1
|
Wiseman RL, Bigos KL, Dastgheyb RM, Barker PB, Rubin LH, Slusher BS. Brain N -acetyl-aspartyl-glutamate is associated with cognitive function in older virally suppressed people with HIV. AIDS 2024; 38:1003-1011. [PMID: 38411600 PMCID: PMC11062820 DOI: 10.1097/qad.0000000000003871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
OBJECTIVES Cognitive impairment persists in virally suppressed people with HIV (VS-PWH) especially in higher order domains. One cortical circuit, linked to these domains, is regulated by N -acetyl-aspartyl glutamate (NAAG), the endogenous agonist of the metabotropic glutamate receptor 3. The enzyme glutamate carboxypeptidase II (GCPII) catabolizes NAAG and is upregulated in aging and disease. Inhibition of GCPII increases brain NAAG and improves learning and memory in rodent and primate models. DESIGN As higher order cognitive impairment is present in VS-PWH, and NAAG has not been investigated in earlier magnetic resonance spectroscopy studies (MRS), we investigated if brain NAAG levels measured by MRS were associated with cognitive function. METHODS We conducted a retrospective analysis of 7-Tesla MRS data from a previously published study on cognition in older VS-PWH. The original study did not separately quantify NAAG, therefore, work for this report focused on relationships between regional NAAG levels in frontal white matter (FWM), left hippocampus, left basal ganglia and domain-specific cognitive performance in 40 VS-PWH after adjusting for confounds. Participants were older than 50 years, negative for affective and neurologic disorders, and had no prior 3-month psychoactive-substance use. RESULTS Higher NAAG levels in FWM were associated with better attention/working memory. Higher left basal ganglia NAAG related to better verbal fluency. There was a positive relationship between hippocampal NAAG and executive function which lost significance after correction for confounds. CONCLUSION These data suggest brain NAAG serves as a biomarker of cognition in VS-PWH. Pharmacological modulation of brain NAAG warrants investigation as a therapeutic approach for cognitive deficits in VS-PWH.
Collapse
Affiliation(s)
- Robyn L. Wiseman
- Department of Pharmacology and Molecular Sciences
- Johns Hopkins Drug Discovery
- Department of Medicine, Division of Clinical Pharmacology
| | - Kristin L. Bigos
- Department of Pharmacology and Molecular Sciences
- Department of Medicine, Division of Clinical Pharmacology
- Department of Psychiatry and Behavioral Sciences
| | | | - Peter B. Barker
- Russell H. Morgan Department of Radiology and Radiological Sciences
| | - Leah H. Rubin
- Department of Psychiatry and Behavioral Sciences
- Department of Neurology
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
| | - Barbara S. Slusher
- Department of Pharmacology and Molecular Sciences
- Johns Hopkins Drug Discovery
- Department of Medicine, Division of Clinical Pharmacology
- Department of Psychiatry and Behavioral Sciences
- Department of Neurology
- Department of Oncology
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Duan A, Qiu Y, Song B, Tao Y, Wang M, Yin Z, Xie M, Chen Z, Wang Z, Sun X. Metabolome-Wide Mendelian Randomization Assessing the Causal Role of Serum and Cerebrospinal Metabolites in Traumatic Brain Injury. Biomedicines 2024; 12:1178. [PMID: 38927385 PMCID: PMC11201266 DOI: 10.3390/biomedicines12061178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Previous studies have identified metabolites as biomarkers or potential therapeutic targets for traumatic brain injury (TBI). However, the causal association between them remains unknown. Therefore, we investigated the causal effect of serum metabolites and cerebrospinal fluid (CSF) metabolites on TBI susceptibility through Mendelian randomization (MR). Genetic variants related to metabolites and TBI were extracted from a corresponding genome-wide association study (GWAS). Causal effects were estimated through the inverse variance weighted approach, supplemented by a weighted median, weight mode, and the MR-Egger test. In addition, sensitivity analyses were further performed to evaluate the stability of the MR results, including the MR-Egger intercept, leave-one-out analysis, Cochrane's Q-test, and the MR-PRESSO global test. Metabolic pathway analysis was applied to uncover the underlying pathways of the significant metabolites in TBI. In blood metabolites, substances such as 4-acetaminophen sulfate and kynurenine showed positive links, whereas beta-hydroxyisovalerate and creatinine exhibited negative correlations. CSF metabolites such as N-formylanthranilic acid were positively related, while kynurenate showed negative associations. The metabolic pathway analysis highlighted the potential biological pathways involved in TBI. Of these 16 serum metabolites, 11 CSF metabolites and metabolic pathways may serve as useful circulating biomarkers in clinical screening and prevention, and may be candidate molecules for the exploration of mechanisms and drug targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (A.D.); (Y.Q.)
| | - Xiaoou Sun
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (A.D.); (Y.Q.)
| |
Collapse
|
3
|
Witkin JM, Shafique H, Cerne R, Smith JL, Marini AM, Lipsky RH, Delery E. Mechanistic and therapeutic relationships of traumatic brain injury and γ-amino-butyric acid (GABA). Pharmacol Ther 2024; 256:108609. [PMID: 38369062 DOI: 10.1016/j.pharmthera.2024.108609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Traumatic brain injury (TBI) is a highly prevalent medical condition for which no medications specific for the prophylaxis or treatment of the condition as a whole exist. The spectrum of symptoms includes coma, headache, seizures, cognitive impairment, depression, and anxiety. Although it has been known for years that the inhibitory neurotransmitter γ-amino-butyric acid (GABA) is involved in TBI, no novel therapeutics based upon this mechanism have been introduced into clinical practice. We review the neuroanatomical, neurophysiological, neurochemical, and neuropharmacological relationships of GABA neurotransmission to TBI with a view toward new potential GABA-based medicines. The long-standing idea that excitatory and inhibitory (GABA and others) balances are disrupted by TBI is supported by the experimental data but has failed to invent novel methods of restoring this balance. The slow progress in advancing new treatments is due to the complexity of the disorder that encompasses multiple dynamically interacting biological processes including hemodynamic and metabolic systems, neurodegeneration and neurogenesis, major disruptions in neural networks and axons, frank brain lesions, and a multitude of symptoms that have differential neuronal and neurohormonal regulatory mechanisms. Although the current and ongoing clinical studies include GABAergic drugs, no novel GABA compounds are being explored. It is suggested that filling the gap in understanding the roles played by specific GABAA receptor configurations within specific neuronal circuits could help define new therapeutic approaches. Further research into the temporal and spatial delivery of GABA modulators should also be useful. Along with GABA modulation, research into the sequencing of GABA and non-GABA treatments will be needed.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | | | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA; Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA
| | - Ann M Marini
- Department of Neurology, Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert H Lipsky
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Elizabeth Delery
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.
| |
Collapse
|
4
|
De Gasperi R, Gama Sosa MA, Perez Garcia G, Perez GM, Pryor D, Morrison CLA, Lind R, Abutarboush R, Kawoos U, Statz JK, Patterson J, Hof PR, Zhu CW, Ahlers ST, Cook DG, Elder GA. Metabotropic Glutamate Receptor 2 Expression Is Chronically Elevated in Male Rats With Post-Traumatic Stress Disorder Related Behavioral Traits Following Repetitive Low-Level Blast Exposure. J Neurotrauma 2024; 41:714-733. [PMID: 37917117 PMCID: PMC10902502 DOI: 10.1089/neu.2023.0252] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
Many military veterans who experienced blast-related traumatic brain injuries in the conflicts in Iraq and Afghanistan currently suffer from chronic cognitive and mental health problems that include depression and post-traumatic stress disorder (PTSD). Male rats exposed to repetitive low-level blast develop cognitive and PTSD-related behavioral traits that are present for more than 1 year after exposure. We previously reported that a group II metabotropic receptor (mGluR2/3) antagonist reversed blast-induced behavioral traits. In this report, we explored mGluR2/3 expression following blast exposure in male rats. Western blotting revealed that mGluR2 protein (but not mGluR3) was increased in all brain regions studied (anterior cortex, hippocampus, and amygdala) at 43 or 52 weeks after blast exposure but not at 2 weeks or 6 weeks. mGluR2 RNA was elevated at 52 weeks while mGluR3 was not. Immunohistochemical staining revealed no changes in the principally presynaptic localization of mGluR2 by blast exposure. Administering the mGluR2/3 antagonist LY341495 after behavioral traits had emerged rapidly reversed blast-induced effects on novel object recognition and cued fear responses 10 months following blast exposure. These studies support alterations in mGluR2 receptors as a key pathophysiological event following blast exposure and provide further support for group II metabotropic receptors as therapeutic targets in the neurobehavioral effects that follow blast injury.
Collapse
Affiliation(s)
- Rita De Gasperi
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Georgina Perez Garcia
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Dylan Pryor
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Chenel L-A. Morrison
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Northeast Regional Alliance Health Careers Opportunity Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rachel Lind
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Jonathan K. Statz
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Jacob Patterson
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carolyn W. Zhu
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, Pharmacology, and Psychiatry, University of Washington, Seattle, Washington, USA
- Department of Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Gregory A. Elder
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
5
|
Ji T, Pang Y, Cheng M, Wang R, Chen X, Zhang C, Liu M, Zhang J, Zhong C. Deletion of glutamate carboxypeptidase II (GCPII), but not GCPIII, provided long-term benefits in mice with traumatic brain injury. CNS Neurosci Ther 2023; 29:3786-3801. [PMID: 37349952 PMCID: PMC10651966 DOI: 10.1111/cns.14299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/28/2023] [Indexed: 06/24/2023] Open
Abstract
MAIN PROBLEM N-acetylaspartylglutamate (NAAG) has neuroprotective effects in traumatic brain injury (TBI) by activating metabotropic glutamate receptor 3 (mGluR3) and reducing glutamate release. Glutamate carboxypeptidase II (GCPII) is the primary enzyme responsible for the hydrolysis of NAAG. It remains unclear whether glutamate carboxypeptidase III (GCPIII), a homolog of GCPII, can partially compensate for GCPII's function. METHODS GCPII-/- , GCPIII-/- , and GCPII/III-/- mice were generated using CRISPR/Cas9 technology. Mice brain injury model was established through moderate controlled cortical impact (CCI). The relationship between GCPII and GCPIII was explored by analyzing injury response signals in the hippocampus and cortex of mice with different genotypes at the acute (1 day) and subacute (7 day) phase after TBI. RESULTS In this study, we found that deletion of GCPII reduced glutamate production, excitotoxicity, and neuronal damage and improved cognitive function, but GCPIII deletion had no significant neuroprotective effect. Additionally, there was no significant difference in the neuroprotective effect between the combination of GCPII and GCPIII deletion and GCPII deletion alone. CONCLUSION These results suggest that GCPII inhibition may be a therapeutic option for TBI, and that GCPIII may not act as a complementary enzyme to GCPII in this context.
Collapse
Affiliation(s)
- Tongjie Ji
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Ying Pang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Meng Cheng
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Rui Wang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Xu Chen
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Chunyu Zhang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Min Liu
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Jing Zhang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
- Institute for Advanced StudyTongji UniversityShanghaiChina
| | - Chunlong Zhong
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| |
Collapse
|
6
|
Arnsten AFT, Ishizawa Y, Xie Z. Scientific rationale for the use of α2A-adrenoceptor agonists in treating neuroinflammatory cognitive disorders. Mol Psychiatry 2023; 28:4540-4552. [PMID: 37029295 PMCID: PMC10080530 DOI: 10.1038/s41380-023-02057-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
Neuroinflammatory disorders preferentially impair the higher cognitive and executive functions of the prefrontal cortex (PFC). This includes such challenging disorders as delirium, perioperative neurocognitive disorder, and the sustained cognitive deficits from "long-COVID" or traumatic brain injury. There are no FDA-approved treatments for these symptoms; thus, understanding their etiology is important for generating therapeutic strategies. The current review describes the molecular rationale for why PFC circuits are especially vulnerable to inflammation, and how α2A-adrenoceptor (α2A-AR) actions throughout the nervous and immune systems can benefit the circuits in PFC needed for higher cognition. The layer III circuits in the dorsolateral PFC (dlPFC) that generate and sustain the mental representations needed for higher cognition have unusual neurotransmission and neuromodulation. They are wholly dependent on NMDAR neurotransmission, with little AMPAR contribution, and thus are especially vulnerable to kynurenic acid inflammatory signaling which blocks NMDAR. Layer III dlPFC spines also have unusual neuromodulation, with cAMP magnification of calcium signaling in spines, which opens nearby potassium channels to rapidly weaken connectivity and reduce neuronal firing. This process must be tightly regulated, e.g. by mGluR3 or α2A-AR on spines, to prevent loss of firing. However, the production of GCPII inflammatory signaling reduces mGluR3 actions and markedly diminishes dlPFC network firing. Both basic and clinical studies show that α2A-AR agonists such as guanfacine can restore dlPFC network firing and cognitive function, through direct actions in the dlPFC, but also by reducing the activity of stress-related circuits, e.g. in the locus coeruleus and amygdala, and by having anti-inflammatory actions in the immune system. This information is particularly timely, as guanfacine is currently the focus of large clinical trials for the treatment of delirium, and in open label studies for the treatment of cognitive deficits from long-COVID.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department Neuroscience, Yale University School of Medicine, New Haven, CT, 056510, USA.
| | - Yumiko Ishizawa
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhongcong Xie
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
7
|
Ji T, Pang Y, Cheng M, Wang R, Chen X, Zhang C, Liu M, Zhang J, Zhong C. mNSCs overexpressing Rimkla transplantation facilitates cognitive recovery in a mouse model of traumatic brain injury. iScience 2023; 26:107913. [PMID: 37810220 PMCID: PMC10550729 DOI: 10.1016/j.isci.2023.107913] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/22/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
N-acetyl aspartyl-glutamate (NAAG) is easily inactivated for the hydrolysis of NAAG peptidase on the surface of glial cells, thereby losing its endogenous neuroprotective effect after traumatic brain injury. In this study, lentiviral vectors were used to over express/knock out NAAG synthetase II (Rimkla) in mouse embryonic neural stem cells (mNSCs) in vitro and these mNSCs were transplanted at the lesion site in a mouse model of controlled cortical impact (CCI). In vivo experiments showed that transplantation of mNSCs overexpressing Rimkla regulated glutamate-glutamine cycling between adjacent astrocytes and neurons in the subacute phase of CCI, thereby enhancing support for neuronal metabolism and promoting neuronal synaptic repair in the hippocampal CA3 region. Taken together, these findings demonstrate that transplantation of neural stem cells overexpressing Rimkla can effectively increase the NAAG concentration in local brain regions, which opens up new ideas for the maintenance of NAAG neuroprotective effects after TBI.
Collapse
Affiliation(s)
- Tongjie Ji
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Pang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Meng Cheng
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rui Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xu Chen
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyu Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Min Liu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Advanced Study, Tongji University, Shanghai, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
8
|
Gao Y, Liu N, Chen J, Zheng P, Niu J, Tang S, Peng X, Wu J, Yu J, Ma L. Neuropharmacological insight into preventive intervention in posttraumatic epilepsy based on regulating glutamate homeostasis. CNS Neurosci Ther 2023; 29:2430-2444. [PMID: 37309302 PMCID: PMC10401093 DOI: 10.1111/cns.14294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 05/15/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Posttraumatic epilepsy (PTE) is one of the most critical complications of traumatic brain injury (TBI), significantly increasing TBI patients' neuropsychiatric symptoms and mortality. The abnormal accumulation of glutamate caused by TBI and its secondary excitotoxicity are essential reasons for neural network reorganization and functional neural plasticity changes, contributing to the occurrence and development of PTE. Restoring glutamate balance in the early stage of TBI is expected to play a neuroprotective role and reduce the risk of PTE. AIMS To provide a neuropharmacological insight for drug development to prevent PTE based on regulating glutamate homeostasis. METHODS We discussed how TBI affects glutamate homeostasis and its relationship with PTE. Furthermore, we also summarized the research progress of molecular pathways for regulating glutamate homeostasis after TBI and pharmacological studies aim to prevent PTE by restoring glutamate balance. RESULTS TBI can lead to the accumulation of glutamate in the brain, which increases the risk of PTE. Targeting the molecular pathways affecting glutamate homeostasis helps restore normal glutamate levels and is neuroprotective. DISCUSSION Taking glutamate homeostasis regulation as a means for new drug development can avoid the side effects caused by direct inhibition of glutamate receptors, expecting to alleviate the diseases related to abnormal glutamate levels in the brain, such as PTE, Parkinson's disease, depression, and cognitive impairment. CONCLUSION It is a promising strategy to regulate glutamate homeostasis through pharmacological methods after TBI, thereby decreasing nerve injury and preventing PTE.
Collapse
Affiliation(s)
- Yuan Gao
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
- Hunan Province Key Laboratory for Antibody‐Based Drug and Intelligent Delivery System, School of Pharmaceutical SciencesHunan University of MedicineHuaihuaChina
| | - Ning Liu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Juan Chen
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Ping Zheng
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jianguo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous RegionNingxia Medical UniversityYinchuanChina
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody‐Based Drug and Intelligent Delivery System, School of Pharmaceutical SciencesHunan University of MedicineHuaihuaChina
| | - Xiaodong Peng
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jing Wu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jianqiang Yu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Lin Ma
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous RegionNingxia Medical UniversityYinchuanChina
| |
Collapse
|
9
|
Molecular Landscape of Tourette's Disorder. Int J Mol Sci 2023; 24:ijms24021428. [PMID: 36674940 PMCID: PMC9865021 DOI: 10.3390/ijms24021428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/12/2023] Open
Abstract
Tourette's disorder (TD) is a highly heritable childhood-onset neurodevelopmental disorder and is caused by a complex interplay of multiple genetic and environmental factors. Yet, the molecular mechanisms underlying the disorder remain largely elusive. In this study, we used the available omics data to compile a list of TD candidate genes, and we subsequently conducted tissue/cell type specificity and functional enrichment analyses of this list. Using genomic data, we also investigated genetic sharing between TD and blood and cerebrospinal fluid (CSF) metabolite levels. Lastly, we built a molecular landscape of TD through integrating the results from these analyses with an extensive literature search to identify the interactions between the TD candidate genes/proteins and metabolites. We found evidence for an enriched expression of the TD candidate genes in four brain regions and the pituitary. The functional enrichment analyses implicated two pathways ('cAMP-mediated signaling' and 'Endocannabinoid Neuronal Synapse Pathway') and multiple biological functions related to brain development and synaptic transmission in TD etiology. Furthermore, we found genetic sharing between TD and the blood and CSF levels of 39 metabolites. The landscape of TD not only provides insights into the (altered) molecular processes that underlie the disease but, through the identification of potential drug targets (such as FLT3, NAALAD2, CX3CL1-CX3CR1, OPRM1, and HRH2), it also yields clues for developing novel TD treatments.
Collapse
|
10
|
Tang Y, Sun L, Zhao Y, Yao J, Feng Z, Liu Z, Zhang G, Sun C. UHPLC-ESI-QE-Orbitrap-MS based metabolomics reveals the antioxidant mechanism of icaritin on mice with cerebral ischemic reperfusion. PeerJ 2023; 11:e14483. [PMID: 36643627 PMCID: PMC9838208 DOI: 10.7717/peerj.14483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/08/2022] [Indexed: 01/12/2023] Open
Abstract
Background Icaritin (ICT) has been previously demonstrated to display protective effects against cerebral ischemic reperfusion (I/R) by inhibiting oxidative stress, but the mechanism remains unclear. This study aimed to explore the mechanism from the perspective of metabolomics. Methods A mice cerebral artery occlusion/reperfusion (MCAO/R) model was explored to mimic cerebral ischemic reperfusion and protective effect of ICT was assessed by neurologic deficit scoring, infarct volume and brain water content. Ultra-high-performance liquid chromatography electrospray ionization orbitrap tandem mass spectrometry (UHPLC-ESI-QE-Orbitrap-MS) based metabolomic was performed to explore potential biomarkers. Brain tissue metabolic profiles were analyzed and metabolic biomarkers were identified through multivariate data analysis. The protein levels of Nrf2, HO-1 and HQO1 were assayed by western blot. The release of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT) were detected using corresponding assay kits. Results The results showed that after ICT treatment, the neurological deficit, cerebral infarction area, brain edema and the level of MDA in brain tissue of MCAO/R mice were significantly reduced. Meanwhile, ICT enhanced the activity of SOD, CAT and GSH-Px. Western blot results confirmed that ICT up-regulated the protein levels of antioxidant-related protein including Nrf2, HO-1 and NQO1. According to the metabolomic profiling of brain tissues, clear separations were observed among the Sham, Model and ICT groups. A total of 44 biomarkers were identified, and the identified biomarkers were mainly related to linoleic acid metabolism, arachidonic acid metabolism, alanine, aspartate and glutamate metabolism, arginine biosynthesis, arginine and proline metabolism, D-glutamine and D-glutamate metabolism, taurine and hypotaurine metabolism and purine metabolism, respectively. At the same time, the inhibitory effect of ICT on arachidonic acid and linoleic acid in brain tissue, as well as the promoting effect on taurine, GABA, NAAG, may be the key factors for the anti-neurooxidative function of mice after MCAO/R injury. Conclusion Our results demonstrate that ICT has benefits for MCAO/R injury, which are partially related to the suppression of oxidative stress via stimulating the Nrf2 signaling and regulating the production of arachidonic acid, linoleic acid, taurine, GABA, NAAG in brain tissue.
Collapse
Affiliation(s)
- Yunfeng Tang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Lixin Sun
- Linyi Traditional Chinese Medicine Hospital, Linyi, Shandong Province, China
| | - Yun Zhao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Jingchun Yao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Zhong Feng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China,School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Zhong Liu
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Chenghong Sun
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| |
Collapse
|
11
|
Dogra S, Putnam J, Conn PJ. Metabotropic glutamate receptor 3 as a potential therapeutic target for psychiatric and neurological disorders. Pharmacol Biochem Behav 2022; 221:173493. [PMID: 36402243 PMCID: PMC9729465 DOI: 10.1016/j.pbb.2022.173493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
Abstract
Glutamate is a major excitatory neurotransmitter in the central nervous system (CNS) and abnormalities in the glutamatergic system underlie various CNS disorders. As metabotropic glutamate receptor 3 (mGlu3 receptor) regulates glutamatergic transmission in various brain areas, emerging literature suggests that targeting mGlu3 receptors can be a novel approach to the treatment of psychiatric and neurological disorders. For example, mGlu3 receptor negative allosteric modulators (NAMs) induce rapid antidepressant-like effects in both acute and chronic stress models. Activation of mGlu3 receptors can enhance cognition in the rodents modeling schizophrenia-like pathophysiology. The mGlu3 receptors expressed in the astrocytes induce neuroprotective effects. Although polymorphisms in GRM3 have been shown to be associated with addiction, there is not significant evidence about the efficacy of mGlu3 receptor ligands in rodent models of addiction. Collectively, drugs targeting mGlu3 receptors may provide an alternative approach to fill the unmet clinical need for safer and more efficacious therapeutics for CNS disorders.
Collapse
Affiliation(s)
- Shalini Dogra
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Jason Putnam
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
12
|
Chen Z, Krishnamachary B, Mironchik Y, Ray Banerjee S, Pomper MG, Bhujwalla ZM. PSMA-specific degradable dextran for multiplexed immunotargeted siRNA therapeutics against prostate cancer. NANOSCALE 2022; 14:14014-14022. [PMID: 36093754 PMCID: PMC9844541 DOI: 10.1039/d2nr02200a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Small interfering RNA (siRNA) is ideal for gene silencing through a sequence-specific RNA interference process. The redundancy and complexity of molecular pathways in cancer create a need for multiplexed targeting that can be achieved with multiplexed siRNA delivery. Here, we delivered multiplexed siRNA with a PSMA-targeted biocompatible dextran nanocarrier to downregulate CD46 and PD-L1 in PSMA expressing prostate cancer cells. The selected gene targets, PD-L1 and CD46, play important roles in the escape of cancer cells from immune surveillance. PSMA, abundantly expressed by prostate cancer cells, allowed the prostate cancer-specific delivery of the nanocarrier. The nanocarrier was modified with acid cleavable acetal bonds for a rapid release of siRNA. Cell imaging and flow cytometry studies confirmed the PSMA-specific delivery of CD46 and PD-L1 siRNA to high PSMA expressing PC-3 PIP cells. Immunoblot, qRT-PCR and flow cytometry methods confirmed the downregulation of CD46 and PD-L1 following treatment with multiplexed siRNA.
Collapse
Affiliation(s)
- Zhihang Chen
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Balaji Krishnamachary
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Yelena Mironchik
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Sangeeta Ray Banerjee
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Zaver M Bhujwalla
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
13
|
Woo E, Datta D, Arnsten AFT. Glutamate Metabotropic Receptor Type 3 (mGlu3) Localization in the Rat Prelimbic Medial Prefrontal Cortex. Front Neuroanat 2022; 16:849937. [PMID: 35444520 PMCID: PMC9013768 DOI: 10.3389/fnana.2022.849937] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Metabotropic glutamate receptors type 3 (mGlu3, encoded by GRM3) are increasingly related to cognitive functioning, including the working memory operations of the prefrontal cortex (PFC). In rhesus monkeys, mGlu3 are most commonly expressed on glia (36%), but are also very prominent on layer III dendritic spines (23%) in the dorsolateral PFC (dlPFC) where they enhance working memory-related neuronal firing. In contrast, mGlu2 are predominately presynaptic in layer III of macaque dlPFC, indicating a pre- vs. post-synaptic dissociation by receptor subtype. The current study examined the cellular and subcellular localizations of mGlu3 in the rat prelimbic medial PFC (PL mPFC), a region needed for spatial working memory performance in rodents. Multiple label immunofluorescence demonstrated mGlu3 expression in neurons and astrocytes, with rare labeling in microglia. Immunoelectron microscopy of layers III and V found that the predominant location for mGlu3 was on axons (layer III: 35.9%; layer V: 44.1%), with labeling especially prominent within the intervaricose segments distant from axon terminals. mGlu3 were also found on glia (likely astrocytes), throughout the glial membrane (layer III: 28.2%; layer V: 29.5%). Importantly, mGlu3 could be seen on dendritic spines, especially in layer III (layer III: 15.6%; layer V: 8.2%), with minor labeling on dendrites. These data show that there are some similarities between mGlu3 expression in rat PL mPFC and macaque dlPFC, but the spine expression enriches and differentiates in the more recently evolved primate dlPFC.
Collapse
|
14
|
Morland C, Nordengen K. N-Acetyl-Aspartyl-Glutamate in Brain Health and Disease. Int J Mol Sci 2022; 23:ijms23031268. [PMID: 35163193 PMCID: PMC8836185 DOI: 10.3390/ijms23031268] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
N-acetyl-aspartyl-glutamate (NAAG) is the most abundant dipeptide in the brain, where it acts as a neuromodulator of glutamatergic synapses by activating presynaptic metabotropic glutamate receptor 3 (mGluR3). Recent data suggest that NAAG is selectively localized to postsynaptic dendrites in glutamatergic synapses and that it works as a retrograde neurotransmitter. NAAG is released in response to glutamate and provides the postsynaptic neuron with a feedback mechanisms to inhibit excessive glutamate signaling. A key regulator of synaptically available NAAG is rapid degradation by the extracellular enzyme glutamate carboxypeptidase II (GCPII). Increasing endogenous NAAG—for instance by inhibiting GCPII—is a promising treatment option for many brain disorders where glutamatergic excitotoxicity plays a role. The main effect of NAAG occurs through increased mGluR3 activation and thereby reduced glutamate release. In the present review, we summarize the transmitter role of NAAG and discuss the involvement of NAAG in normal brain physiology. We further present the suggested roles of NAAG in various neurological and psychiatric diseases and discuss the therapeutic potential of strategies aiming to enhance NAAG levels.
Collapse
Affiliation(s)
- Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, 1068 Oslo, Norway
- Correspondence: (C.M.); (K.N.); Tel.: +47-22844937; (C.M.); +47-23073580 (K.N.)
| | - Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
- Correspondence: (C.M.); (K.N.); Tel.: +47-22844937; (C.M.); +47-23073580 (K.N.)
| |
Collapse
|
15
|
Hollinger KR, Sharma A, Tallon C, Lovell L, Thomas AG, Zhu X, Wiseman R, Wu Y, Kambhampati SP, Liaw K, Sharma R, Rojas C, Rais R, Kannan S, Kannan RM, Slusher BS. Dendrimer-2PMPA selectively blocks upregulated microglial GCPII activity and improves cognition in a mouse model of multiple sclerosis. Nanotheranostics 2022; 6:126-142. [PMID: 34976589 PMCID: PMC8671953 DOI: 10.7150/ntno.63158] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/19/2022] Open
Abstract
Cognitive impairment is a common aspect of multiple sclerosis (MS) for which there are no treatments. Reduced brain N-acetylaspartylglutamate (NAAG) levels are linked to impaired cognition in various neurological diseases, including MS. NAAG levels are regulated by glutamate carboxypeptidase II (GCPII), which hydrolyzes the neuropeptide to N-acetyl-aspartate and glutamate. GCPII activity is upregulated multifold in microglia following neuroinflammation. Although several GCPII inhibitors, such as 2-PMPA, elevate brain NAAG levels and restore cognitive function in preclinical studies when given at high systemic doses or via direct brain injection, none are clinically available due to poor bioavailability and limited brain penetration. Hydroxyl-dendrimers have been successfully used to selectively deliver drugs to activated glia. Methods: We attached 2-PMPA to hydroxyl polyamidoamine (PAMAM) dendrimers (D-2PMPA) using a click chemistry approach. Cy5-labelled-D-2PMPA was used to visualize selective glial uptake in vitro and in vivo. D-2PMPA was evaluated for anti-inflammatory effects in LPS-treated glial cultures. In experimental autoimmune encephalomyelitis (EAE)-immunized mice, D-2PMPA was dosed biweekly starting at disease onset and cognition was assessed using the Barnes maze, and GCPII activity was measured in CD11b+ hippocampal cells. Results: D-2PMPA showed preferential uptake into microglia and robust anti-inflammatory activity, including elevations in NAAG, TGFβ, and mGluR3 in glial cultures. D-2PMPA significantly improved cognition in EAE mice, even though physical severity was unaffected. GCPII activity increased >20-fold in CD11b+ cells from EAE mice, which was significantly mitigated by D-2PMPA treatment. Conclusions: Hydroxyl dendrimers facilitate targeted drug delivery to activated microglia. These data support further development of D-2PMPA to attenuate elevated microglial GCPII activity and treat cognitive impairment in MS.
Collapse
Affiliation(s)
| | - Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Carolyn Tallon
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Lyndah Lovell
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Xiaolei Zhu
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Robyn Wiseman
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Siva P Kambhampati
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Kevin Liaw
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Rishi Sharma
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA.,Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.,Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
de Godoy LL, Studart-Neto A, Wylezinska-Arridge M, Tsunemi MH, Moraes NC, Yassuda MS, Coutinho AM, Buchpiguel CA, Nitrini R, Bisdas S, da Costa Leite C. The Brain Metabolic Signature in Superagers Using In Vivo 1H-MRS: A Pilot Study. AJNR Am J Neuroradiol 2021; 42:1790-1797. [PMID: 34446458 DOI: 10.3174/ajnr.a7262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 05/28/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Youthful memory performance in older adults may reflect an underlying resilience to the conventional pathways of aging. Subjects having this unusual characteristic have been recently termed "superagers." This study aimed to explore the significance of imaging biomarkers acquired by 1H-MRS to characterize superagers and to differentiate them from their normal-aging peers. MATERIALS AND METHODS Fifty-five patients older than 80 years of age were screened using a detailed neuropsychological protocol, and 25 participants, comprising 12 superagers and 13 age-matched controls, were statistically analyzed. We used state-of-the-art 3T 1H-MR spectroscopy to quantify 18 neurochemicals in the posterior cingulate cortex of our subjects. All 1H-MR spectroscopy data were analyzed using LCModel. Results were further processed using 2 approaches to investigate the technique accuracy: 1) comparison of the average concentration of metabolites estimated with Cramer-Rao lower bounds <20%; and 2) calculation and comparison of the weighted means of metabolites' concentrations. RESULTS The main finding observed was a higher total N-acetyl aspartate concentration in superagers than in age-matched controls using both approaches (P = .02 and P = .03 for the weighted means), reflecting a positive association of total N-acetyl aspartate with higher cognitive performance. CONCLUSIONS 1H-MR spectroscopy emerges as a promising technique to unravel neurochemical mechanisms related to cognitive aging in vivo and providing a brain metabolic signature in superagers. This may contribute to monitoring future interventional therapies to avoid or postpone the pathologic processes of aging.
Collapse
Affiliation(s)
- L L de Godoy
- From the Department of Radiology and Oncology (L.L.d.G., C.d.C.L.), Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- The National Hospital of Neurology and Neurosurgery (M.W.-A., S.B.), University College London, London, UK
| | - A Studart-Neto
- Department of Neurology (A.S.-N., N.C.M., M.S.Y., R.N.), Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - M Wylezinska-Arridge
- The National Hospital of Neurology and Neurosurgery (M.W.-A., S.B.), University College London, London, UK
| | - M H Tsunemi
- Department of Biostatistics, Institute of Biosciences (M.H.T.), Universidade Estadual Paulista, Botucatu, Sao Paulo, SP, Brazil
| | - N C Moraes
- Department of Neurology (A.S.-N., N.C.M., M.S.Y., R.N.), Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - M S Yassuda
- Department of Neurology (A.S.-N., N.C.M., M.S.Y., R.N.), Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - A M Coutinho
- Division and Laboratory of Nuclear Medicine (A.M.C., C.A.B.), Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - C A Buchpiguel
- Division and Laboratory of Nuclear Medicine (A.M.C., C.A.B.), Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - R Nitrini
- Department of Neurology (A.S.-N., N.C.M., M.S.Y., R.N.), Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - S Bisdas
- The National Hospital of Neurology and Neurosurgery (M.W.-A., S.B.), University College London, London, UK
| | - C da Costa Leite
- From the Department of Radiology and Oncology (L.L.d.G., C.d.C.L.), Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
17
|
Frontera A, Bauzá A. Glutamate carboxypeptidase II as a model system for designing host-guest units: a theoretical approach. Org Biomol Chem 2021; 19:7816-7821. [PMID: 34549237 DOI: 10.1039/d1ob01209f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Through the combination of X-ray structure analysis and density functional theory (DFT) theoretical calculations at the B3LYP-D3/def2-TZVP level of theory, we designed and investigated two novel host units for the recognition of neutral (e.g. halobenzenes) and charged (e.g. chloride and nitrate) guests inspired by a glutamate carboxypeptidase II (GCPII) system and its inhibitors.
Collapse
Affiliation(s)
- Antonio Frontera
- Department of Chemistry, Universitat de les Illes Balears, Crts de Valldemossa km 7.6, 07122 Palma de Mallorca (Baleares), Spain.
| | - Antonio Bauzá
- Department of Chemistry, Universitat de les Illes Balears, Crts de Valldemossa km 7.6, 07122 Palma de Mallorca (Baleares), Spain.
| |
Collapse
|
18
|
Inampudi C, Ciccotosto GD, Cappai R, Crack PJ. Genetic Modulators of Traumatic Brain Injury in Animal Models and the Impact of Sex-Dependent Effects. J Neurotrauma 2021; 37:706-723. [PMID: 32027210 DOI: 10.1089/neu.2019.6955] [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] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major health problem causing disability and death worldwide. There is no effective treatment, due in part to the complexity of the injury pathology and factors affecting its outcome. The extent of brain injury depends on the type of insult, age, sex, lifestyle, genetic risk factors, socioeconomic status, other co-injuries, and underlying health problems. This review discusses the genes that have been directly tested in TBI models, and whether their effects are known to be sex-dependent. Sex differences can affect the incidence, symptom onset, pathology, and clinical outcomes following injury. Adult males are more susceptible at the acute phase and females show greater injury in the chronic phase. TBI is not restricted to a single sex; despite variations in the degree of symptom onset and severity, it is important to consider both female and male animals in TBI pre-clinical research studies.
Collapse
Affiliation(s)
- Chaitanya Inampudi
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Roberto Cappai
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Nordengen K, Morland C, Slusher BS, Gundersen V. Dendritic Localization and Exocytosis of NAAG in the Rat Hippocampus. Cereb Cortex 2021; 30:1422-1435. [PMID: 31504271 PMCID: PMC7132944 DOI: 10.1093/cercor/bhz176] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022] Open
Abstract
While a lot is known about classical, anterograde neurotransmission, less is known about the mechanisms and molecules involved in retrograde neurotransmission. Our hypothesis is that N-acetylaspartylglutamate (NAAG), the most abundant dipeptide in the brain, may act as a retrograde transmitter in the brain. NAAG was predominantly localized in dendritic compartments of glutamatergic synapses in the intact hippocampus, where it was present in close proximity to synaptic-like vesicles. In acute hippocampal slices, NAAG was depleted from postsynaptic dendritic elements during neuronal stimulation induced by depolarizing concentrations of potassium or by exposure to glutamate receptor (GluR) agonists. The depletion was completely blocked by botulinum toxin B and strictly dependent on extracellular calcium, indicating exocytotic release. In contrast, there were low levels of NAAG and no effect by depolarization or GluR agonists in presynaptic glutamatergic terminals or GABAergic pre- and postsynaptic elements. Together these data suggest a possible role for NAAG as a retrograde signaling molecule at glutamatergic synapses via exocytotic release.
Collapse
Affiliation(s)
- K Nordengen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0317, Norway.,Department of Neurology, Akershus University Hospital, Lørenskog N-1478, Norway
| | - C Morland
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0317, Norway.,Department of Pharmaceutical Biosciences, Institute of Pharmacy, University of Oslo, Oslo NO-0317, Norway
| | - B S Slusher
- Department of Neurology and Johns Hopkins Drug Discovery, John Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - V Gundersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0317, Norway.,Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo N-0424, Norway.,Department of Neurology, Institute of Clinical Medicine, University of Oslo, Oslo NO-0317, Norway
| |
Collapse
|
20
|
Li Y, Liu K, Li C, Guo Y, Fang J, Tong H, Tang Y, Zhang J, Sun J, Jiao F, Zhang Q, Jin R, Xiong K, Chen X. 18F-FDG PET Combined With MR Spectroscopy Elucidates the Progressive Metabolic Cerebral Alterations After Blast-Induced Mild Traumatic Brain Injury in Rats. Front Neurosci 2021; 15:593723. [PMID: 33815036 PMCID: PMC8012735 DOI: 10.3389/fnins.2021.593723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/19/2021] [Indexed: 11/21/2022] Open
Abstract
A majority of blast-induced mild traumatic brain injury (mTBI) patients experience persistent neurological dysfunction with no findings on conventional structural MR imaging. It is urgent to develop advanced imaging modalities to detect and understand the pathophysiology of blast-induced mTBI. Fluorine-18 fluorodeoxyglucose positron emission tomography (18F-FDG PET) could detect neuronal function and activity of the injured brain, while MR spectroscopy provides complementary information and assesses metabolic irregularities following injury. This study aims to investigate the effectiveness of combining 18F-FDG PET with MR spectroscopy to evaluate acute and subacute metabolic cerebral alterations caused by blast-induced mTBI. Thirty-two adult male Sprague–Dawley rats were exposed to a single blast (mTBI group) and 32 rats were not exposed to the blast (sham group), followed by 18F-FDG PET, MRI, and histological evaluation at baseline, 1–3 h, 1 day, and 7 days post-injury in three separate cohorts. 18F-FDG uptake showed a transient increase in the amygdala and somatosensory cortex, followed by a gradual return to baseline from day 1 to 7 days post-injury and a continuous rise in the motor cortex. In contrast, decreased 18F-FDG uptake was seen in the midbrain structures (inferior and superior colliculus). Analysis of MR spectroscopy showed that inflammation marker myo-inositol (Ins), oxidative stress marker glutamine + glutamate (Glx), and hypoxia marker lactate (Lac) levels markedly elevated over time in the somatosensory cortex, while the major osmolyte taurine (Tau) level immediately increased at 1–3 h and 1 day, and then returned to sham level on 7 days post-injury, which could be due to the disruption of the blood–brain barrier. Increased 18F-FDG uptake and elevated Ins and Glx levels over time were confirmed by histology analysis which showed increased microglial activation and gliosis in the frontal cortex. These results suggest that 18F-FDG PET and MR spectroscopy can be used together to reflect more comprehensive neuropathological alterations in vivo, which could improve our understanding of the complex alterations in the brain after blast-induced mTBI.
Collapse
Affiliation(s)
- Yang Li
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China.,Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China.,Department of Medical Imaging, Air Force Hospital of Western Theater Command, Chengdu, China
| | - Kaijun Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Chang Li
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Tang
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jinju Sun
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Fangyang Jiao
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Qianhui Zhang
- Department of Foreign Language, Army Medical University, Chongqing, China
| | - Rongbing Jin
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Kunlin Xiong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China.,Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| |
Collapse
|
21
|
Becker I, Wang-Eckhardt L, Lodder-Gadaczek J, Wang Y, Grünewald A, Eckhardt M. Mice deficient in the NAAG synthetase II gene Rimkla are impaired in a novel object recognition task. J Neurochem 2021; 157:2008-2023. [PMID: 33638175 DOI: 10.1111/jnc.15333] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/11/2021] [Accepted: 02/21/2021] [Indexed: 12/27/2022]
Abstract
N-acetylaspartylglutamate (NAAG) is an abundant neuropeptide in the mammalian nervous system, synthesized by two related NAAG synthetases I and II (NAAGS-I and -II) encoded by the genes Rimklb and Rimkla, respectively. NAAG plays a role in cognition and memory, according to studies using inhibitors of the NAAG hydrolase glutamate carboxypeptidase II that increase NAAG concentration. To examine consequences of reduced NAAG concentration, Rimkla-deficient (Rimkla-/- ) mice were generated. These mice exhibit normal NAAG level at birth, likely because of the intact Rimklb gene, but have significantly reduced NAAG levels in all brain regions in adulthood. In wild type mice NAAGS-II was most abundant in brainstem and spinal cord, as demonstrated using a new NAAGS-II antiserum. In the hippocampus, NAAGS-II was only detectable in neurons expressing parvalbumin, a marker of GABAergic interneurons. Apart from reduced open field activity, general behavior of adult (6 months old) Rimkla-/- mice examined in different tests (dark-light transition, optokinetic behavior, rotarod, and alternating T-maze) was not significantly altered. However, Rimkla-/- mice were impaired in a short-term novel object recognition test. This was also the case for mice lacking NAA synthase Nat8l, which are devoid of NAAG. Together with results from previous studies showing that inhibition of the NAAG degrading enzyme glutamate carboxypeptidase II is associated with a significant improvement in object recognition, these results suggest a direct involvement of NAAG synthesized by NAAGS-II in the memory consolidation underlying the novel object recognition task.
Collapse
Affiliation(s)
- Ivonne Becker
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lihua Wang-Eckhardt
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Julia Lodder-Gadaczek
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Yong Wang
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Agathe Grünewald
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Matthias Eckhardt
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
22
|
Nguyen T, Kirsch BJ, Asaka R, Nabi K, Quinones A, Tan J, Antonio MJ, Camelo F, Li T, Nguyen S, Hoang G, Nguyen K, Udupa S, Sazeides C, Shen YA, Elgogary A, Reyes J, Zhao L, Kleensang A, Chaichana KL, Hartung T, Betenbaugh MJ, Marie SK, Jung JG, Wang TL, Gabrielson E, Le A. Uncovering the Role of N-Acetyl-Aspartyl-Glutamate as a Glutamate Reservoir in Cancer. Cell Rep 2020; 27:491-501.e6. [PMID: 30970252 PMCID: PMC6472703 DOI: 10.1016/j.celrep.2019.03.036] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/14/2019] [Accepted: 03/08/2019] [Indexed: 11/13/2022] Open
Abstract
N-acetyl-aspartyl-glutamate (NAAG) is a peptide-based neurotransmitter that has been extensively studied in many neurological diseases. In this study, we show a specific role of NAAG in cancer. We found that NAAG is more abundant in higher grade cancers and is a source of glutamate in cancers expressing glutamate carboxypeptidase II (GCPII), the enzyme that hydrolyzes NAAG to glutamate and N-acetyl-aspartate (NAA). Knocking down GCPII expression through genetic alteration or pharmacological inhibition of GCPII results in a reduction of both glutamate concentrations and cancer growth. Moreover, targeting GCPII in combination with glutaminase inhibition accentuates these effects. These findings suggest that NAAG serves as an important reservoir to provide glutamate to cancer cells through GCPII when glutamate production from other sources is limited. Thus, GCPII is a viable target for cancer therapy, either alone or in combination with glutaminase inhibition. Nguyen et al. show that NAAG is more abundant in higher grade cancers and a source of glutamate in cancers expressing GCPII, the enzyme that hydrolyzes NAAG to glutamate and NAA. The results suggest that GCPII is a viable target for cancer therapy, either alone or in combination with glutaminase inhibition.
Collapse
Affiliation(s)
- Tu Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brian James Kirsch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Ryoichi Asaka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Karim Nabi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Addison Quinones
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jessica Tan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Felipe Camelo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ting Li
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephanie Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Giang Hoang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kiet Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sunag Udupa
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christos Sazeides
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yao-An Shen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amira Elgogary
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Juvenal Reyes
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Liang Zhao
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Andre Kleensang
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kaisorn Lee Chaichana
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; University of Konstanz, 78464 Konstanz, Germany
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Suely K Marie
- Department of Neurology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Jin G Jung
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anne Le
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
23
|
Fu C, Wu Q, Zhang Z, Xia Z, Ji H, Lu H, Wang Y. UPLC-ESI-IT-TOF-MS metabolomic study of the therapeutic effect of Xuefu Zhuyu decoction on rats with traumatic brain injury. JOURNAL OF ETHNOPHARMACOLOGY 2019; 245:112149. [PMID: 31401321 DOI: 10.1016/j.jep.2019.112149] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 06/10/2023]
Abstract
It has been widely reported that Xuefu Zhuyu decoction (XFZYD), a traditional Chinese medicine, is effective in the treatment of traumatic brain injury (TBI). However, the mechanism of the therapeutic process is still not fully understood. Metabolomic technique can be used to explore the mechanisms underlying the treatment of TBI with XFZYD. The purpose of this work was to investigate the metabolic characteristics of blood samples from rats with and without XFZYD treatment and the dynamic changes in metabolite profiles on days 1, 3, 7, 14 and 21 after injury (within the severe phase of TBI) based on untargeted UPLC-ESI-IT-TOF-MS analysis. Pattern recognition, clustering analysis and metabolic pathway analysis were used to analyse the metabolomic data of three groups (a sham-operated group, a TBI model, and an XFZYD-treated TBI model). The results showed that XFZYD reversed the abnormalities in the levels of small-molecule metabolites (such as L-acetylcarnitine, L-tryptophan, indoleacrylic acid, γ-aminobutyric acid, hypotaurine, LysoPC(18:1)(11Z), creatine, L-phenylalanine and L-leucine) in TBI rats through six metabolic pathways (including phenylalanine, tyrosine and tryptophan biosynthesis; phenylalanine metabolism; valine, leucine and isoleucine biosynthesis; taurine and hypotaurine metabolism; tryptophan metabolism; and alanine, aspartate and glutamate metabolism) involved in the therapy process. XFZYD regulated the metabolic disorders of endogenous markers by the possible mechanisms of neuroprotection, energy metabolism, inflammatory response and oxidative stress. This study revealed the holistic and dynamic metabolic changes caused by XFZYD in rats with TBI and provided important research methods and approaches for exploring the multiple metabolites and metabolic pathways involved in the therapeutic effect of XFZYD on TBI.
Collapse
Affiliation(s)
- Chunyan Fu
- College of Chemistry and Chemical Engineering, Central South University, Hunan, Changsha, PR China; College of Pharmacy, Shaoyang University, Hunan, Shaoyang, PR China
| | - Qian Wu
- College of Chemistry and Chemical Engineering, Central South University, Hunan, Changsha, PR China
| | - Zhimin Zhang
- College of Chemistry and Chemical Engineering, Central South University, Hunan, Changsha, PR China
| | - Zian Xia
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Hunan, Changsha, PR China
| | - Hongchao Ji
- College of Chemistry and Chemical Engineering, Central South University, Hunan, Changsha, PR China
| | - Hongmei Lu
- College of Chemistry and Chemical Engineering, Central South University, Hunan, Changsha, PR China.
| | - Yang Wang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Hunan, Changsha, PR China.
| |
Collapse
|
24
|
Neale JH, Yamamoto T. N-acetylaspartylglutamate (NAAG) and glutamate carboxypeptidase II: An abundant peptide neurotransmitter-enzyme system with multiple clinical applications. Prog Neurobiol 2019; 184:101722. [PMID: 31730793 DOI: 10.1016/j.pneurobio.2019.101722] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/24/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
N-Acetylaspartylglutamate (NAAG) is the third most prevalent neurotransmitter in the mammalian nervous system, yet its therapeutic potential is only now being fully recognized. Drugs that inhibit the inactivation of NAAG by glutamate carboxypeptidase II (GCPII) increase its extracellular concentration and its activation of its receptor, mGluR3. These drugs warrant attention, as they are effective in animal models of several clinical disorders including stroke, traumatic brain injury and schizophrenia. In inflammatory and neuropathic pain studies, GCPII inhibitors moderated both the primary and secondary pain responses when given systemically, locally or in brain regions associated with the pain perception pathway. The finding that GCPII inhibition also moderated the motor and cognitive effects of ethanol intoxication led to the discovery of their procognitive efficacy in long-term memory tests in control mice and in short-term memory in a mouse model of Alzheimer's disease. NAAG and GCPII inhibitors respectively reduce cocaine self-administration and the rewarding effects of a synthetic stimulant. Most recently, GCPII inhibition also has been reported to be efficacious in a model of inflammatory bowel disease. GCPII was first discovered as a protein expressed by and released from metastatic prostate cells where it is known as prostate specific membrane antigen (PSMA). GCPII inhibitors with high affinity for this protein have been developed as prostate imaging and radiochemical therapies for prostate cancer. Taken together, these data militate in favor of the development and application of GCPII inhibitors in more advanced preclinical research as a prelude to clinical trials.
Collapse
Affiliation(s)
- Joseph H Neale
- Department of Biology, Georgetown University, 37(th) and O Sts., NW, Washington, DC, 20057, USA.
| | - Tatsuo Yamamoto
- Dept. of Anesthesiology, Kumamoto University., Kumamoto, Japan
| |
Collapse
|
25
|
Chen L, Chao Y, Cheng P, Li N, Zheng H, Yang Y. UPLC-QTOF/MS-Based Metabolomics Reveals the Protective Mechanism of Hydrogen on Mice with Ischemic Stroke. Neurochem Res 2019; 44:1950-1963. [PMID: 31236794 DOI: 10.1007/s11064-019-02829-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/11/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022]
Abstract
As a reductive gas, hydrogen plays an antioxidant role by selectively scavenging oxygen free radicals. It has been reported that hydrogen has protective effects against nerve damage caused by ischemia-reperfusion in stroke, but the specific mechanism is still unclear. Therefore, this study aims to investigate the protective effects of hydrogen on stroke-induced ischemia-reperfusion injury and its detailed mechanism. Two weeks after the inhalation of high concentrations (66.7%) of hydrogen, middle cerebral artery occlusion (MCAO) was induced in mice using the thread occlusion technique to establish an animal model of the focal cerebral ischemia-reperfusion. Then, a metabolomics analysis of mouse cerebral cortex tissues was first performed by ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF/MS) to study the metabolic changes and protective mechanisms of hydrogen on stroke ischemia-reperfusion injury. According to the metabolomic profiling of cortex tissues, 29 different endogenous metabolites were screened, including palmitoyl-L-carnitine, citric acid, glutathione, taurine, acetyl-L-carnitine, N-acetylaspartylglutamic acid (NAAG), L-aspartic acid, lysophosphatidylcholine (LysoPC) and lysophosphatidylethanolamine (LysoPE). Through pathway analysis, the metabolic pathways were concentrate on the glutathione pathway and the taurine pathway, mitochondrial energy metabolism and phospholipid metabolism that related to the oxidative stress process. This result reveals that hydrogen may protect against ischemic stroke by reducing oxidative stress during ischemia-reperfusion, thereby protecting nerve cells from reactive oxygen species(ROS).
Collapse
Affiliation(s)
- Lilin Chen
- College of Basic Medicine, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Yufan Chao
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Pengchao Cheng
- College of Basic Medicine, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Na Li
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Hongnan Zheng
- Department of Natural Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi, People's Republic of China.
| | - Yajuan Yang
- Department of Nursing, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
26
|
Hollinger KR, Alt J, Rais R, Kaplin AI, Slusher BS. The NAAG’ing Concerns of Modeling Human Alzheimer’s Disease in Mice. J Alzheimers Dis 2019; 68:939-945. [DOI: 10.3233/jad-181251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kristen R. Hollinger
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Rana Rais
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Adam I. Kaplin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Barbara S. Slusher
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
27
|
Neale JH, Olszewski R. A role for N-acetylaspartylglutamate (NAAG) and mGluR3 in cognition. Neurobiol Learn Mem 2019; 158:9-13. [PMID: 30630041 DOI: 10.1016/j.nlm.2019.01.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/03/2018] [Accepted: 01/05/2019] [Indexed: 12/18/2022]
Abstract
The peptide transmitter N-acetylaspartylglutamate (NAAG) and its receptor, the type 3 metabotropic glutamate receptor (mGluR3, GRM3), are prevalent and widely distributed in the mammalian nervous system. Drugs that inhibit the inactivation of synaptically released NAAG have procognitive activity in object recognition and other behavioral models. These inhibitors also reverse cognitive deficits in animal models of clinical disorders. Antagonists of mGluR3 block these actions and mice that are null mutant for this receptor are insensitive to the actions of these procognitive drugs. A positive allosteric modulator of this receptor also has procognitive activity. While some data suggest that drugs acting on mGluR3 achieve their procognitive action by increasing arousal during acquisition training, exploration of the procognitive efficacy of NAAG is in its early stages and thus substantial opportunities exist to define the breadth and nature of this activity.
Collapse
Affiliation(s)
- Joseph H Neale
- Department of Biology, Georgetown University, Washington, DC, USA.
| | - Rafal Olszewski
- Department of Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
28
|
Metabolite differences between glutamate carboxypeptidase II gene knockout mice and their wild-type littermates after traumatic brain injury: a 7-tesla 1H-MRS study. BMC Neurosci 2018; 19:75. [PMID: 30458729 PMCID: PMC6245916 DOI: 10.1186/s12868-018-0473-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 11/03/2018] [Indexed: 11/30/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a complex condition and remains a prominent public and medical health issue in individuals of all ages. A rapid increase in extracellular glutamate occurs after TBI, leading to glutamate-induced excitotoxicity, which causes neuronal damage and further functional impairments. Although inhibition of glutamate carboxypeptidase II (GCP II) is considered a potential approach for reducing glutamate-induced excitotoxicity after TBI, further detailed evidence regarding its efficacy is required. Therefore, in this study, we examined the differences in the metabolite status between wild-type (WT) and GCP II gene-knockout (KO) mice after TBI using proton magnetic resonance spectroscopy (1H-MRS) and T2-weighted magnetic resonance (MR) imaging with a 7-tesla imaging system, and brain water-content analysis. Results Evaluation of glutamate and N-acetylaspartate concentrations revealed a decrease in both levels in the ipsilateral hippocampus at 24 h post-TBI; however, the reduction in glutamate and N-acetylaspartate levels was less marked in GCP II-KO mice than in WT mice (p < 0.05). T2 MR data and brain water-content analysis demonstrated that the extent of cortical edema and brain swelling was less in KO than in WT mice after TBI (p < 0.05). Conclusion Using two non-invasive methods, 1H-MRS and T2 MR imaging, as well as in vitro brain-water content measurements, we demonstrated that the mechanism underlying the neuroprotective effects of GCP II-KO against brain swelling in TBI involves changes in glutamate and N-acetylaspartate levels. This knowledge may contribute towards the development of therapeutic strategies for TBI.
Collapse
|
29
|
de Matos NM, Hock A, Wyss M, Ettlin DA, Brügger M. Neurochemical dynamics of acute orofacial pain in the human trigeminal brainstem nuclear complex. Neuroimage 2017; 162:162-172. [DOI: 10.1016/j.neuroimage.2017.08.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 01/25/2023] Open
|
30
|
Singh K, Trivedi R, Verma A, D'souza MM, Koundal S, Rana P, Baishya B, Khushu S. Altered metabolites of the rat hippocampus after mild and moderate traumatic brain injury - a combined in vivo and in vitro 1 H-MRS study. NMR IN BIOMEDICINE 2017; 30:e3764. [PMID: 28759166 DOI: 10.1002/nbm.3764] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 05/13/2017] [Accepted: 05/28/2017] [Indexed: 06/07/2023]
Abstract
Traumatic brain injury (TBI) has been shown to affect hippocampus-associated learning, memory and higher cognitive functions, which may be a consequence of metabolic alterations. Hippocampus-associated disorders may vary depending on the severity of injury [mild TBI (miTBI) and moderate TBI (moTBI)] and time since injury. The underlying hippocampal metabolic irregularities may provide an insight into the pathological process following TBI. In this study, in vivo and in vitro proton magnetic resonance spectroscopy (1 H-MRS) data were acquired from the hippocampus region of controls and TBI groups (miTBI and moTBI) at D0 (pre-injury), 4 h, Day 1 and Day 5 post-injury (PI). In vitro MRS results indicated trauma-induced changes in both miTBI and moTBI; however, in vivo MRS showed metabolic alterations in moTBI only. miTBI and moTBI showed elevated levels of osmolytes indicating injury-induced edema. Altered levels of citric acid cycle intermediates, glutamine/glutamate and amino acid metabolism indicated injury-induced aberrant bioenergetics, excitotoxicity and oxidative stress. An overall similar pattern of pathological process was observed in both miTBI and moTBI, with the distinction of depleted N-acetylaspartate levels (indicating neuronal loss) at 4 h and Day 1 and enhanced lactate production (indicating heightened energy depletion leading to the commencement of the anaerobic pathway) at Day 5 in moTBI. To the best of our knowledge, this is the first study to investigate the hippocampus metabolic profile in miTBI and moTBI simultaneously using in vivo and in vitro MRS.
Collapse
Affiliation(s)
- Kavita Singh
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Richa Trivedi
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Ajay Verma
- Centre for Biomedical Magnetic Resonance (CBMR), SGPGIMS Campus, Lucknow, Uttar Pradesh, India
| | - Maria M D'souza
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Sunil Koundal
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Poonam Rana
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Bikash Baishya
- Centre for Biomedical Magnetic Resonance (CBMR), SGPGIMS Campus, Lucknow, Uttar Pradesh, India
| | - Subash Khushu
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
31
|
Knedlík T, Vorlová B, Navrátil V, Tykvart J, Sedlák F, Vaculín Š, Franěk M, Šácha P, Konvalinka J. Mouse glutamate carboxypeptidase II (GCPII) has a similar enzyme activity and inhibition profile but a different tissue distribution to human GCPII. FEBS Open Bio 2017; 7:1362-1378. [PMID: 28904865 PMCID: PMC5586342 DOI: 10.1002/2211-5463.12276] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/23/2017] [Accepted: 07/19/2017] [Indexed: 11/09/2022] Open
Abstract
Glutamate carboxypeptidase II (GCPII), also known as prostate-specific membrane antigen (PSMA) or folate hydrolase, is a metallopeptidase expressed predominantly in the human brain and prostate. GCPII expression is considerably increased in prostate carcinoma, and the enzyme also participates in glutamate excitotoxicity in the brain. Therefore, GCPII represents an important diagnostic marker of prostate cancer progression and a putative target for the treatment of both prostate cancer and neuronal disorders associated with glutamate excitotoxicity. For the development of novel therapeutics, mouse models are widely used. However, although mouse GCPII activity has been characterized, a detailed comparison of the enzymatic activity and tissue distribution of the mouse and human GCPII orthologs remains lacking. In this study, we prepared extracellular mouse GCPII and compared it with human GCPII. We found that mouse GCPII possesses lower catalytic efficiency but similar substrate specificity compared with the human protein. Using a panel of GCPII inhibitors, we discovered that inhibition constants are generally similar for mouse and human GCPII. Furthermore, we observed highest expression of GCPII protein in the mouse kidney, brain, and salivary glands. Importantly, we did not detect GCPII in the mouse prostate. Our data suggest that the differences in enzymatic activity and inhibition profile are rather small; therefore, mouse GCPII can approximate human GCPII in drug development and testing. On the other hand, significant differences in GCPII tissue expression must be taken into account when developing novel GCPII-based anticancer and therapeutic methods, including targeted anticancer drug delivery systems, and when using mice as a model organism.
Collapse
Affiliation(s)
- Tomáš Knedlík
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic.,Department of Biochemistry Faculty of Science Charles University Prague Czech Republic
| | - Barbora Vorlová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic.,First Faculty of Medicine Charles University Prague Czech Republic
| | - Václav Navrátil
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic.,Department of Biochemistry Faculty of Science Charles University Prague Czech Republic
| | - Jan Tykvart
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic.,Department of Biochemistry Faculty of Science Charles University Prague Czech Republic.,Present address: Donnelly Centre for Cellular and Biomolecular Research University of Toronto Toronto ON Canada
| | - František Sedlák
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic.,First Faculty of Medicine Charles University Prague Czech Republic.,Department of Genetics and Microbiology Faculty of Science Charles University Prague Czech Republic
| | - Šimon Vaculín
- Department of Normal, Pathological and Clinical Physiology Third Faculty of Medicine Charles University Prague Czech Republic
| | - Miloslav Franěk
- Department of Normal, Pathological and Clinical Physiology Third Faculty of Medicine Charles University Prague Czech Republic
| | - Pavel Šácha
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic
| | - Jan Konvalinka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic.,Department of Biochemistry Faculty of Science Charles University Prague Czech Republic
| |
Collapse
|
32
|
Azimi N, Yadollahikhales G, Argenti JP, Cunningham MG. Discrepancies in stereotaxic coordinate publications and improving precision using an animal-specific atlas. J Neurosci Methods 2017; 284:15-20. [PMID: 28392415 DOI: 10.1016/j.jneumeth.2017.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
Rodent brain atlases have traditionally been used to identify brain structures in three-dimensional space for a variety of stereotaxic procedures. As neuroscience becomes increasingly sophisticated, higher levels of precision and consistency are needed. Observations of various atlases currently in use across labs reveal numerous coordinate discrepancies. Here we provide examples of inconsistencies by comparing the coordinates of the boundaries of various brain structures across six atlas publications. We conclude that the coordinates determined by any particular atlas should be considered as only a first approximation of the actual target coordinates for the experimental animal for a particular study. Furthermore, the coordinates determined by one research team cannot be assumed to be universally applicable and accurate in other experimental settings. To optimize precision, we describe a simple protocol for the construction of a customized atlas that is specific to the surgical approach and to the species, gender, and age of the animal used in any given study.
Collapse
Affiliation(s)
- Nima Azimi
- Laboratory for Neural Reconstruction, McLean Hospital Department of Psychiatry, 115 Mill Street Belmont, MA 02478, United States
| | - Golnaz Yadollahikhales
- Laboratory for Neural Reconstruction, McLean Hospital Department of Psychiatry, 115 Mill Street Belmont, MA 02478, United States
| | - John Paul Argenti
- Laboratory for Neural Reconstruction, McLean Hospital Department of Psychiatry, 115 Mill Street Belmont, MA 02478, United States
| | - Miles G Cunningham
- Laboratory for Neural Reconstruction, McLean Hospital Department of Psychiatry, 115 Mill Street Belmont, MA 02478, United States.
| |
Collapse
|
33
|
Olszewski RT, Janczura KJ, Bzdega T, Der EK, Venzor F, O'Rourke B, Hark TJ, Craddock KE, Balasubramanian S, Moussa C, Neale JH. NAAG Peptidase Inhibitors Act via mGluR3: Animal Models of Memory, Alzheimer's, and Ethanol Intoxication. Neurochem Res 2017; 42:2646-2657. [PMID: 28285415 PMCID: PMC5603630 DOI: 10.1007/s11064-017-2181-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 01/08/2017] [Accepted: 01/16/2017] [Indexed: 11/26/2022]
Abstract
Glutamate carboxypeptidase II (GCPII) inactivates the peptide neurotransmitter N-acetylaspartylglutamate (NAAG) following synaptic release. Inhibitors of GCPII increase extracellular NAAG levels and are efficacious in animal models of clinical disorders via NAAG activation of a group II metabotropic glutamate receptor. mGluR2 and mGluR3 knock-out (ko) mice were used to test the hypothesis that mGluR3 mediates the activity of GCPII inhibitors ZJ43 and 2-PMPA in animal models of memory and memory loss. Short- (1.5 h) and long- (24 h) term novel object recognition tests were used to assess memory. Treatment with ZJ43 or 2-PMPA prior to acquisition trials increased long-term memory in mGluR2, but not mGluR3, ko mice. Nine month-old triple transgenic Alzheimer's disease model mice exhibited impaired short-term novel object recognition memory that was rescued by treatment with a NAAG peptidase inhibitor. NAAG peptidase inhibitors and the group II mGluR agonist, LY354740, reversed the short-term memory deficit induced by acute ethanol administration in wild type mice. 2-PMPA also moderated the effect of ethanol on short-term memory in mGluR2 ko mice but failed to do so in mGluR3 ko mice. LY354740 and ZJ43 blocked ethanol-induced motor activation. Both GCPII inhibitors and LY354740 also significantly moderated the loss of motor coordination induced by 2.1 g/kg ethanol treatment. These data support the conclusion that inhibitors of glutamate carboxypeptidase II are efficacious in object recognition models of normal memory and memory deficits via an mGluR3 mediated process, actions that could have widespread clinical applications.
Collapse
Affiliation(s)
- Rafal T Olszewski
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Karolina J Janczura
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Tomasz Bzdega
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Elise K Der
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Faustino Venzor
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Brennen O'Rourke
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Timothy J Hark
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Kirsten E Craddock
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Shankar Balasubramanian
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA
| | - Charbel Moussa
- Department of Neuroscience, Georgetown University, Washington, D.C., 20057, USA
| | - Joseph H Neale
- Department of Biology, Georgetown University, 37th and O Sts., N.W., Washington, D.C., 20057-1225, USA.
| |
Collapse
|
34
|
Nonaka T, Yamada T, Ishimura T, Zuo D, Moffett JR, Neale JH, Yamamoto T. A role for the locus coeruleus in the analgesic efficacy of N-acetylaspartylglutamate peptidase (GCPII) inhibitors ZJ43 and 2-PMPA. Mol Pain 2017; 13:1744806917697008. [PMID: 28326936 PMCID: PMC5407666 DOI: 10.1177/1744806917697008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 11/16/2022] Open
Abstract
N-acetylaspartylglutamate (NAAG) is the third most prevalent and widely distributed neurotransmitter in the mammalian nervous system. NAAG activates a group II metabotropic glutamate receptor (mGluR3) and is inactivated by an extracellular enzyme, glutamate carboxypeptidase II (GCPII) in vivo. Inhibitors of this enzyme are analgesic in animal models of inflammatory, neuropathic and bone cancer pain. NAAG and GCPII are present in the locus coeruleus, a center for the descending noradrenergic inhibitory pain system. In the formalin footpad model, systemic treatment with GCPII inhibitors reduces both phases of the inflammatory pain response and increases release of spinal noradrenaline. This analgesic efficacy is blocked by systemic injection of a group II mGluR antagonist, by intrathecal (spinal) injection of an alpha 2 adrenergic receptor antagonist and by microinjection of an α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor antagonist directly into the contralateral locus coeruleus. Footpad inflammation increases release of glutamate in the contralateral locus coeruleus and systemic treatment with a GCPII inhibitor blocks this increase. Direct injection of GCPII inhibitors into the contralateral or ipsilateral locus coeruleus reduces both phases of the inflammatory pain response in a dose-dependent manner and the contralateral effect also is blocked by intrathecal injection of an alpha 2 adrenergic receptor antagonist. These data support the hypothesis that the analgesic efficacy of systemically administered GCPII inhibitors is mediated, at least in part, by the contralateral locus coeruleus via group II mGluR, AMPA and alpha 2 adrenergic receptors.
Collapse
Affiliation(s)
- Takahiro Nonaka
- Department of Anesthesiology, School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Toshihiko Yamada
- Department of Anesthesiology, School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Tatsuhiro Ishimura
- Department of Anesthesiology, School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - John R Moffett
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joseph H Neale
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Tatsuo Yamamoto
- Department of Anesthesiology, School of Medical Science, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
35
|
Evans JC, Malhotra M, Cryan JF, O'Driscoll CM. The therapeutic and diagnostic potential of the prostate specific membrane antigen/glutamate carboxypeptidase II (PSMA/GCPII) in cancer and neurological disease. Br J Pharmacol 2016; 173:3041-3079. [PMID: 27526115 PMCID: PMC5056232 DOI: 10.1111/bph.13576] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
Prostate specific membrane antigen (PSMA) otherwise known as glutamate carboxypeptidase II (GCPII) is a membrane bound protein that is highly expressed in prostate cancer and in the neovasculature of a wide variety of tumours including glioblastomas, breast and bladder cancers. This protein is also involved in a variety of neurological diseases including schizophrenia and ALS. In recent years, there has been a surge in the development of both diagnostics and therapeutics that take advantage of the expression and activity of PSMA/GCPII. These include gene therapy, immunotherapy, chemotherapy and radiotherapy. In this review, we discuss the biological roles that PSMA/GCPII plays, both in normal and diseased tissues, and the current therapies exploiting its activity that are at the preclinical stage. We conclude by giving an expert opinion on the future direction of PSMA/GCPII based therapies and diagnostics and hurdles that need to be overcome to make them effective and viable.
Collapse
Affiliation(s)
- James C Evans
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | |
Collapse
|
36
|
Ravin R, Blank PS, Busse B, Ravin N, Vira S, Bezrukov L, Waters H, Guerrero-Cazares H, Quinones-Hinojosa A, Lee PR, Fields RD, Bezrukov SM, Zimmerberg J. Blast shockwaves propagate Ca(2+) activity via purinergic astrocyte networks in human central nervous system cells. Sci Rep 2016; 6:25713. [PMID: 27162174 PMCID: PMC4861979 DOI: 10.1038/srep25713] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/21/2016] [Indexed: 12/26/2022] Open
Abstract
In a recent study of the pathophysiology of mild, blast-induced traumatic brain injury (bTBI) the exposure of dissociated, central nervous system (CNS) cells to simulated blast resulted in propagating waves of elevated intracellular Ca2+. Here we show, in dissociated human CNS cultures, that these calcium waves primarily propagate through astrocyte-dependent, purinergic signaling pathways that are blocked by P2 antagonists. Human, compared to rat, astrocytes had an increased calcium response and prolonged calcium wave propagation kinetics, suggesting that in our model system rat CNS cells are less responsive to simulated blast. Furthermore, in response to simulated blast, human CNS cells have increased expressions of a reactive astrocyte marker, glial fibrillary acidic protein (GFAP) and a protease, matrix metallopeptidase 9 (MMP-9). The conjoint increased expression of GFAP and MMP-9 and a purinergic ATP (P2) receptor antagonist reduction in calcium response identifies both potential mechanisms for sustained changes in brain function following primary bTBI and therapeutic strategies targeting abnormal astrocyte activity.
Collapse
Affiliation(s)
- Rea Ravin
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA.,Celoptics Inc., Rockville, MD 20852, USA
| | - Paul S Blank
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| | - Brad Busse
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| | - Nitay Ravin
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA.,Celoptics Inc., Rockville, MD 20852, USA
| | - Shaleen Vira
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| | - Ludmila Bezrukov
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| | - Hang Waters
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| | | | | | - Philip R Lee
- Section on Nervous System Development and Plasticity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-3713, USA
| | - R Douglas Fields
- Section on Nervous System Development and Plasticity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-3713, USA
| | - Sergey M Bezrukov
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-0924, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| |
Collapse
|
37
|
Cao Y, Gao Y, Xu S, Bao J, Lin Y, Luo X, Wang Y, Luo Q, Jiang J, Neale JH, Zhong C. Glutamate carboxypeptidase II gene knockout attenuates oxidative stress and cortical apoptosis after traumatic brain injury. BMC Neurosci 2016; 17:15. [PMID: 27091009 PMCID: PMC4836105 DOI: 10.1186/s12868-016-0251-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/13/2016] [Indexed: 12/15/2022] Open
Abstract
Background Glutamate carboxypeptidase II (GCPII) inactivates the peptide co-transmitter N-acetylaspartylglutamate following synaptic release. Inhibition of GCPII elevates extracellular levels of the peptide, inhibits glutamate release and is neuroprotective in an animal model of traumatic brain injury. GCPII gene knockout mice were used to examine the cellular mechanisms underlying the neuroprotective efficacy of this transmitter system. Results Following controlled cortical impact injury, GCPII knockout (KO) mice exhibited reduced TUNEL-positive nuclei in the contusion margin of the cerebral cortex relative to wild type mice. Impact injury reduced glutathione levels and superoxide dismutase and glutathione peroxidase activities and increased malondialdehyde. Each of these effects was moderated in KO mice relative to wild type. Similarly, the injury-induced increases in cleaved caspase-3, cytosolic cytochrome c levels and Bcl-2/Bax ratio observed in wild type mice were attenuated in the knockout mice. Conclusions These data support the hypothesis that the neuroprotective efficacy of GCPII KO in traumatic brain injury is mediated via a reduction in oxidative stress.
Collapse
Affiliation(s)
- Yang Cao
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yang Gao
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Siyi Xu
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Jingang Bao
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yingying Lin
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Xingguang Luo
- Department of Psychiatry, Yale University, School of Medicine, West Haven, CT, 06516, USA
| | - Yong Wang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Qizhong Luo
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Jiyao Jiang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Joseph H Neale
- Department of Biology, Georgetown University, Washington, DC, 20075, USA
| | - Chunlong Zhong
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
38
|
Khacho P, Wang B, Bergeron R. The Good and Bad Sides of NAAG. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 76:311-49. [PMID: 27288081 DOI: 10.1016/bs.apha.2016.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Why has such a small peptide been the source of controversy in neuroscience over the last 5 decades? Is N-acetyl-aspartyl-glutamate (NAAG) a neurotransmitter? Is NAAG located in neuronal tissue or in astrocytes? Is NAAG involved in neuropsychiatric and neurodegenerative disorders? Is NAAG therapeutically beneficial in the treatment of stroke or in initiating cascades of events leading to psychosis? After many years of intense research there is no clear consensus within the scientific community on how NAAG behaves in the brain. One of the major controversies about NAAG is its physiological action at N-methyl-d-aspartate (NMDA) receptors. While some researchers strongly argue that NAAG acts as a weak agonist at NMDA receptors, others have suggested that NAAG could behave as a potent antagonist. Published data from our laboratory demonstrate that the effect of NAAG on NMDA receptors could be influenced by a number of factors including the subcellular localization and subunit composition of NMDA receptors, as well as protons. In this chapter, we will summarize the knowledge of the literature on NAAG, however, we will place emphasis on our recently published data. More specifically, we have reported interesting findings on the effects of NAAG on NMDA receptors at synaptic and extrasynaptic sites using a pharmacological paradigm to distinguish the two populations of NMDA receptors. Additionally, we have evaluated the role of NAAG on GluN2A- and GluN2B-containing NMDA receptors using a HEK293 cell recombinant system. Finally, we have studied the effects of NAAG on GluN2A- and GluN2B-containing NMDA receptors in different extracellular pH conditions. We believe that our findings could potentially resolve some aspects of the debate regarding the role of NAAG at NMDA receptors.
Collapse
Affiliation(s)
- P Khacho
- University of Ottawa, Ottawa, ON, Canada
| | - B Wang
- University of Ottawa, Ottawa, ON, Canada
| | - R Bergeron
- University of Ottawa, Ottawa, ON, Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
39
|
Still NAAG’ing After All These Years. NEUROPSYCHOPHARMACOLOGY: A TRIBUTE TO JOSEPH T. COYLE 2016; 76:215-55. [DOI: 10.1016/bs.apha.2016.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
40
|
Src Family Kinases in Brain Edema After Acute Brain Injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:185-90. [PMID: 26463946 DOI: 10.1007/978-3-319-18497-5_33] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Brain edema, the first stage of intracranial hypertension, has been associated with poor prognosis and increased mortality after acute brain injury such as ischemic stroke, intracranial hemorrhage (ICH), and traumatic brain injury (TBI). Acute brain injury often initiates release of many molecules, including glutamate, adenosine, thrombin, oxyhemoglobin, cytokines, reactive oxygen species (ROS), damage-associated molecular pattern molecules (DAMPs), and others. Most of these molecules activate Src family kinases (SFKs), a family of proto-oncogenic non-receptor tyrosine kinases, resulting in blood-brain barrier (BBB) disruption and brain edema at the acute stage after brain injury. However, SFKs also contribute to BBB self-repair and brain edema resolution in the chronic stage that follows brain injury. In this review, we summarize possible pathways through which SFKs are implicated in both brain edema formation and its eventual resolution.
Collapse
|
41
|
Rais R, Wozniak K, Wu Y, Niwa M, Stathis M, Alt J, Giroux M, Sawa A, Rojas C, Slusher BS. Selective CNS Uptake of the GCP-II Inhibitor 2-PMPA following Intranasal Administration. PLoS One 2015; 10:e0131861. [PMID: 26151906 PMCID: PMC4494705 DOI: 10.1371/journal.pone.0131861] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 06/09/2015] [Indexed: 11/19/2022] Open
Abstract
Glutamate carboxypeptidase II (GCP-II) is a brain metallopeptidase that hydrolyzes the abundant neuropeptide N-acetyl-aspartyl-glutamate (NAAG) to NAA and glutamate. Small molecule GCP-II inhibitors increase brain NAAG, which activates mGluR3, decreases glutamate, and provide therapeutic utility in a variety of preclinical models of neurodegenerative diseases wherein excess glutamate is presumed pathogenic. Unfortunately no GCP-II inhibitor has advanced clinically, largely due to their highly polar nature resulting in insufficient oral bioavailability and limited brain penetration. Herein we report a non-invasive route for delivery of GCP-II inhibitors to the brain via intranasal (i.n.) administration. Three structurally distinct classes of GCP-II inhibitors were evaluated including DCMC (urea-based), 2-MPPA (thiol-based) and 2-PMPA (phosphonate-based). While all showed some brain penetration following i.n. administration, 2-PMPA exhibited the highest levels and was chosen for further evaluation. Compared to intraperitoneal (i.p.) administration, equivalent doses of i.n. administered 2-PMPA resulted in similar plasma exposures (AUC0-t, i.n./AUC0-t, i.p. = 1.0) but dramatically enhanced brain exposures in the olfactory bulb (AUC0-t, i.n./AUC0-t, i.p. = 67), cortex (AUC0-t, i.n./AUC0-t, i.p. = 46) and cerebellum (AUC0-t, i.n./AUC0-t, i.p. = 6.3). Following i.n. administration, the brain tissue to plasma ratio based on AUC0-t in the olfactory bulb, cortex, and cerebellum were 1.49, 0.71 and 0.10, respectively, compared to an i.p. brain tissue to plasma ratio of less than 0.02 in all areas. Furthermore, i.n. administration of 2-PMPA resulted in complete inhibition of brain GCP-II enzymatic activity ex-vivo confirming target engagement. Lastly, because the rodent nasal system is not similar to humans, we evaluated i.n. 2-PMPA also in a non-human primate. We report that i.n. 2-PMPA provides selective brain delivery with micromolar concentrations. These studies support intranasal delivery of 2-PMPA to deliver therapeutic concentrations in the brain and may facilitate its clinical development.
Collapse
Affiliation(s)
- Rana Rais
- Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Krystyna Wozniak
- Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Ying Wu
- Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Minae Niwa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Marigo Stathis
- Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Jesse Alt
- Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Marc Giroux
- Kurve Technology, Inc., Bothell, Washington, United States of America
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Camilo Rojas
- Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Barbara S. Slusher
- Brain Science Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
42
|
N -acetyl-aspartyl-glutamate and inhibition of glutamate carboxypeptidases protects against soman-induced neuropathology. Neurotoxicology 2015; 48:180-91. [DOI: 10.1016/j.neuro.2015.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/26/2015] [Accepted: 03/13/2015] [Indexed: 12/31/2022]
|
43
|
Gao Y, Xu S, Cui Z, Zhang M, Lin Y, Cai L, Wang Z, Luo X, Zheng Y, Wang Y, Luo Q, Jiang J, Neale JH, Zhong C. Mice lacking glutamate carboxypeptidase II develop normally, but are less susceptible to traumatic brain injury. J Neurochem 2015; 134:340-53. [PMID: 25872793 DOI: 10.1111/jnc.13123] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 04/04/2015] [Accepted: 04/08/2015] [Indexed: 11/26/2022]
Affiliation(s)
- Yang Gao
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Siyi Xu
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Zhenwen Cui
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Mingkun Zhang
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Yingying Lin
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Lei Cai
- Shanghai Research Center for Model Organisms; Shanghai China
| | - Zhugang Wang
- Shanghai Research Center for Model Organisms; Shanghai China
| | - Xingguang Luo
- Department of Psychiatry; Yale University School of Medicine; West Haven Connecticut USA
| | - Yan Zheng
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Yong Wang
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Qizhong Luo
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Jiyao Jiang
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| | - Joseph H. Neale
- Department of Biology; Georgetown University; Washington DC USA
| | - Chunlong Zhong
- Department of Neurosurgery; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University; Shanghai China
| |
Collapse
|
44
|
Zhong C, Luo Q, Jiang J. Blockade ofN-acetylaspartylglutamate peptidases: a novel protective strategy for brain injuries and neurological disorders. Int J Neurosci 2014; 124:867-73. [DOI: 10.3109/00207454.2014.890935] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
45
|
High-field proton magnetic resonance spectroscopy reveals metabolic effects of normal brain aging. Neurobiol Aging 2014; 35:1686-94. [PMID: 24559659 DOI: 10.1016/j.neurobiolaging.2014.01.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 12/19/2013] [Accepted: 01/19/2014] [Indexed: 11/24/2022]
Abstract
Altered brain metabolism is likely to be an important contributor to normal cognitive decline and brain pathology in elderly individuals. To characterize the metabolic changes associated with normal brain aging, we used high-field proton magnetic resonance spectroscopy in vivo to quantify 20 neurochemicals in the hippocampus and sensorimotor cortex of young adult and aged rats. We found significant differences in the neurochemical profile of the aged brain when compared with younger adults, including lower aspartate, ascorbate, glutamate, and macromolecules, and higher glucose, myo-inositol, N-acetylaspartylglutamate, total choline, and glutamine. These neurochemical biomarkers point to specific cellular mechanisms that are altered in brain aging, such as bioenergetics, oxidative stress, inflammation, cell membrane turnover, and endogenous neuroprotection. Proton magnetic resonance spectroscopy may be a valuable translational approach for studying mechanisms of brain aging and pathology, and for investigating treatments to preserve or enhance cognitive function in aging.
Collapse
|
46
|
Yuan Y, Pan S, Sun Z, Dan Q, Liu J. Brain-derived neurotrophic factor-modified umbilical cord mesenchymal stem cell transplantation improves neurological deficits in rats with traumatic brain injury. Int J Neurosci 2013; 124:524-31. [DOI: 10.3109/00207454.2013.859144] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
47
|
Alves JL. Blood–brain barrier and traumatic brain injury. J Neurosci Res 2013; 92:141-7. [DOI: 10.1002/jnr.23300] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 05/10/2013] [Accepted: 08/29/2013] [Indexed: 12/29/2022]
Affiliation(s)
- José Luís Alves
- Doctoral Programme in Health SciencesFaculdade de Medicina da Universidade de CoimbraCoimbra Portugal
| |
Collapse
|
48
|
Gurkoff GG, Feng JF, Van KC, Izadi A, Ghiasvand R, Shahlaie K, Song M, Lowe DA, Zhou J, Lyeth BG. NAAG peptidase inhibitor improves motor function and reduces cognitive dysfunction in a model of TBI with secondary hypoxia. Brain Res 2013; 1515:98-107. [PMID: 23562458 DOI: 10.1016/j.brainres.2013.03.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 03/22/2013] [Accepted: 03/27/2013] [Indexed: 10/27/2022]
Abstract
Immediately following traumatic brain injury (TBI) and TBI with hypoxia, there is a rapid and pathophysiological increase in extracellular glutamate, subsequent neuronal damage and ultimately diminished motor and cognitive function. N-acetyl-aspartyl glutamate (NAAG), a prevalent neuropeptide in the CNS, is co-released with glutamate, binds to the presynaptic group II metabotropic glutamate receptor subtype 3 (mGluR3) and suppresses glutamate release. However, the catalytic enzyme glutamate carboxypeptidase II (GCP II) rapidly hydrolyzes NAAG into NAA and glutamate. Inhibition of the GCP II enzyme with NAAG peptidase inhibitors reduces the concentration of glutamate both by increasing the duration of NAAG activity on mGluR3 and by reducing degradation into NAA and glutamate resulting in reduced cell death in models of TBI and TBI with hypoxia. In the following study, rats were administered the NAAG peptidase inhibitor PGI-02776 (10mg/kg) 30 min following TBI combined with a hypoxic second insult. Over the two weeks following injury, PGI-02776-treated rats had significantly improved motor function as measured by increased duration on the rota-rod and a trend toward improved performance on the beam walk. Furthermore, two weeks post-injury, PGI-02776-treated animals had a significant decrease in latency to find the target platform in the Morris water maze as compared to vehicle-treated animals. These findings demonstrate that the application of NAAG peptidase inhibitors can reduce the deleterious motor and cognitive effects of TBI combined with a second hypoxic insult in the weeks following injury.
Collapse
Affiliation(s)
- Gene G Gurkoff
- Department of Neurological Surgery, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Janczura KJ, Olszewski RT, Bzdega T, Bacich DJ, Heston WD, Neale JH. NAAG peptidase inhibitors and deletion of NAAG peptidase gene enhance memory in novel object recognition test. Eur J Pharmacol 2012. [PMID: 23200894 DOI: 10.1016/j.ejphar.2012.11.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The peptide neurotransmitter N-acetylaspartylglutamate (NAAG) is inactivated by the extracellular enzyme glutamate carboxypeptidase II. Inhibitors of this enzyme reverse dizocilpine (MK-801)-induced impairment of short-term memory in the novel object recognition test. The objective of this study was to test the hypothesis that NAAG peptidase inhibition enhances long-term (24h delay) memory of C57BL mice. These mice and mice in which glutamate carboxypeptidase II had been knocked out were presented with two identical objects to explore for 10min on day 1 and tested with one of these familiar objects and one novel object on day 2. Memory was assessed as the degree to which the mice recalled the familiar object and explored the novel object to a greater extent on day 2. Uninjected mice or mice injected with saline prior to the acquisition session on day 1 demonstrated a lack of memory of the acquisition experience by exploring the familiar and novel objects to the same extent on day 2. Mice treated with glutamate carboxypeptidase II inhibitors ZJ43 or 2-PMPA prior to the acquisition trial explored the novel object significantly more time than the familiar object on day 2. Consistent with these results, mice in which glutamate carboxypeptidase II had been knocked out distinguished the novel from the familiar object on day 2 while their heterozygous colony mates did not. Inhibition of glutamate carboxypeptidase II enhances recognition memory, a therapeutic action that might be useful in treatment of memory deficits related to age and neurological disorders.
Collapse
|
50
|
Yamada T, Zuo D, Yamamoto T, Olszewski RT, Bzdega T, Moffett JR, Neale JH. NAAG peptidase inhibition in the periaqueductal gray and rostral ventromedial medulla reduces flinching in the formalin model of inflammation. Mol Pain 2012; 8:67. [PMID: 22971334 PMCID: PMC3539905 DOI: 10.1186/1744-8069-8-67] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 05/21/2012] [Indexed: 11/28/2022] Open
Abstract
Background Metabotropic glutamate receptors (mGluRs) have been identified as significant analgesic targets. Systemic treatments with inhibitors of the enzymes that inactivate the peptide transmitter N-acetylaspartylglutamate (NAAG), an mGluR3 agonist, have an analgesia-like effect in rat models of inflammatory and neuropathic pain. The goal of this study was to begin defining locations within the central pain pathway at which NAAG activation of its receptor mediates this effect. Results NAAG immunoreactivity was found in neurons in two brain regions that mediate nociceptive processing, the periaqueductal gray (PAG) and the rostral ventromedial medulla (RVM). Microinjection of the NAAG peptidase inhibitor ZJ43 into the PAG contralateral, but not ipsilateral, to the formalin injected footpad reduced the rapid and slow phases of the nociceptive response in a dose-dependent manner. ZJ43 injected into the RVM also reduced the rapid and slow phase of the response. The group II mGluR antagonist LY341495 blocked these effects of ZJ43 on the PAG and RVM. NAAG peptidase inhibition in the PAG and RVM did not affect the thermal withdrawal response in the hot plate test. Footpad inflammation also induced a significant increase in glutamate release in the PAG. Systemic injection of ZJ43 increased NAAG levels in the PAG and RVM and blocked the inflammation-induced increase in glutamate release in the PAG. Conclusion These data demonstrate a behavioral and neurochemical role for NAAG in the PAG and RVM in regulating the spinal motor response to inflammation and that NAAG peptidase inhibition has potential as an approach to treating inflammatory pain via either the ascending (PAG) and/or the descending pain pathways (PAG and RVM) that warrants further study.
Collapse
Affiliation(s)
- Toshihiko Yamada
- Department of Biology, Georgetown University, Washington, DC, USA
| | | | | | | | | | | | | |
Collapse
|