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Zachary F Stielper
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Heather L Cancienne
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Scott Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Patricia E Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Jason W Middleton
- Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
29
|
Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat Rev Neurol 2019; 16:9-29. [DOI: 10.1038/s41582-019-0284-z] [Citation(s) in RCA: 320] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
|
30
|
Monoacylglycerol Lipase Inactivation by Using URB602 Mitigates Myocardial Damage in a Rat Model of Cardiac Arrest. Crit Care Med 2019; 47:e144-e151. [PMID: 30431495 DOI: 10.1097/ccm.0000000000003552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Monoacylglycerol lipase participates in organ protection by regulating the hydrolysis of the endocannabinoid 2-arachidonoylglycerol. This study investigated whether blocking monoacylglycerol lipase protects against postresuscitation myocardial injury and improves survival in a rat model of cardiac arrest and cardiopulmonary resuscitation. DESIGN Prospective randomized laboratory study. SETTING University research laboratory. SUBJECTS Male Sprague-Dawley rat (n = 96). INTERVENTIONS Rats underwent 8-minute asphyxia-based cardiac arrest and resuscitation. Surviving rats were randomly divided into cardiopulmonary resuscitation + URB602 group, cardiopulmonary resuscitation group, and sham group. One minute after successful resuscitation, rats in the cardiopulmonary resuscitation + URB602 group received a single dose of URB602 (5 mg/kg), a small-molecule monoacylglycerol lipase inhibitor, whereas rats in the cardiopulmonary resuscitation group received an equivalent volume of vehicle solution. The sham rats underwent all of the procedures performed on rats in the cardiopulmonary resuscitation and cardiopulmonary resuscitation + URB602 groups minus cardiac arrest and asphyxia. MEASUREMENTS AND MAIN RESULTS Survival was recorded 168 hours after the return of spontaneous circulation (n = 22 in each group). Compared with vehicle treatment (31.8%), URB602 treatment markedly improved survival (63.6%) 168 hours after cardiopulmonary resuscitation. Next, we used additional surviving rats to evaluate myocardial and mitochondrial injury 6 hours after return of spontaneous circulation, and we found that URB602 significantly reduced myocardial injury and prevented myocardial mitochondrial damage. In addition, URB602 attenuated the dysregulation of endocannabinoid and eicosanoid metabolism 6 hours after return of spontaneous circulation and prevented the acceleration of mitochondrial permeability transition 15 minutes after return of spontaneous circulation. CONCLUSIONS Monoacylglycerol lipase blockade may reduce myocardial and mitochondrial injury and significantly improve the resuscitation effect after cardiac arrest and cardiopulmonary resuscitation.
Collapse
|
31
|
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: 10] [Impact Index Per Article: 2.0] [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
|
32
|
Chinnadurai A, Berger G, Burkovskiy I, Zhou J, Cox A, Lynch M, Lehmann C. Monoacylglycerol lipase inhibition as potential treatment for interstitial cystitis. Med Hypotheses 2019; 131:109321. [PMID: 31443753 DOI: 10.1016/j.mehy.2019.109321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022]
Abstract
Interstitial cystitis is a chronic inflammatory condition of the urinary bladder with an unclear etiology. Currently, there are no widely accepted long-term treatment options available for patients with IC, with the European Association of Urology (EAU, 2017 guidelines), American Urology Association (AUA, 2014 guidelines), and the Royal College of Obstetricians and Gynaecologists (RCOG, 2016 guidelines) all suggesting various different conservative, pharmacological, intravesical, and surgical interventions. The endocannabinoid system represents a potential target for IC treatment and management. Activation of cannabinoid receptor 2 (CBR2) with various agonists has previously been shown to reduce leukocyte differentiation and migration, in addition to inhibiting the release of pro-inflammatory cytokines at the site of inflammation. These receptors have been identified in the detrusor and sensory nerves of the urothelium in various mammalian species, including humans. We hypothesize that by inhibiting the enzymes responsible for the catabolism of endogenous cannabinoids locally, bladder concentrations of CBR2 agonists will increase, particularly 2-arachidonyl glycerol, resulting in a diminished inflammatory response.
Collapse
Affiliation(s)
- Anu Chinnadurai
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Geraint Berger
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian Burkovskiy
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Juan Zhou
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Ashley Cox
- Department of Urology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mary Lynch
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Christian Lehmann
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
33
|
Bittar A, Bhatt N, Hasan TF, Montalbano M, Puangmalai N, McAllen S, Ellsworth A, Carretero Murillo M, Taglialatela G, Lucke-Wold B, Logsdon A, Rosen C, Turner RC, Kayed R. Neurotoxic tau oligomers after single versus repetitive mild traumatic brain injury. Brain Commun 2019; 1:fcz004. [PMID: 31608324 PMCID: PMC6777515 DOI: 10.1093/braincomms/fcz004] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 02/05/2023] Open
Abstract
Mild traumatic brain injury accounts for the majority of head injuries and has been correlated with neurodegeneration and dementia. While repetitive mild traumatic brain injury is highly correlated to neurodegeneration, the correlation of a single mild traumatic brain injury with neurodegeneration is still unclear. Because tau aggregates are the main form of mild traumatic brain injury induced pathology, toxic forms of tau protein most likely play a role in the development of post-mild traumatic brain injury neurodegeneration. Therefore, it becomes crucial to characterize the properties of soluble tau aggregates in single versus repetitive mild traumatic brain injury. Herein, we isolated tau oligomers from wild-type mice exposed to single or repetitive mild traumatic brain injury and characterized the tau aggregates at functional, biochemical and biophysical levels. We demonstrated that single versus repetitive mild traumatic brain injuries frequencies lead to the formation of different tau oligomeric polymorphisms. These polymorphisms express different long-term potentiation impairment potencies, toxicity potentials, morphologies and strain indicating properties. To our knowledge, this is the first evidence that soluble tau oligomers derived from single versus repetitive mild traumatic brain injuries form distinct polymorphisms that possibly correlate with the risk of neurodegeneration after mild traumatic brain injury.
Collapse
Affiliation(s)
- Alice Bittar
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| | - Nemil Bhatt
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| | - Tasneem F Hasan
- Department of Neurology, Ochsner Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA
| | - Mauro Montalbano
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| | - Nicha Puangmalai
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| | - Salome McAllen
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| | - Anna Ellsworth
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| | | | - Giulio Taglialatela
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| | - Aric Logsdon
- Department of Psychiatry, University of Washington, Seattle, WA 98195, USA
| | - Charles Rosen
- Central Illinois Neural Health Sciences, Bloomington, IL 61701, USA
| | - Ryan C Turner
- Department of Neurosurgery, Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Rakez Kayed
- Department of Neurology, The Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555-1045, USA
| |
Collapse
|
34
|
Ko J, Hemphill M, Yang Z, Beard K, Sewell E, Shallcross J, Schweizer M, Sandsmark DK, Diaz-Arrastia R, Kim J, Meaney D, Issadore D. Multi-Dimensional Mapping of Brain-Derived Extracellular Vesicle MicroRNA Biomarker for Traumatic Brain Injury Diagnostics. J Neurotrauma 2019; 37:2424-2434. [PMID: 30950328 DOI: 10.1089/neu.2018.6220] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The diagnosis and prognosis of traumatic brain injury (TBI) is complicated by variability in the type and severity of injuries and the multiple endophenotypes that describe each patient's response and recovery to the injury. It has been challenging to capture the multiple dimensions that describe an injury and its recovery to provide clinically useful information. To address this challenge, we have performed an open-ended search for panels of microRNA (miRNA) biomarkers, packaged inside of brain-derived extracellular vesicles (EVs), that can be combined algorithmically to accurately classify various states of injury. We mapped GluR2+ EV miRNA across a variety of injury types, injury intensities, history of injuries, and time elapsed after injury, and sham controls in a pre-clinical murine model (n = 116), as well as in clinical samples (n = 36). We combined next-generation sequencing with a technology recently developed by our lab, Track Etched Magnetic Nanopore (TENPO) sorting, to enrich for GluR2+ EVs and profile their miRNA. By mapping and comparing brain-derived EV miRNA between various injuries, we have identified signaling pathways in the packaged miRNA that connect these biomarkers to underlying mechanisms of TBI. Many of these pathways are shared between the pre-clinical model and the clinical samples, and present distinct signatures across different injury models and times elapsed after injury. Using this map of EV miRNA, we applied machine learning to define a panel of biomarkers to successfully classify specific states of injury, paving the way for a prognostic blood test for TBI. We generated a panel of eight miRNAs (miR-150-5p, miR-669c-5p, miR-488-3p, miR-22-5p, miR-9-5p, miR-6236, miR-219a.2-3p, miR-351-3p) for injured mice versus sham mice and four miRNAs (miR-203b-5p, miR-203a-3p, miR-206, miR-185-5p) for TBI patients versus healthy controls.
Collapse
Affiliation(s)
- Jina Ko
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew Hemphill
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zijian Yang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kryshawna Beard
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emily Sewell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jamie Shallcross
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Melissa Schweizer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Danielle K Sandsmark
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Junhyong Kim
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Computer and Information Science, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
Belardo C, Iannotta M, Boccella S, Rubino RC, Ricciardi F, Infantino R, Pieretti G, Stella L, Paino S, Marabese I, Maisto R, Luongo L, Maione S, Guida F. Oral Cannabidiol Prevents Allodynia and Neurological Dysfunctions in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2019; 10:352. [PMID: 31040777 PMCID: PMC6476923 DOI: 10.3389/fphar.2019.00352] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/21/2019] [Indexed: 01/28/2023] Open
Abstract
Neurological dysfunctions are the most impactful and persistent consequences of traumatic brain injury (TBI). Indeed, previous reports suggest that an association between TBI and chronic pain syndromes, as well anxio-depressive behaviors, tends to be more common in patients with mild forms of TBI. At present, no effective treatment options are available for these symptoms. In the present study, we used a weight drop mild TBI mouse model to investigate the effect of a commercially available 10% Cannabidiol (CBD) oil on both the sensorial and neuropsychiatric dysfunctions associated with mild TBI through behavioral and biomolecular approaches. TBI mice developed chronic pain associated with anxious and aggressive behavior, followed by a late depressive-like behavior and impaired social interaction. Such behaviors were related with specific changes in neurotransmitters release at cortical levels. CBD oral treatment restored the behavioral alterations and partially normalized the cortical biochemical changes. In conclusion, our data show some of the brain modifications probably responsible for the behavioral phenotype associated with TBI and suggest the CBD as a pharmacological tool to improve neurological dysfunctions caused by the trauma.
Collapse
Affiliation(s)
- Carmela Belardo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Flavia Ricciardi
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Salvatore Paino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
36
|
McGuire JL, DePasquale EAK, Watanabe M, Anwar F, Ngwenya LB, Atluri G, Romick-Rosendale LE, McCullumsmith RE, Evanson NK. Chronic Dysregulation of Cortical and Subcortical Metabolism After Experimental Traumatic Brain Injury. Mol Neurobiol 2019; 56:2908-2921. [PMID: 30069831 PMCID: PMC7584385 DOI: 10.1007/s12035-018-1276-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/23/2018] [Indexed: 02/03/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and long-term disability worldwide. Although chronic disability is common after TBI, effective treatments remain elusive and chronic TBI pathophysiology is not well understood. Early after TBI, brain metabolism is disrupted due to unregulated ion release, mitochondrial damage, and interruption of molecular trafficking. This metabolic disruption causes at least part of the TBI pathology. However, it is not clear how persistent or pervasive metabolic injury is at later stages of injury. Using untargeted 1H-NMR metabolomics, we examined ex vivo hippocampus, striatum, thalamus, frontal cortex, and brainstem tissue in a rat lateral fluid percussion model of chronic brain injury. We found altered tissue concentrations of metabolites in the hippocampus and thalamus consistent with dysregulation of energy metabolism and excitatory neurotransmission. Furthermore, differential correlation analysis provided additional evidence of metabolic dysregulation, most notably in brainstem and frontal cortex, suggesting that metabolic consequences of injury are persistent and widespread. Interestingly, the patterns of network changes were region-specific. The individual metabolic signatures after injury in different structures of the brain at rest may reflect different compensatory mechanisms engaged to meet variable metabolic demands across brain regions.
Collapse
Affiliation(s)
- Jennifer L McGuire
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, 45267, USA.
| | - Erica A K DePasquale
- Graduate Program in Biomedical Informatics, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Miki Watanabe
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Fatima Anwar
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Laura B Ngwenya
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, 45267, USA
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Gowtham Atluri
- Graduate Program in Biomedical Informatics, University of Cincinnati, Cincinnati, OH, 45267, USA
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45267, USA
| | | | - Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Nathan K Evanson
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, 45267, USA
- Division of Pediatric Rehabilitation Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| |
Collapse
|
37
|
Fucich EA, Mayeux JP, McGinn MA, Gilpin NW, Edwards S, Molina PE. A Novel Role for the Endocannabinoid System in Ameliorating Motivation for Alcohol Drinking and Negative Behavioral Affect after Traumatic Brain Injury in Rats. J Neurotrauma 2019; 36:1847-1855. [PMID: 30638118 DOI: 10.1089/neu.2018.5854] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with psychiatric dysfunction-including pain, cognitive impairment, anxiety, and increased alcohol use. We previously demonstrated that inhibiting endocannabinoid degradation post-TBI with JZL184 attenuates neuroinflammation and neuronal hyperexcitability at the site of injury and improves neurobehavioral recovery. This study aimed to determine the effect of JZL184 on post-TBI behavioral changes related to psychiatric dysfunction and post-TBI neuroadaptations in brain regions associated with these behaviors. We hypothesized that JZL184 would attenuate post-TBI behavioral and neural changes in alcohol-drinking rats. Adult male Wistar rats were trained to operantly self-administer alcohol before receiving lateral fluid percussion injury. Thirty minutes post-TBI, rats received JZL184 (16 mg/kg, i.p.) or vehicle. Spatial memory (Y-maze), anxiety-like behavior (open field), alcohol motivation (progressive ratio responding), and mechanosensitivity (Von Frey) were measured 3-10 days post-injury, and ventral striatum (VS) and central amygdala (CeA) tissue were collected for western blot analysis of phosphorylated glutamate receptor subunit 1 (GluR1) and glucocorticoid receptor (GR). TBI impaired spatial memory, increased anxiety-like behavior, and increased motivated alcohol drinking. JZL184 prevented these changes. TBI also increased phosphorylated GluR1 and GR in the CeA (but not the VS) compared with sham controls. JZL184 attenuated post-TBI GR phosphorylation in the CeA. These findings suggest that TBI produces comorbid cognitive dysfunction, increased alcohol motivation, and anxiety-like behavior, possibly related to amygdala dysfunction, and these changes are prevented by systemic post-TBI endocannabinoid degradation inhibition. Thus, boosting endocannabinoid tone post-TBI may represent a viable therapeutic strategy for TBI-related psychiatric comorbidities such as alcohol use disorder and anxiety.
Collapse
Affiliation(s)
- Elizabeth A Fucich
- 1 Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,2 Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Jacques P Mayeux
- 1 Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,2 Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - M Adrienne McGinn
- 1 Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,2 Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Nicholas W Gilpin
- 1 Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,2 Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Scott Edwards
- 1 Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,2 Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Patricia E Molina
- 1 Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,2 Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
38
|
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: 3.2] [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
|
39
|
Leishman E, Kunkler PE, Hurley JH, Miller S, Bradshaw HB. Bioactive Lipids in Cancer, Inflammation and Related Diseases : Acute and Chronic Mild Traumatic Brain Injury Differentially Changes Levels of Bioactive Lipids in the CNS Associated with Headache. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1161:193-217. [PMID: 31562631 DOI: 10.1007/978-3-030-21735-8_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Headache is a common complaint after mild traumatic brain injury (mTBI). Changes in the CNS lipidome were previously associated with acrolein-induced headache in rodents. mTBI caused similar headache-like symptoms in rats; therefore, we tested the hypothesis that mTBI might likewise alter the lipidome. Using a stereotaxic impactor, rats were given either a single mTBI or a series of 4 mTBIs 48 h apart. 72 h later for single mTBI and 7 days later for repeated mTBI, the trigeminal ganglia (TG), trigeminal nucleus (TNC), and cerebellum (CER) were isolated. Using HPLC/MS/MS, ~80 lipids were measured in each tissue and compared to sham controls. mTBI drove widespread alterations in lipid levels. Single mTBI increased arachidonic acid and repeated mTBI increased prostaglandins in all 3 tissue types. mTBI affected multiple TRPV agonists, including N-arachidonoyl ethanolamine (AEA), which increased in the TNC and CER after single mTBI. After repeated mTBI, AEA increased in the TG, but decreased in the TNC. Common to all tissue types in single and repeated mTBI was an increase the AEA metabolite, N-arachidonoyl glycine, a potent activator of microglial migration. Changes in the CNS lipidome associated with mTBI likely play a role in headache and in long-term neurodegenerative effects of repeated mTBI.
Collapse
Affiliation(s)
- Emma Leishman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Phillip E Kunkler
- Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joyce H Hurley
- Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sally Miller
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Heather B Bradshaw
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
40
|
Belardo C, Iannotta M, Boccella S, Rubino RC, Ricciardi F, Infantino R, Pieretti G, Stella L, Paino S, Marabese I, Maisto R, Luongo L, Maione S, Guida F. Oral Cannabidiol Prevents Allodynia and Neurological Dysfunctions in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2019. [PMID: 31040777 DOI: 10.3389/fphar.2019.00352/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Neurological dysfunctions are the most impactful and persistent consequences of traumatic brain injury (TBI). Indeed, previous reports suggest that an association between TBI and chronic pain syndromes, as well anxio-depressive behaviors, tends to be more common in patients with mild forms of TBI. At present, no effective treatment options are available for these symptoms. In the present study, we used a weight drop mild TBI mouse model to investigate the effect of a commercially available 10% Cannabidiol (CBD) oil on both the sensorial and neuropsychiatric dysfunctions associated with mild TBI through behavioral and biomolecular approaches. TBI mice developed chronic pain associated with anxious and aggressive behavior, followed by a late depressive-like behavior and impaired social interaction. Such behaviors were related with specific changes in neurotransmitters release at cortical levels. CBD oral treatment restored the behavioral alterations and partially normalized the cortical biochemical changes. In conclusion, our data show some of the brain modifications probably responsible for the behavioral phenotype associated with TBI and suggest the CBD as a pharmacological tool to improve neurological dysfunctions caused by the trauma.
Collapse
Affiliation(s)
- Carmela Belardo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Flavia Ricciardi
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Salvatore Paino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
41
|
Xu X, Jiang S, Xu E, Wu X, Zhao R. Inhibition of CB1 receptor ameliorates spatial learning and memory impairment in mice with traumatic brain injury. Neurosci Lett 2018; 696:127-131. [PMID: 30576711 DOI: 10.1016/j.neulet.2018.12.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/11/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) is an increasingly prevalent condition affecting people of all ages and genders. The impairment of spatial learning and memory is one of the most common effects of TBI. Unfortunately, it currently lacks effective therapeutic interventions. The endocannabinoid (EC) system regulates a diverse array of physiological processes. Here, we found a 6.7-fold increase of 2-AG levels at 1 d post-TBI, declining thereafter. After 5 d, the levels were still 3.3-fold higher than in the controls. AM281, a CB1 receptor antagonist, reversed the TBI-reduced NMDA receptor subunits NR2B in the hippocampus and ameliorate the spatial learning and memory impairment at 7 d post-TBI, suggesting CB1 receptor is involved in the TBI-induced hippocampal-dependent spatial learning and memory impairment.
Collapse
Affiliation(s)
- Xiaoming Xu
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China.
| | - Shukun Jiang
- Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Enyu Xu
- Department of Forensic Toxicological Analysis, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Xu Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Rui Zhao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, PR China
| |
Collapse
|
42
|
Pharmacological inhibition of 2-arachidonoilglycerol hydrolysis enhances memory consolidation in rats through CB2 receptor activation and mTOR signaling modulation. Neuropharmacology 2018; 138:210-218. [DOI: 10.1016/j.neuropharm.2018.05.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/09/2018] [Accepted: 05/24/2018] [Indexed: 11/24/2022]
|
43
|
Hulsebus HJ, Curtis BJ, Molina PE, Afshar M, Boule LA, Morris N, Keshavarzian A, Kolls JK, Yeligar SM, Price ME, Wyatt TA, Choudhry MA, Kovacs EJ. Summary of the 2017 Alcohol and Immunology Research Interest Group (AIRIG) meeting. Alcohol 2018; 69:51-56. [PMID: 29654985 PMCID: PMC5930121 DOI: 10.1016/j.alcohol.2017.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 02/08/2023]
Abstract
On June 24, 2017, the 22nd annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held as a satellite conference during the annual Research Society on Alcoholism (RSA) Scientific Meeting in Denver, Colorado. The 2017 meeting focused broadly on mechanisms that link alcohol to tissue injury and inflammation, and how this research can be translated to improve human health. Two plenary sessions composed the meeting, which first explored the association between alcohol and trauma/tissue injury, and finished with a discussion of alcohol and mucosal inflammation. The presentations encompassed diverse areas of alcohol research, from effects on the brain, to airway and pulmonary systems, to gut barrier disruption. The discussions also thoughtfully highlighted how current laboratory and clinical research can be used to prevent or treat alcohol-related morbidity and mortality.
Collapse
Affiliation(s)
- Holly J. Hulsebus
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Immunology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Brenda J. Curtis
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Patricia E. Molina
- Department of Physiology and Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Majid Afshar
- Division of Pulmonary and Critical Care Medicine, Loyola University Chicago, Maywood, IL, USA,Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Lisbeth A. Boule
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Niya Morris
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Ali Keshavarzian
- Department of Gastroenterology, Rush University Medical Center, Chicago, IL, USA
| | - Jay K. Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University, New Orleans, LA, USA
| | - Samantha M. Yeligar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep, Emory University and Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Michael E. Price
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Todd A. Wyatt
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mashkoor A. Choudhry
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Elizabeth J. Kovacs
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Immunology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA,Corresponding author: Elizabeth J. Kovacs, Ph.D., 12700 East 19th Ave, Research Complex 2, Mailstop #8620, Aurora, CO, 80045. Phone: 303-724-8243.
| |
Collapse
|
44
|
Karoly HC, Bidwell LC, Mueller RL, Hutchison KE. Investigating the Relationships Between Alcohol Consumption, Cannabis Use, and Circulating Cytokines: A Preliminary Analysis. Alcohol Clin Exp Res 2018; 42:531-539. [PMID: 29286537 DOI: 10.1111/acer.13592] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND In recent years, human and animal studies have converged to support altered inflammatory signaling as a molecular mechanism underlying the pathophysiology of alcohol use disorders (AUDs). Alcohol binds to receptors on immune cells, triggering signaling pathways that produce pro-inflammatory cytokines. Chronic inflammation is associated with tissue damage, which may contribute to negative effects of AUD. Conversely, cannabis is associated with decreased inflammatory signaling, and animal studies suggest that cannabinoids may impact alcohol-induced inflammation. Thus, the impact of cannabis on inflammation in AUDs in humans warrants examination. METHODS We explored the relationship between self-reported alcohol and cannabis use and circulating levels of the pro-inflammatory cytokines interleukin 6 (IL-6), IL-8, and IL-1β in the blood. Among 66 regular drinkers (mean age = 30.08), we examined circulating cytokines and administered questionnaires assessing alcohol consumption and days of cannabis use over the past 90 days. We examined whether alcohol consumption, cannabis use, and gender were associated with changes in circulating cytokines, and whether there was a significant interaction between alcohol and cannabis use predicting blood levels of circulating cytokines. RESULTS A positive association between alcohol and IL-6 emerged. We also observed a negative association between cannabis and IL-1β. Follow-up moderation analyses indicated a cannabis by alcohol interaction predicting circulating IL-6, such that cannabis nonusers showed a stronger relationship between alcohol and IL-6 compared to cannabis users. CONCLUSIONS These preliminary findings suggest that cannabinoid compounds may serve to mitigate inflammation associated with alcohol use. In addition, the present results provide data to inform future investigations, with the goal of ultimately leveraging knowledge of the role of inflammation in AUDs to develop more effective treatments focused on novel immune targets.
Collapse
Affiliation(s)
- Hollis C Karoly
- Department of Psychology & Neuroscience , University of Colorado Boulder, Boulder, Colorado
| | - L Cinnamon Bidwell
- Institute of Cognitive Science , University of Colorado Boulder, Boulder, Colorado
| | - Raeghan L Mueller
- Department of Psychology & Neuroscience , University of Colorado Boulder, Boulder, Colorado
| | - Kent E Hutchison
- Department of Psychology & Neuroscience , University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
45
|
Therapeutic Potentials of Synapses after Traumatic Brain Injury: A Comprehensive Review. Neural Plast 2017; 2017:4296075. [PMID: 28491479 PMCID: PMC5405590 DOI: 10.1155/2017/4296075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/09/2017] [Accepted: 03/14/2017] [Indexed: 12/26/2022] Open
Abstract
Massive studies have focused on the understanding of the pathobiology of cellular and molecular changes and injury mechanisms after traumatic brain injury (TBI), but very few studies have specially discussed the role of synapses in the context of TBI. This paper specifically highlights the role and therapeutic potentials of synapses after TBI. First, we review and conclude how synapses interact with constant structural, metabolic, neuroendocrine, and inflammatory mechanisms after TBI. Second, we briefly describe several key synaptic proteins involved in neuroplasticity, which may be novel neuronal targets for specific intervention. Third, we address therapeutic interventions in association with synapses after TBI. Finally, we concisely discuss the study gaps in the synapses after TBI, in hopes that this would provide more insights for future studies. Synapses play an important role in TBI; while the understandings on the synaptic participation in the treatments and prognosis of TBI are lacking, more studies in this area are warranted.
Collapse
|
46
|
Guida F, Boccella S, Iannotta M, De Gregorio D, Giordano C, Belardo C, Romano R, Palazzo E, Scafuro MA, Serra N, de Novellis V, Rossi F, Maione S, Luongo L. Palmitoylethanolamide Reduces Neuropsychiatric Behaviors by Restoring Cortical Electrophysiological Activity in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2017; 8:95. [PMID: 28321191 PMCID: PMC5337754 DOI: 10.3389/fphar.2017.00095] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/14/2017] [Indexed: 11/25/2022] Open
Abstract
Traumatic brain injury (TBI) represents a major public health problem, which is associated with neurological dysfunction. In severe or moderate cases of TBI, in addition to its high mortality rate, subjects may encounter diverse behavioral dysfunctions. Previous reports suggest that an association between TBI and chronic pain syndromes tends to be more common in patients with mild forms of brain injury. Despite causing minimal brain damage, mild TBI (mTBI) often leads to persistent psychologically debilitating symptoms, which can include anxiety, various forms of memory and learning deficits, and depression. At present, no effective treatment options are available for these symptoms, and little is known about the complex cellular activity affecting neuronal activity that occurs in response to TBI during its late phase. Here, we used a mouse model to investigate the effect of Palmitoylethanolamide (PEA) on both the sensorial and neuropsychiatric dysfunctions associated with mTBI through behavioral, electrophysiological, and biomolecular approaches. Fourteen-day mTBI mice developed anxious, aggressive, and reckless behavior, whilst depressive-like behavior and impaired social interactions were observed from the 60th day onward. Altered behavior was associated with changes in interleukin 1 beta (IL-1β) expression levels and neuronal firing activity in the medial prefrontal cortex. Compared with vehicle, PEA restored the behavioral phenotype and partially normalized the biochemical and functional changes occurring at the supraspinal level. In conclusion, our findings reveal some of the supraspinal modifications responsible for the behavioral alterations associated with mTBI and suggest PEA as a pharmacological tool to ameliorate neurological dysfunction induced by the trauma.
Collapse
Affiliation(s)
- Francesca Guida
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN)Naples, Italy; Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle RicerchePozzuoli, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Danilo De Gregorio
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Catia Giordano
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Rosaria Romano
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Maria A Scafuro
- Department of Anesthesiology, Surgery and Emergency, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Nicola Serra
- Department of Radiology, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Vito de Novellis
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN)Naples, Italy; Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle RicerchePozzuoli, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN)Naples, Italy; Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle RicerchePozzuoli, Italy; Young Against Pain (YAP) Italian Group, NaplesItaly
| |
Collapse
|
47
|
Schurman LD, Lichtman AH. Endocannabinoids: A Promising Impact for Traumatic Brain Injury. Front Pharmacol 2017; 8:69. [PMID: 28261100 PMCID: PMC5314139 DOI: 10.3389/fphar.2017.00069] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/02/2017] [Indexed: 02/01/2023] Open
Abstract
The endogenous cannabinoid (endocannabinoid) system regulates a diverse array of physiological processes and unsurprisingly possesses considerable potential targets for the potential treatment of numerous disease states, including two receptors (i.e., CB1 and CB2 receptors) and enzymes regulating their endogenous ligands N-arachidonoylethanolamine (anandamide) and 2-arachidonyl glycerol (2-AG). Increases in brain levels of endocannabinoids to pathogenic events suggest this system plays a role in compensatory repair mechanisms. Traumatic brain injury (TBI) pathology remains mostly refractory to currently available drugs, perhaps due to its heterogeneous nature in etiology, clinical presentation, and severity. Here, we review pre-clinical studies assessing the therapeutic potential of cannabinoids and manipulations of the endocannabinoid system to ameliorate TBI pathology. Specifically, manipulations of endocannabinoid degradative enzymes (e.g., fatty acid amide hydrolase, monoacylglycerol lipase, and α/β-hydrolase domain-6), CB1 and CB2 receptors, and their endogenous ligands have shown promise in modulating cellular and molecular hallmarks of TBI pathology such as; cell death, excitotoxicity, neuroinflammation, cerebrovascular breakdown, and cell structure and remodeling. TBI-induced behavioral deficits, such as learning and memory, neurological motor impairments, post-traumatic convulsions or seizures, and anxiety also respond to manipulations of the endocannabinoid system. As such, the endocannabinoid system possesses potential drugable receptor and enzyme targets for the treatment of diverse TBI pathology. Yet, full characterization of TBI-induced changes in endocannabinoid ligands, enzymes, and receptor populations will be important to understand that role this system plays in TBI pathology. Promising classes of compounds, such as the plant-derived phytocannabinoids, synthetic cannabinoids, and endocannabinoids, as well as their non-cannabinoid receptor targets, such as TRPV1 receptors, represent important areas of basic research and potential therapeutic interest to treat TBI.
Collapse
Affiliation(s)
| | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, RichmondVA, USA
| |
Collapse
|
48
|
Abou-El-Hassan H, Sukhon F, Assaf EJ, Bahmad H, Abou-Abbass H, Jourdi H, Kobeissy FH. Degradomics in Neurotrauma: Profiling Traumatic Brain Injury. Methods Mol Biol 2017; 1598:65-99. [PMID: 28508358 DOI: 10.1007/978-1-4939-6952-4_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Degradomics has recently emerged as a subdiscipline in the omics era with a focus on characterizing signature breakdown products implicated in various disease processes. Driven by promising experimental findings in cancer, neuroscience, and metabolomic disorders, degradomics has significantly promoted the notion of disease-specific "degradome." A degradome arises from the activation of several proteases that target specific substrates and generate signature protein fragments. Several proteases such as calpains, caspases, cathepsins, and matrix metalloproteinases (MMPs) are involved in the pathogenesis of numerous diseases that disturb the physiologic balance between protein synthesis and protein degradation. While regulated proteolytic activities are needed for development, growth, and regeneration, uncontrolled proteolysis initiated under pathological conditions ultimately culminates into apoptotic and necrotic processes. In this chapter, we aim to review the protease-substrate repertoires in neural injury concentrating on traumatic brain injury. A striking diversity of protease substrates, essential for neuronal and brain structural and functional integrity, namely, encryptic biomarker neoproteins, have been characterized in brain injury. These include cytoskeletal proteins, transcription factors, cell cycle regulatory proteins, synaptic proteins, and cell junction proteins. As these substrates are subject to proteolytic fragmentation, they are ceaselessly exposed to activated proteases. Characterization of these molecules allows for a surge of "possible" therapeutic approaches of intervention at various levels of the proteolytic cascade.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Fares Sukhon
- Faculty of Medicine, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Edwyn Jeremy Assaf
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hisham Bahmad
- Faculty of Medical, Neuroscience Research Center, Beirut Arab University, Beirut, Lebanon
- Faculty of Medicine, Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Hussein Abou-Abbass
- Faculty of Medical Sciences, Neuroscience Research Center, Lebanese University, Beirut, Lebanon
- Faculty of Medicine, Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Hussam Jourdi
- Faculty of Science¸ Department of Biology, University of Balamand, Souk-el-Gharb Campus, Aley, Lebanon
| | - Firas H Kobeissy
- Faculty of Medicine, Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.
- Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research, University of Florida, Gainesville, FL, USA.
| |
Collapse
|