1
|
Yuan L, Wang Y, Hu X, Zuo Y, Jin T, Li X, Li X, Cheng L, Zhang H, Zhang T. Time-of-Day-Dependent Effects of Rehabilitation on Motor Recovery After Experimental Focal Cerebral Ischemia. Stroke 2025. [PMID: 40276875 DOI: 10.1161/strokeaha.125.050084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Rehabilitation is an efficacious method to improve poststroke motor dysfunction. Various rehabilitative techniques have become popular in this field of research. However, it has not been reported whether better outcomes can be achieved if rehabilitation training is conducted at the optimal time of the day. METHODS A model of photothrombotic ischemic stroke was used in C57/BL6 mice, and poststroke 24-hour activity cycles were evaluated. We found an activity peak around Zeitgeber time (ZT)13 (21:00) and a trough around ZT20 (04:00) during the dark phase. In addition, we selected ZT6 (14:00) as the daytime training group (corresponding to the ZT13 training group, ZT20 training group, and ZT6 training group, respectively). The 3 groups underwent treadmill training for 4 weeks. Functional and histological recovery levels were compared among groups. In addition, bulk RNA sequencing analysis was used to explore the possible molecular mechanisms. RESULTS The results showed that stroke-induced mice maintained a regular nocturnal locomotor rhythm with reduced amplitude. Motor recovery was greater in the ZT13 training group than in the ZT6 group, with a trend toward better outcomes at ZT13 than ZT20. The ZT13 group also showed superior neuronal survival and neurogenesis compared with ZT6, while improvements between ZT13 and ZT20 were less pronounced. Bulk RNA sequencing suggested that synaptic plasticity, calcium signaling, cAMP signaling, and MAPK signaling pathways contributed to neural repair differences between ZT13 and ZT6. In addition, the results showed a similar pattern of motor recovery in female and aged mice trained at ZT13 compared with ZT6, reinforcing the benefits of time-of-day training. CONCLUSIONS In conclusion, rehabilitative training during the most active phase is beneficial for enhanced recovery outcomes. Our study suggests that rehabilitation should be conducted when the body is in an optimal physiological state-that is, a time-of-day-dependent rehabilitation strategy.
Collapse
Affiliation(s)
- Linran Yuan
- Qilu Hospital of Shandong University, Jinan, China (L.Y., T.Z.)
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (L.Y., T.Z.)
| | - Yunlei Wang
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
| | - Xueyan Hu
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China (X.H., Xudong Li, Xingzhu Li, L.C., T.Z.)
| | - Yao Zuo
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China (Y.Z.)
| | - Tianyu Jin
- Department of Rehabilitation Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, China (T.J.)
| | - Xudong Li
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China (X.H., Xudong Li, Xingzhu Li, L.C., T.Z.)
| | - Xingzhu Li
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China (X.H., Xudong Li, Xingzhu Li, L.C., T.Z.)
| | - Lingna Cheng
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China (X.H., Xudong Li, Xingzhu Li, L.C., T.Z.)
| | - Haojie Zhang
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
| | - Tong Zhang
- Qilu Hospital of Shandong University, Jinan, China (L.Y., T.Z.)
- China Rehabilitation Research Center, Beijing Bo'ai Hospital (L.Y., Y.W., X.H., Xudong Li, Xingzhu Li, L.C., H.Z., T.Z.)
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (L.Y., T.Z.)
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China (X.H., Xudong Li, Xingzhu Li, L.C., T.Z.)
| |
Collapse
|
2
|
Xu P, Jetmore HD, Chen R, Shen M. Enzyme-modified Pt nanoelectrodes for glutamate detection. Faraday Discuss 2025; 257:165-181. [PMID: 39465674 DOI: 10.1039/d4fd00138a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
We present here a glutamate oxidase (GluOx)-modified platinum (Pt) nanoelectrode with a planar geometry for glutamate detection. The Pt nanoelectrode was characterized using electrochemistry and scanning electron microscopy (SEM). The radius of the Pt nanoelectrode measured using SEM is ∼210 nm. GluOx-modified Pt nanoelectrodes were generated by dip coating GluOx on the Pt nanoelectrode in a solution of 0.9% (wt%) bovine serum albumin (BSA), 0.126% (wt%) glutaraldehyde, and 100 U mL-1 GluOx. An increase in current was observed at +0.7 V vs. Ag/AgCl/1 M KCl with adding increasing concentrations of glutamate. Two-sample t-test results showed that there is a significant difference for current at +0.7 V between the blank and the added lowest glutamate concentration, as well as between adjacent glutamate concentrations, confirming that the increase in current is related to the increased glutamate concentration. The experimental current-concentration curve of glutamate detection fitted well to the theoretical Michaelis-Menten curve. At the low concentration range (50 μM to 200 μM), a linear relationship between the current and glutamate concentration was observed. The Michaelis-Menten constants of Imax and Km were calculated to be 1.093 pA and 0.227 mM, respectively. Biosensor efficiency (the ratio of glutamate sensitivity to H2O2 sensitivity) is calculated to be 57.9%. Enzact (Imax/H2O2 sensitivity, an indicator of the amount of enzyme loaded on the electrode) of the GluOx-modified Pt nanoelectrode is 0.243 mM. We further compared the sensitivity of a GluOx-modified Pt nanoelectrode with a GluOx-modified carbon fiber microelectrode (7 μm diameter and a sensing length of ∼350 μm). Glutamate detection on the GluOx-modified carbon fiber microelectrode fitted well to a Michaelis-Menten like response. Based on the fitting, the GluOx-modified carbon fiber microelectrode exhibited an Imax of 0.689 nA and a Km of 301.2 μM towards glutamate detection. The best linear range of glutamate detection on the GluOx-modified carbon fiber microelectrode is from 50 μM to 150 μM glutamate. The GluOx-modified carbon fiber microelectrode exhibited a higher potential requirement for glutamate detection compared to the GluOx-modified Pt nanoelectrode.
Collapse
Affiliation(s)
- Peibo Xu
- Chan Zuckerberg Biohub Chicago, USA
| | - Henry David Jetmore
- Department of Chemistry, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA.
| | - Ran Chen
- Department of Chemistry, University of Illinois at Urbana Champaign, Urbana, IL, 61801, USA.
| | - Mei Shen
- Chan Zuckerberg Biohub Chicago, USA
| |
Collapse
|
3
|
Arizanovska D, Emodogo JA, Lally AP, Palavicino-Maggio CB, Liebl DJ, Folorunso OO. Cross species review of the physiological role of D-serine in translationally relevant behaviors. Amino Acids 2023; 55:1501-1517. [PMID: 37833512 PMCID: PMC10689556 DOI: 10.1007/s00726-023-03338-6] [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: 06/01/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023]
Abstract
Bridging the gap between preclinical models of neurological and psychiatric disorders with their human manifestations is necessary to understand their underlying mechanisms, identify biomarkers, and develop novel therapeutics. Cognitive and social impairments underlie multiple neuropsychiatric and neurological disorders and are often comorbid with sleep disturbances, which can exacerbate poor outcomes. Importantly, many symptoms are conserved between vertebrates and invertebrates, although they may have subtle differences. Therefore, it is essential to determine the molecular mechanisms underlying these behaviors across different species and their translatability to humans. Genome-wide association studies have indicated an association between glutamatergic gene variants and both the risk and frequency of psychiatric disorders such as schizophrenia, bipolar disorder, and autism spectrum disorder. For example, changes in glutamatergic neurotransmission, such as glutamate receptor subtype N-methyl-D-aspartate receptor (NMDAR) hypofunction, have been shown to contribute to the pathophysiology of schizophrenia. Furthermore, in neurological disorders, such as traumatic brain injury and Alzheimer's disease, hyperactivation of NMDARs leads to synaptic damage. In addition to glutamate binding, NMDARs require the binding of a co-agonist D-serine or glycine to the GluN1 subunit to open. D-serine, which is racemized from L-serine by the neuronal enzyme serine racemase (SRR), and both SRR and D-serine are enriched in cortico-limbic brain regions. D-serine is critical for complex behaviors, such as cognition and social behavior, where dysregulation of its synthesis and release has been implicated in many pathological conditions. In this review, we explore the role of D-serine in behaviors that are translationally relevant to multiple psychiatric and neurological disorders in different models across species.
Collapse
Affiliation(s)
- Dena Arizanovska
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jada A Emodogo
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA
| | - Anna P Lally
- Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA, USA
| | - Caroline B Palavicino-Maggio
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Neurobiological Mechanisms of Aggression Laboratory, McLean Hospital, Belmont, MA, USA
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oluwarotimi O Folorunso
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
4
|
Teneqexhi P, Khalid A, Nisbett KE, Job GA, Messer WS, Ragozzino ME. The Partial M 1 Muscarinic Cholinergic Receptor Agonist, CDD-0102A, Differentially Modulates Glutamate Efflux in Striatal Subregions during Stereotyped Motor Behavior in the BTBR Mouse Model of Autism. ACS Chem Neurosci 2023; 14:2699-2709. [PMID: 37434313 PMCID: PMC10401636 DOI: 10.1021/acschemneuro.3c00260] [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: 04/19/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023] Open
Abstract
The BTBR T+ Itpr3tf/J (BTBR) mouse displays elevated repetitive motor behaviors. Treatment with the partial M1 muscarinic receptor agonist, CDD-0102A, attenuates stereotyped motor behaviors in BTBR mice. The present experiment investigated whether CDD-0102A modifies changes in striatal glutamate concentrations during stereotyped motor behavior in BTBR and B6 mice. Using glutamate biosensors, change in striatal glutamate efflux was measured during bouts of digging and grooming behavior with a 1 s time resolution. Mice displayed both decreases and increases in glutamate efflux during such behaviors. Magnitude of changes in glutamate efflux (decreases and increases) from dorsomedial and dorsolateral striatum were significantly greater in BTBR mice compared to those of B6 mice. In BTBR mice, CDD-0102A (1.2 mg/kg) administered 30 min prior to testing significantly reduced the magnitude change in glutamate decreases and increases from the dorsolateral striatum and decreased grooming behavior. Conversely, CDD-0102A treatment in B6 mice potentiated glutamate decreases and increases in the dorsolateral striatum and elevated grooming behavior. The findings suggest that activation of M1 muscarinic receptors modifies glutamate transmission in the dorsolateral striatum and self-grooming behavior.
Collapse
Affiliation(s)
- Pamela Teneqexhi
- Department
of Psychology, University of Illinois Chicago, 1007 West Harrison Street, Chicago, Illinois 60607, United States
| | - Alina Khalid
- Department
of Psychology, University of Illinois Chicago, 1007 West Harrison Street, Chicago, Illinois 60607, United States
| | - Khalin E. Nisbett
- Department
of Psychology, University of Illinois Chicago, 1007 West Harrison Street, Chicago, Illinois 60607, United States
- Graduate
Program in Neuroscience, University of Illinois
Chicago, Chicago, Illinois 60607, United States
| | - Greeshma A. Job
- Department
of Psychology, University of Illinois Chicago, 1007 West Harrison Street, Chicago, Illinois 60607, United States
| | - William S. Messer
- Departments
of Pharmacology and Experimental Therapeutics, and Medicinal and Biological
Chemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Michael E. Ragozzino
- Department
of Psychology, University of Illinois Chicago, 1007 West Harrison Street, Chicago, Illinois 60607, United States
| |
Collapse
|
5
|
Downs AM, Plaxco KW. Real-Time, In Vivo Molecular Monitoring Using Electrochemical Aptamer Based Sensors: Opportunities and Challenges. ACS Sens 2022; 7:2823-2832. [PMID: 36205360 PMCID: PMC9840907 DOI: 10.1021/acssensors.2c01428] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The continuous, real-time measurement of specific molecules in situ in the body would greatly improve our ability to understand, diagnose, and treat disease. The vast majority of continuous molecular sensing technologies, however, either (1) rely on the chemical or enzymatic reactivity of their targets, sharply limiting their scope, or (2) have never been shown (and likely will never be shown) to operate in the complex environments found in vivo. Against this background, here we review electrochemical aptamer-based (EAB) sensors, an electrochemical approach to real-time molecular monitoring that has now seen 15 years of academic development. The strengths of the EAB platform are significant: to date it is the only molecular measurement technology that (1) functions independently of the chemical reactivity of its targets, and is thus general, and (2) supports in vivo measurements. Specifically, using EAB sensors we, and others, have already reported the real-time, seconds-resolved measurements of multiple, unrelated drugs and metabolites in situ in the veins and tissues of live animals. Against these strengths, we detail the platform's remaining weaknesses, which include still limited measurement duration (hours, rather than the more desirable days) and the difficulty in obtaining sufficiently high performance aptamers against new targets, before then detailing promising approaches overcoming these hurdles. Finally, we close by exploring the opportunities we believe this potentially revolutionary technology (as well as a few, possibly competing, technologies) will create for both researchers and clinicians.
Collapse
Affiliation(s)
- Alex M. Downs
- Sandia National Laboratories, Albuquerque, NM 87106, USA
| | - Kevin W. Plaxco
- Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA,Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA,Corresponding author:
| |
Collapse
|
6
|
Needham H, Torpey G, Flores CC, Davis CJ, Vanderheyden WM, Gerstner JR. A Dichotomous Role for FABP7 in Sleep and Alzheimer's Disease Pathogenesis: A Hypothesis. Front Neurosci 2022; 16:798994. [PMID: 35844236 PMCID: PMC9280343 DOI: 10.3389/fnins.2022.798994] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/10/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty acid binding proteins (FABPs) are a family of intracellular lipid chaperone proteins known to play critical roles in the regulation of fatty acid uptake and transport as well as gene expression. Brain-type fatty acid binding protein (FABP7) is enriched in astrocytes and has been implicated in sleep/wake regulation and neurodegenerative diseases; however, the precise mechanisms underlying the role of FABP7 in these biological processes remain unclear. FABP7 binds to both arachidonic acid (AA) and docosahexaenoic acid (DHA), resulting in discrete physiological responses. Here, we propose a dichotomous role for FABP7 in which ligand type determines the subcellular translocation of fatty acids, either promoting wakefulness aligned with Alzheimer's pathogenesis or promoting sleep with concomitant activation of anti-inflammatory pathways and neuroprotection. We hypothesize that FABP7-mediated translocation of AA to the endoplasmic reticulum of astrocytes increases astrogliosis, impedes glutamatergic uptake, and enhances wakefulness and inflammatory pathways via COX-2 dependent generation of pro-inflammatory prostaglandins. Conversely, we propose that FABP7-mediated translocation of DHA to the nucleus stabilizes astrocyte-neuron lactate shuttle dynamics, preserves glutamatergic uptake, and promotes sleep by activating anti-inflammatory pathways through the peroxisome proliferator-activated receptor-γ transcriptional cascade. Importantly, this model generates several testable hypotheses applicable to other neurodegenerative diseases, including amyotrophic lateral sclerosis and Parkinson's disease.
Collapse
Affiliation(s)
- Hope Needham
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Grace Torpey
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - William M. Vanderheyden
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
7
|
Kinlein SA, Wallace NK, Savenkova MI, Karatsoreos IN. Chronic hypothalamic-pituitary-adrenal axis disruption alters glutamate homeostasis and neural responses to stress in male C57Bl6/N mice. Neurobiol Stress 2022; 19:100466. [PMID: 35720261 PMCID: PMC9198473 DOI: 10.1016/j.ynstr.2022.100466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 11/24/2022] Open
Abstract
It is now well-established that stress elicits brain- and body-wide changes in physiology and has significant impacts on many aspects of health. The hypothalamic-pituitary-adrenal (HPA) axis is the major neuroendocrine system mediating the integrated response to stress. Appropriate engagement and termination of HPA activity enhances survival and optimizes physiological and behavioral responses to stress, while dysfunction of this system is linked to negative health outcomes such as depression, anxiety, and post-traumatic stress disorder. Glutamate signaling plays a large role in the transmission of stress-related information throughout the brain. Furthermore, aberrant glutamate signaling has negative consequences for neural plasticity and synaptic function and is linked to stress-related pathology. However, the connection between HPA dysfunction and glutamate signaling is not fully understood. We tested how HPA axis dysfunction (using low dose chronic corticosterone in the drinking water) affects glutamate homeostasis and neural responses under baseline and acute stress in male C57BL/6N mice. Using laser microdissection and transcriptomic analyses, we show that chronic disruption of the HPA axis alters the expression of genes related to glutamate signaling in the medial prefrontal cortex (mPFC), hippocampus, and amygdala. While neural responses to stress (as measured by FOS) in the hippocampus and amygdala were not affected in our model of HPA dysfunction, we observed an exaggerated response to stress in the mPFC. To further probe this we undertook in vivo biosensor measurements of the dynamics of extracellular glutamate responses to stress in the mPFC in real-time, and found glutamate dynamics in the mPFC were significantly altered by chronic HPA dysfunction. Together, these findings support the hypothesis that chronic HPA axis dysfunction alters glutamatergic signaling in regions known to regulate emotional behavior, providing more evidence linking HPA dysfunction and stress vulnerability.
Collapse
|
8
|
Huang K, Yang Q, Han Y, Zhang Y, Wang Z, Wang L, Wei P. An Easily Compatible Eye-tracking System for Freely-moving Small Animals. Neurosci Bull 2022; 38:661-676. [PMID: 35325370 PMCID: PMC9206064 DOI: 10.1007/s12264-022-00834-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
Measuring eye movement is a fundamental approach in cognitive science as it provides a variety of insightful parameters that reflect brain states such as visual attention and emotions. Combining eye-tracking with multimodal neural recordings or manipulation techniques is beneficial for understanding the neural substrates of cognitive function. Many commercially-available and custom-built systems have been widely applied to awake, head-fixed small animals. However, the existing eye-tracking systems used in freely-moving animals are still limited in terms of their compatibility with other devices and of the algorithm used to detect eye movements. Here, we report a novel system that integrates a general-purpose, easily compatible eye-tracking hardware with a robust eye feature-detection algorithm. With ultra-light hardware and a detachable design, the system allows for more implants to be added to the animal's exposed head and has a precise synchronization module to coordinate with other neural implants. Moreover, we systematically compared the performance of existing commonly-used pupil-detection approaches, and demonstrated that the proposed adaptive pupil feature-detection algorithm allows the analysis of more complex and dynamic eye-tracking data in free-moving animals. Synchronized eye-tracking and electroencephalogram recordings, as well as algorithm validation under five noise conditions, suggested that our system is flexibly adaptable and can be combined with a wide range of neural manipulation and recording technologies.
Collapse
Affiliation(s)
- Kang Huang
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qin Yang
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yaning Han
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yulin Zhang
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhiyi Wang
- Harbin Institute of Technology Shenzhen, Shenzhen, 518055, China
| | - Liping Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengfei Wei
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
9
|
Nakajima T, Takeda S, Ito Y, Oyama A, Takami Y, Takeya Y, Yamamoto K, Sugimoto K, Shimizu H, Shimamura M, Rakugi H, Morishita R. A novel chronic dural port platform for continuous collection of cerebrospinal fluid and intrathecal drug delivery in free-moving mice. Fluids Barriers CNS 2022; 19:31. [PMID: 35505336 PMCID: PMC9066940 DOI: 10.1186/s12987-022-00331-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/19/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cerebrospinal fluid (CSF) provides a close representation of pathophysiological changes occurring in the central nervous system (CNS); therefore, it has been employed in pathogenesis research and biomarker development for CNS disorders. CSF obtained from valid mouse models relevant to CNS disorders can be an important resource for successful biomarker and drug development. However, the limited volume of CSF that can be collected from tiny intrathecal spaces and the technical difficulties involved in CSF sampling has been a bottleneck that has hindered the detailed analysis of CSF in mouse models. METHODS We developed a novel chronic dural port (CDP) method without cannulation for CSF collection of mice. This method enables easy and repeated access to the intrathecal space in a free-moving, unanesthetized mouse, thereby enabling continuous long-term CSF collection with minimal tissue damage and providing a large volume of high-quality CSF from a single mouse. When combined with chemical biosensors, the CDP method allows for real-time monitoring of the dynamic changes in neurochemicals in the CSF at a one-second temporal resolution in free-moving mice. Moreover, the CDP can serve as a direct access point for the intrathecal injection of CSF tracers and drugs. RESULTS We established a CDP implantation and continuous CSF collection protocol. The CSF collected using CDP was not contaminated with blood and maintained physiological concentrations of basic electrolytes and proteins. The CDP method did not affect mouse's physiological behavior or induce tissue damage, thereby enabling a stable CSF collection for up to four weeks. The spatio-temporal distribution of CSF tracers delivered using CDP revealed that CSF metabolism in different brain areas is dynamic. The direct intrathecal delivery of centrally acting drugs using CDP enabled real-time behavioral assessments in free-moving mice. CONCLUSIONS The CDP method enables the collection of a large volume of high-quality CSF and direct intrathecal drug administration with real-time behavioral assessment in free-moving mice. Combined with animal models relevant to CNS disorders, this method provides a unique and valuable platform for biomarker and therapeutic drug research.
Collapse
Affiliation(s)
- Tsuneo Nakajima
- grid.136593.b0000 0004 0373 3971Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan
| | - Shuko Takeda
- grid.136593.b0000 0004 0373 3971Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan ,Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, Osaka 573- 0022 Japan
| | - Yuki Ito
- grid.136593.b0000 0004 0373 3971Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan ,Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, Osaka 573- 0022 Japan
| | - Akane Oyama
- grid.136593.b0000 0004 0373 3971Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan ,Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, Osaka 573- 0022 Japan
| | - Yoichi Takami
- grid.136593.b0000 0004 0373 3971Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan
| | - Yasushi Takeya
- grid.136593.b0000 0004 0373 3971Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Department of Clinical Nursing Division of Health Sciences Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan
| | - Koichi Yamamoto
- grid.136593.b0000 0004 0373 3971Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan
| | - Ken Sugimoto
- grid.136593.b0000 0004 0373 3971Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan ,grid.415086.e0000 0001 1014 2000General and Geriatric Medicine, Kawasaki Medical School General Medical Center, Okayama, 700-8505 Japan
| | - Hideo Shimizu
- grid.136593.b0000 0004 0373 3971Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan ,grid.412378.b0000 0001 1088 0812Department of Internal Medicine, Osaka Dental University, Hirakata, Osaka 573-1121 Japan
| | - Munehisa Shimamura
- grid.136593.b0000 0004 0373 3971Department of Neurology, Department of Health Development and Medicine, Osaka University, Suita, Osaka 565-0871 Japan
| | - Hiromi Rakugi
- grid.136593.b0000 0004 0373 3971Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan
| | - Ryuichi Morishita
- grid.136593.b0000 0004 0373 3971Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871 Japan
| |
Collapse
|
10
|
DiNuzzo M, Mangia S, Moraschi M, Mascali D, Hagberg GE, Giove F. Perception is associated with the brain's metabolic response to sensory stimulation. eLife 2022; 11:71016. [PMID: 35225790 PMCID: PMC9038191 DOI: 10.7554/elife.71016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Processing of incoming sensory stimulation triggers an increase of cerebral perfusion and blood oxygenation (neurovascular response) as well as an alteration of the metabolic neurochemical profile (neurometabolic response). Here we show in human primary visual cortex (V1) that perceived and unperceived isoluminant chromatic flickering stimuli designed to have similar neurovascular responses as measured by blood oxygenation level dependent functional MRI (BOLD-fMRI) have markedly different neurometabolic responses as measured by functional MRS. In particular, a significant regional buildup of lactate, an index of aerobic glycolysis, and glutamate, an index of malate-aspartate shuttle, occurred in V1 only when the flickering was perceived, without any relation with behavioral or physiological variables. Whereas the BOLD-fMRI signal in V1, a proxy for input to V1, was insensitive to flickering perception by design, the BOLD-fMRI signal in secondary visual areas was larger during perceived than unperceived flickering, indicating increased output from V1. These results demonstrate that the upregulation of energy metabolism induced by visual stimulation depends on the type of information processing taking place in V1, and that 1H-fMRS provides unique information about local input/output balance that is not measured by BOLD fMRI.
Collapse
Affiliation(s)
- Mauro DiNuzzo
- Museo Storico della Fisica e Centro Studi e Ricerche Enrico Fermi, Rome, Italy
| | - Silvia Mangia
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, United States
| | - Marta Moraschi
- Department of Radiation Oncology, University of Rome, Rome, Italy
| | - Daniele Mascali
- Dipartimento di Neuroscienze, Università Gabriele D'Annunzio, Chieti, Italy
| | - Gisela E Hagberg
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics and Biomedical Magnetic Resonance, Tübingen, Germany
| | - Federico Giove
- Museo Storico della Fisica e Centro Studi e Ricerche Enrico Fermi, Rome, Italy
| |
Collapse
|
11
|
Wallace NK, Pollard F, Savenkova M, Karatsoreos IN. Effect of Aging on Daily Rhythms of Lactate Metabolism in the Medial Prefrontal Cortex of Male Mice. Neuroscience 2020; 448:300-310. [PMID: 32717298 DOI: 10.1016/j.neuroscience.2020.07.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 07/11/2020] [Accepted: 07/18/2020] [Indexed: 01/24/2023]
Abstract
Aging is associated with reduced amplitude and earlier timing of circadian (daily) rhythms in sleep, brain function, and behavior. We examined whether age-related circadian dysfunction extends to the metabolic function of the brain, particularly in the prefrontal cortex (PFC). Using enzymatic amperometric biosensors, we recorded lactate concentration changes in the PFC in Young (7 mos) and Aged (19 mos) freely-behaving C57BL/6N male mice. Both Young and Aged mice displayed diurnal and circadian rhythms of lactate, with the Aged rhythm slightly phase advanced. Under constant conditions, the Aged rhythm showed a reduced amplitude not seen in the Young mice. We simultaneously observed a relationship between arousal state and PFC lactate rhythm via electroencephalography, which was modified by aging. Finally, using RT-qPCR, we found that aging affects the daily expression pattern of Glucose Transporter 1 (GLUT-1).
Collapse
Affiliation(s)
- Naomi K Wallace
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Felicity Pollard
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Marina Savenkova
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Ilia N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
12
|
Baer AG, Bourdon AK, Price JM, Campagna SR, Jacobson DA, Baghdoyan HA, Lydic R. Isoflurane anesthesia disrupts the cortical metabolome. J Neurophysiol 2020; 124:2012-2021. [PMID: 33112692 PMCID: PMC7814899 DOI: 10.1152/jn.00375.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Identifying similarities and differences in the brain metabolome during different states of consciousness has broad relevance for neuroscience and state-dependent autonomic function. This study focused on the prefrontal cortex (PFC) as a brain region known to modulate states of consciousness. Anesthesia was used as a tool to eliminate wakefulness. Untargeted metabolomic analyses were performed on microdialysis samples obtained from mouse PFC during wakefulness and during isoflurane anesthesia. Analyses detected 2,153 molecules, 91 of which could be identified. Analytes were grouped as detected during both wakefulness and anesthesia (n = 61) and as unique to wakefulness (n = 23) or anesthesia (n = 7). Data were analyzed using univariate and multivariate approaches. Relative to wakefulness, during anesthesia there was a significant (q < 0.0001) fourfold change in 21 metabolites. During anesthesia 11 of these 21 molecules decreased and 10 increased. The Kyoto Encyclopedia of Genes and Genomes database was used to relate behavioral state-specific changes in the metabolome to metabolic pathways. Relative to wakefulness, most of the amino acids and analogs measured were significantly decreased during isoflurane anesthesia. Nucleosides and analogs were significantly increased during anesthesia. Molecules associated with carbohydrate metabolism, maintenance of lipid membranes, and normal cell functions were significantly decreased during anesthesia. Significant state-specific changes were also discovered among molecules comprising lipids and fatty acids, monosaccharides, and organic acids. Considered together, these molecules regulate point-to-point transmission, volume conduction, and cellular metabolism. The results identify a novel ensemble of candidate molecules in PFC as putative modulators of wakefulness and the loss of wakefulness. NEW & NOTEWORTHY The loss of wakefulness caused by a single concentration of isoflurane significantly altered levels of interrelated metabolites in the prefrontal cortex. The results support the interpretation that states of consciousness reflect dynamic interactions among cortical neuronal networks involving a humbling number of molecules that comprise the brain metabolome.
Collapse
Affiliation(s)
- Aaron G Baer
- Department of Anesthesiology, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Allen K Bourdon
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee
| | - Joshua M Price
- Office of Information Technology, University of Tennessee, Knoxville, Tennessee
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee.,Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, Tennessee
| | - Daniel A Jacobson
- Department of Psychology, University of Tennessee, Knoxville, Tennessee.,Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Helen A Baghdoyan
- Department of Anesthesiology, University of Tennessee Medical Center, Knoxville, Tennessee.,Department of Psychology, University of Tennessee, Knoxville, Tennessee.,Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Ralph Lydic
- Department of Anesthesiology, University of Tennessee Medical Center, Knoxville, Tennessee.,Department of Psychology, University of Tennessee, Knoxville, Tennessee.,Oak Ridge National Laboratory, Oak Ridge, Tennessee
| |
Collapse
|
13
|
An active site mutation in 6-hydroxy-l-Nicotine oxidase from Arthrobacter nicotinovorans changes the substrate specificity in favor of (S)-nicotine. Arch Biochem Biophys 2020; 692:108520. [DOI: 10.1016/j.abb.2020.108520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 07/17/2020] [Accepted: 07/27/2020] [Indexed: 11/20/2022]
|
14
|
Ou Y, Buchanan AM, Witt CE, Hashemi P. Frontiers in Electrochemical Sensors for Neurotransmitter Detection: Towards Measuring Neurotransmitters as Chemical Diagnostics for Brain Disorders. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2019; 11:2738-2755. [PMID: 32724337 PMCID: PMC7386554 DOI: 10.1039/c9ay00055k] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
It is extremely challenging to chemically diagnose disorders of the brain. There is hence great interest in designing and optimizing tools for direct detection of chemical biomarkers implicated in neurological disorders to improve diagnosis and treatment. Tools that are capable of monitoring brain chemicals, neurotransmitters in particular, need to be biocompatible, perform with high spatiotemporal resolution, and ensure high selectivity and sensitivity. Recent advances in electrochemical methods are addressing these criteria; the resulting devices demonstrate great promise for in vivo neurotransmitter detection. None of these devices are currently used for diagnostic purposes, however these cutting-edge technologies are promising more sensitive, selective, faster, and less invasive measurements. Via this review we highlight significant technical advances and in vivo studies, performed in the last 5 years, that we believe will facilitate the development of diagnostic tools for brain disorders.
Collapse
Affiliation(s)
- Yangguang Ou
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia SC
| | - Anna Marie Buchanan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia SC
| | - Colby E. Witt
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia SC
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia SC
| |
Collapse
|
15
|
Bellesi M, Haswell JD, de Vivo L, Marshall W, Roseboom PH, Tononi G, Cirelli C. Myelin modifications after chronic sleep loss in adolescent mice. Sleep 2019; 41:4850494. [PMID: 29741724 DOI: 10.1093/sleep/zsy034] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Indexed: 01/28/2023] Open
Abstract
Study Objectives Previous studies found that sleep loss can suppress the expression of genes implicated in myelination and can have adverse effects on oligodendrocyte precursor cells. On the other hand, sleep may favor myelination by promoting the expression of genes involved in its formation and maintenance. Albeit limited, these results suggest that sleep loss can have detrimental effects on the formation and maintenance of myelin. Methods Here, we tested this hypothesis by evaluating ultrastructural modifications of myelin in two brain regions (corpus callosum and lateral olfactory tract) of mice exposed to different periods of sleep loss, from a few hours of sleep deprivation to ~5 days of chronic sleep restriction. In addition, we measured the internodal length-the distance between consecutive nodes of Ranvier along the axon-and plasma corticosterone levels. Results We find that g-ratio-the ratio of the diameter of the axon itself to the outer diameter of the myelinated fiber-increases after chronic sleep loss. This effect is mediated by a reduction in myelin thickness and is not associated with changes in the internodal length. Relative to sleep, plasma corticosterone levels increase after acute sleep deprivation, but show only a trend to increase after chronic sleep loss. Conclusions Chronic sleep loss may negatively affect myelin.
Collapse
Affiliation(s)
- Michele Bellesi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | | | - Luisa de Vivo
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - William Marshall
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | | | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
16
|
Piao CS, Holloway AL, Hong-Routson S, Wainwright MS. Depression following traumatic brain injury in mice is associated with down-regulation of hippocampal astrocyte glutamate transporters by thrombin. J Cereb Blood Flow Metab 2019; 39:58-73. [PMID: 29135354 PMCID: PMC6311670 DOI: 10.1177/0271678x17742792] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Depression after traumatic brain injury (TBI) is common but the mechanisms by which TBI causes depression are unknown. TBI decreases glutamate transporters GLT-1 and GLAST and allows extravasation of thrombin. We examined the effects of thrombin on transporter expression in primary hippocampal astrocytes. Application of a PAR-1 agonist caused down-regulation of GLT-1, which was prevented by inhibition of Rho kinase (ROCK). To confirm these mechanisms in vivo, we subjected mice to closed-skull TBI. Thrombin activity in the hippocampus increased one day following TBI. Seven days following TBI, expression of GLT-1 and GLAST was reduced in the hippocampus, and this was prevented by administration of the PAR-1 antagonist SCH79797. Inhibition of ROCK attenuated the decrease in GLT-1, but not GLAST, after TBI. We measured changes in glutamate levels in the hippocampus seven days after TBI using an implanted biosensor. Stress-induced glutamate levels were significantly increased following TBI and this was attenuated by treatment with the ROCK inhibitor fasudil. We quantified depressive behavior following TBI and found that inhibition of PAR-1 or ROCK decreased these behaviors. These results identify a novel mechanism by which TBI results in down-regulation of astrocyte glutamate transporters and implicate astrocyte and glutamate transporter dysfunction in depression following TBI.
Collapse
Affiliation(s)
- Chun-Shu Piao
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ashley L Holloway
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sue Hong-Routson
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,3 Division of Critical Care, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mark S Wainwright
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,3 Division of Critical Care, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
17
|
Arroyo-Currás N, Ortega G, Copp DA, Ploense KL, Plaxco ZA, Kippin TE, Hespanha JP, Plaxco KW. High-Precision Control of Plasma Drug Levels Using Feedback-Controlled Dosing. ACS Pharmacol Transl Sci 2018; 1:110-118. [PMID: 32219207 PMCID: PMC7088981 DOI: 10.1021/acsptsci.8b00033] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Indexed: 12/30/2022]
Abstract
By, in effect, rendering pharmacokinetics an experimentally adjustable parameter, the ability to perform feedback-controlled dosing informed by high-frequency in vivo drug measurements would prove a powerful tool for both pharmacological research and clinical practice. Efforts to this end, however, have historically been thwarted by an inability to measure in vivo drug levels in real time and with sufficient convenience and temporal resolution. In response, we describe a closed-loop, feedback-controlled delivery system that uses drug level measurements provided by an in vivo electrochemical aptamer-based (E-AB) sensor to adjust dosing rates every 7 s. The resulting system supports the maintenance of either constant or predefined time-varying plasma drug concentration profiles in live rats over many hours. For researchers, the resultant high-precision control over drug plasma concentrations provides an unprecedented opportunity to (1) map the relationships between pharmacokinetics and clinical outcomes, (2) eliminate inter- and intrasubject metabolic variation as a confounding experimental variable, (3) accurately simulate human pharmacokinetics in animal models, and (4) measure minute-to-minute changes in a drug's pharmacokinetic behavior in response to changing health status, diet, drug-drug interactions, or other intrinsic and external factors. In the clinic, feedback-controlled drug delivery would improve our ability to accurately maintain therapeutic drug levels in the face of large, often unpredictable intra- and interpatient metabolic variation. This, in turn, would improve the efficacy and safety of therapeutic intervention, particularly for the most gravely ill patients, for whom metabolic variability is highest and the margin for therapeutic error is smallest.
Collapse
Affiliation(s)
- Netzahualcóyotl Arroyo-Currás
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States,E-mail: . Tel.: (410) 955-3569
| | - Gabriel Ortega
- ‡Department of Chemistry and Biochemistry, §Center for Bioengineering, ⊥Center for Control,
Dynamical Systems, and Computation, #Department of Psychological and Brain Sciences, and ∇The Neuroscience
Research Institute and Department of Molecular, Cellular, and Developmental
Biology, University of California Santa
Barbara, Santa
Barbara, California 93106, United States,CIC
bioGUNE, Bizkaia Technology Park, Ed. 801A, 48160, Derio, Spain
| | - David A. Copp
- ‡Department of Chemistry and Biochemistry, §Center for Bioengineering, ⊥Center for Control,
Dynamical Systems, and Computation, #Department of Psychological and Brain Sciences, and ∇The Neuroscience
Research Institute and Department of Molecular, Cellular, and Developmental
Biology, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| | - Kyle L. Ploense
- ‡Department of Chemistry and Biochemistry, §Center for Bioengineering, ⊥Center for Control,
Dynamical Systems, and Computation, #Department of Psychological and Brain Sciences, and ∇The Neuroscience
Research Institute and Department of Molecular, Cellular, and Developmental
Biology, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| | - Zoe A. Plaxco
- ‡Department of Chemistry and Biochemistry, §Center for Bioengineering, ⊥Center for Control,
Dynamical Systems, and Computation, #Department of Psychological and Brain Sciences, and ∇The Neuroscience
Research Institute and Department of Molecular, Cellular, and Developmental
Biology, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| | - Tod E. Kippin
- ‡Department of Chemistry and Biochemistry, §Center for Bioengineering, ⊥Center for Control,
Dynamical Systems, and Computation, #Department of Psychological and Brain Sciences, and ∇The Neuroscience
Research Institute and Department of Molecular, Cellular, and Developmental
Biology, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| | - João P. Hespanha
- ‡Department of Chemistry and Biochemistry, §Center for Bioengineering, ⊥Center for Control,
Dynamical Systems, and Computation, #Department of Psychological and Brain Sciences, and ∇The Neuroscience
Research Institute and Department of Molecular, Cellular, and Developmental
Biology, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| | - Kevin W. Plaxco
- ‡Department of Chemistry and Biochemistry, §Center for Bioengineering, ⊥Center for Control,
Dynamical Systems, and Computation, #Department of Psychological and Brain Sciences, and ∇The Neuroscience
Research Institute and Department of Molecular, Cellular, and Developmental
Biology, University of California Santa
Barbara, Santa
Barbara, California 93106, United States,E-mail: . Tel.: (805) 893-5558
| |
Collapse
|
18
|
Ryu IS, Kim J, Seo SY, Yang JH, Oh JH, Lee DK, Cho HW, Lee K, Yoon SS, Seo JW, Shim I, Choe ES. Repeated Administration of Cigarette Smoke Condensate Increases Glutamate Levels and Behavioral Sensitization. Front Behav Neurosci 2018; 12:47. [PMID: 29615877 PMCID: PMC5864865 DOI: 10.3389/fnbeh.2018.00047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/28/2018] [Indexed: 01/09/2023] Open
Abstract
Nicotine, a nicotinic acetylcholine receptor agonist, produces the reinforcing effects of tobacco dependence by potentiating dopaminergic and glutamatergic neurotransmission. Non-nicotine alkaloids in tobacco also contribute to dependence by activating the cholinergic system. However, glutamatergic neurotransmission in the dorsal striatum associated with behavioral changes in response to cigarette smoking has not been investigated. In this study, the authors investigated alterations in glutamate levels in the rat dorsal striatum related to behavioral alterations after repeated administration of cigarette smoke condensate (CSC) using the real-time glutamate biosensing and an open-field behavioral assessment. Repeated administration of CSC including 0.4 mg nicotine (1.0 mL/kg/day, subcutaneous) for 14 days significantly increased extracellular glutamate concentrations more than repeated nicotine administration. In parallel with the hyperactivation of glutamate levels, repeated administration of CSC-evoked prolonged hypersensitization of psychomotor activity, including locomotor and rearing activities. These findings suggest that the CSC-induced psychomotor activities are closely associated with the elevation of glutamate concentrations in the rat dorsal striatum.
Collapse
Affiliation(s)
- In Soo Ryu
- Department of Biological Sciences, Pusan National University, Busan, South Korea.,Research Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Jieun Kim
- Department of Biological Sciences, Pusan National University, Busan, South Korea
| | - Su Yeon Seo
- Department of Biological Sciences, Pusan National University, Busan, South Korea.,Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Ju Hwan Yang
- Department of Biological Sciences, Pusan National University, Busan, South Korea
| | - Jeong Hwan Oh
- College of Fisheries Sciences, National Institute of Fisheries (NIFS), Busan, South Korea
| | - Dong Kun Lee
- Department of Physiology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Hyun-Wook Cho
- Department of Biology, Sunchon National University, Sunchon, South Korea
| | - Kyuhong Lee
- Inhalation Toxicology Research Center, Korea Institute of Toxicology, Jeongeup, South Korea
| | - Seong Shoon Yoon
- Research Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Joung-Wook Seo
- Research Center for Safety Pharmacology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Insop Shim
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Eun Sang Choe
- Department of Biological Sciences, Pusan National University, Busan, South Korea
| |
Collapse
|
19
|
Yeon C, Kim D, Kim K, Chung E. Visual Evoked Potential Recordings in Mice Using a Dry Non-invasive Multi-channel Scalp EEG Sensor. J Vis Exp 2018. [PMID: 29364268 DOI: 10.3791/56927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
For scalp EEG research environments with laboratory mice, we designed a dry-type 16 channel EEG sensor which is non-invasive, deformable, and re-usable because of the plunger-spring-barrel structural facet and mechanical strengths resulting from metal materials. The whole process for acquiring the VEP responses in vivo from a mouse consists of four steps: (1) sensor assembly, (2) animal preparation, (3) VEP measurement, and (4) signal processing. This paper presents representative measurements of VEP responses from multiple mice with a submicro-voltage signal resolution and sub-hundred millisecond temporal resolution. Although the proposed method is safer and more convenient compared to other previously reported animal EEG acquiring methods, there are remaining issues including how to enhance the signal-to-noise ratio and how to apply this technique with freely moving animals. The proposed method utilizes easily available resources and shows a repetitive VEP response with a satisfactory signal quality. Therefore, this method could be utilized for longitudinal experimental studies and reliable translational research exploiting non-invasive paradigms.
Collapse
Affiliation(s)
- Chanmi Yeon
- Department of Biomedical Science and Engineering (BMSE), Gwangju Institute of Science and Technology (GIST)
| | - Donghyeon Kim
- School of Electrical Engineering and Computer Science (EECS), Gwangju Institute of Science and Technology (GIST)
| | - Kiseon Kim
- School of Electrical Engineering and Computer Science (EECS), Gwangju Institute of Science and Technology (GIST)
| | - Euiheon Chung
- Department of Biomedical Science and Engineering (BMSE), Gwangju Institute of Science and Technology (GIST); School of Mechanical Engineering (SME), Gwangju Institute of Science and Technology (GIST);
| |
Collapse
|
20
|
Behavioral changes after nicotine challenge are associated with α7 nicotinic acetylcholine receptor-stimulated glutamate release in the rat dorsal striatum. Sci Rep 2017; 7:15009. [PMID: 29118361 PMCID: PMC5678080 DOI: 10.1038/s41598-017-15161-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/23/2017] [Indexed: 01/28/2023] Open
Abstract
Neurochemical alterations associated with behavioral responses induced by re-exposure to nicotine have not been sufficiently characterized in the dorsal striatum. Herein, we report on changes in glutamate concentrations in the rat dorsal striatum associated with behavioral alterations after nicotine challenge. Nicotine challenge (0.4 mg/kg/day, subcutaneous) significantly increased extracellular glutamate concentrations up to the level observed with repeated nicotine administration. This increase occurred in parallel with an increase in behavioral changes in locomotor and rearing activities. In contrast, acute nicotine administration and nicotine withdrawal on days 1 and 6 did not alter glutamate levels or behavioral changes. Blockade of α7 nicotinic acetylcholine receptors (nAChRs) significantly decreased the nicotine challenge-induced increases in extracellular glutamate concentrations and locomotor and rearing activities. These findings suggest that behavioral changes in locomotor and rearing activities after re-exposure to nicotine are closely associated with hyperactivation of the glutamate response by stimulating α7 nAChRs in the rat dorsal striatum.
Collapse
|
21
|
DiNuzzo M, Nedergaard M. Brain energetics during the sleep-wake cycle. Curr Opin Neurobiol 2017; 47:65-72. [PMID: 29024871 PMCID: PMC5732842 DOI: 10.1016/j.conb.2017.09.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/06/2017] [Accepted: 09/16/2017] [Indexed: 12/11/2022]
Abstract
Brain activity during wakefulness is associated with high metabolic rates that are believed to support information processing and memory encoding. In spite of loss of consciousness, sleep still carries a substantial energy cost. Experimental evidence supports a cerebral metabolic shift taking place during sleep that suppresses aerobic glycolysis, a hallmark of environment-oriented waking behavior and synaptic plasticity. Recent studies reveal that glial astrocytes respond to the reduction of wake-promoting neuromodulators by regulating volume, composition and glymphatic drainage of interstitial fluid. These events are accompanied by changes in neuronal discharge patterns, astrocyte-neuron interactions, synaptic transactions and underlying metabolic features. Internally-generated neuronal activity and network homeostasis are proposed to account for the high sleep-related energy demand.
Collapse
Affiliation(s)
- Mauro DiNuzzo
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Maiken Nedergaard
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY 14640, USA
| |
Collapse
|
22
|
Nikonova EV, Gilliland JDA, Tanis KQ, Podtelezhnikov AA, Rigby AM, Galante RJ, Finney EM, Stone DJ, Renger JJ, Pack AI, Winrow CJ. Transcriptional Profiling of Cholinergic Neurons From Basal Forebrain Identifies Changes in Expression of Genes Between Sleep and Wake. Sleep 2017; 40:3608773. [PMID: 28419375 PMCID: PMC6075396 DOI: 10.1093/sleep/zsx059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Study objective To assess differences in gene expression in cholinergic basal forebrain cells between sleeping and sleep-deprived mice sacrificed at the same time of day. Methods Tg(ChAT-eGFP)86Gsat mice expressing enhanced green fluorescent protein (eGFP) under control of the choline acetyltransferase (Chat) promoter were utilized to guide laser capture of cholinergic cells in basal forebrain. Messenger RNA expression levels in these cells were profiled using microarrays. Gene expression in eGFP(+) neurons was compared (1) to that in eGFP(-) neurons and to adjacent white matter, (2) between 7:00 am (lights on) and 7:00 pm (lights off), (3) between sleep-deprived and sleeping animals at 0, 3, 6, and 9 hours from lights on. Results There was a marked enrichment of ChAT and other markers of cholinergic neurons in eGFP(+) cells. Comparison of gene expression in these eGFP(+) neurons between 7:00 am and 7:00 pm revealed expected differences in the expression of clock genes (Arntl2, Per1, Per2, Dbp, Nr1d1) as well as mGluR3. Comparison of expression between spontaneous sleep and sleep-deprived groups sacrificed at the same time of day revealed a number of transcripts (n = 55) that had higher expression in sleep deprivation compared to sleep. Genes upregulated in sleep deprivation predominantly were from the protein folding pathway (25 transcripts, including chaperones). Among 42 transcripts upregulated in sleep was the cold-inducible RNA-binding protein. Conclusions Cholinergic cell signatures were characterized. Whether the identified genes are changing as a consequence of differences in behavioral state or as part of the molecular regulatory mechanism remains to be determined.
Collapse
Affiliation(s)
- Elena V Nikonova
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - Jason DA Gilliland
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, PA
| | - Keith Q Tanis
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - Alexei A Podtelezhnikov
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - Alison M Rigby
- Department of Neuroscience, Merck & Co., Inc., West Point, PA
| | - Raymond J Galante
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, PA
| | - Eva M Finney
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - David J Stone
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - John J Renger
- Department of Neuroscience, Merck & Co., Inc., West Point, PA
| | - Allan I Pack
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
23
|
Sandsmark DK, Elliott JE, Lim MM. Sleep-Wake Disturbances After Traumatic Brain Injury: Synthesis of Human and Animal Studies. Sleep 2017; 40:3074241. [PMID: 28329120 PMCID: PMC6251652 DOI: 10.1093/sleep/zsx044] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2017] [Indexed: 12/23/2022] Open
Abstract
Sleep-wake disturbances following traumatic brain injury (TBI) are increasingly recognized as a serious consequence following injury and as a barrier to recovery. Injury-induced sleep-wake disturbances can persist for years, often impairing quality of life. Recently, there has been a nearly exponential increase in the number of primary research articles published on the pathophysiology and mechanisms underlying sleep-wake disturbances after TBI, both in animal models and in humans, including in the pediatric population. In this review, we summarize over 200 articles on the topic, most of which were identified objectively using reproducible online search terms in PubMed. Although these studies differ in terms of methodology and detailed outcomes; overall, recent research describes a common phenotype of excessive daytime sleepiness, nighttime sleep fragmentation, insomnia, and electroencephalography spectral changes after TBI. Given the heterogeneity of the human disease phenotype, rigorous translation of animal models to the human condition is critical to our understanding of the mechanisms and of the temporal course of sleep-wake disturbances after injury. Arguably, this is most effectively accomplished when animal and human studies are performed by the same or collaborating research programs. Given the number of symptoms associated with TBI that are intimately related to, or directly stem from sleep dysfunction, sleep-wake disorders represent an important area in which mechanistic-based therapies may substantially impact recovery after TBI.
Collapse
Affiliation(s)
| | - Jonathan E Elliott
- VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Miranda M Lim
- VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR; Department of Behavioral Neuroscience, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR
| |
Collapse
|
24
|
Soto RJ, Hall JR, Brown MD, Taylor JB, Schoenfisch MH. In Vivo Chemical Sensors: Role of Biocompatibility on Performance and Utility. Anal Chem 2017; 89:276-299. [PMID: 28105839 PMCID: PMC6773264 DOI: 10.1021/acs.analchem.6b04251] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Robert J. Soto
- Department of Chemistry, University of North Carolina at Chapel Hill, CB 3290, Chapel Hill, NC 27599
| | - Jackson R. Hall
- Department of Chemistry, University of North Carolina at Chapel Hill, CB 3290, Chapel Hill, NC 27599
| | - Micah D. Brown
- Department of Chemistry, University of North Carolina at Chapel Hill, CB 3290, Chapel Hill, NC 27599
| | - James B. Taylor
- Department of Chemistry, University of North Carolina at Chapel Hill, CB 3290, Chapel Hill, NC 27599
| | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, CB 3290, Chapel Hill, NC 27599
| |
Collapse
|
25
|
Weltin A, Kieninger J, Urban GA. Microfabricated, amperometric, enzyme-based biosensors for in vivo applications. Anal Bioanal Chem 2016; 408:4503-21. [PMID: 26935934 PMCID: PMC4909808 DOI: 10.1007/s00216-016-9420-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/08/2016] [Accepted: 02/12/2016] [Indexed: 01/19/2023]
Abstract
Miniaturized electrochemical in vivo biosensors allow the measurement of fast extracellular dynamics of neurotransmitter and energy metabolism directly in the tissue. Enzyme-based amperometric biosensing is characterized by high specificity and precision as well as high spatial and temporal resolution. Aside from glucose monitoring, many systems have been introduced mainly for application in the central nervous system in animal models. We compare the microsensor principle with other methods applied in biomedical research to show advantages and drawbacks. Electrochemical sensor systems are easily miniaturized and fabricated by microtechnology processes. We review different microfabrication approaches for in vivo sensor platforms, ranging from simple modified wires and fibres to fully microfabricated systems on silicon, ceramic or polymer substrates. The various immobilization methods for the enzyme such as chemical cross-linking and entrapment in polymer membranes are discussed. The resulting sensor performance is compared in detail. We also examine different concepts to reject interfering substances by additional membranes, aspects of instrumentation and biocompatibility. Practical considerations are elaborated, and conclusions for future developments are presented. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Andreas Weltin
- Laboratory for Sensors, Department of Microsystems Engineering – IMTEK, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany
| | - Jochen Kieninger
- Laboratory for Sensors, Department of Microsystems Engineering – IMTEK, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany
| | - Gerald A. Urban
- Laboratory for Sensors, Department of Microsystems Engineering – IMTEK, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany
| |
Collapse
|
26
|
Ahnaou A, de Boer P, Lavreysen H, Huysmans H, Sinha V, Raeymaekers L, Van De Casteele T, Cid J, Van Nueten L, Macdonald G, Kemp J, Drinkenburg W. Translational neurophysiological markers for activity of the metabotropic glutamate receptor (mGluR2) modulator JNJ-40411813: Sleep EEG correlates in rodents and healthy men. Neuropharmacology 2016; 103:290-305. [DOI: 10.1016/j.neuropharm.2015.11.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/28/2015] [Accepted: 11/28/2015] [Indexed: 12/31/2022]
|
27
|
Li C, Limnuson K, Wu Z, Amin A, Narayan A, Golanov EV, Ahn CH, Hartings JA, Narayan RK. Single probe for real-time simultaneous monitoring of neurochemistry and direct-current electrocorticography. Biosens Bioelectron 2016; 77:62-8. [DOI: 10.1016/j.bios.2015.09.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 01/25/2023]
|
28
|
mGlu2 Receptor Agonism, but Not Positive Allosteric Modulation, Elicits Rapid Tolerance towards Their Primary Efficacy on Sleep Measures in Rats. PLoS One 2015; 10:e0144017. [PMID: 26658273 PMCID: PMC4684355 DOI: 10.1371/journal.pone.0144017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 11/12/2015] [Indexed: 12/17/2022] Open
Abstract
G-protein-coupled receptor (GPCR) agonists are known to induce both cellular adaptations resulting in tolerance to therapeutic effects and withdrawal symptoms upon treatment discontinuation. Glutamate neurotransmission is an integral part of sleep-wake mechanisms, which processes have translational relevance for central activity and target engagement. Here, we investigated the efficacy and tolerance potential of the metabotropic glutamate receptors (mGluR2/3) agonist LY354740 versus mGluR2 positive allosteric modulator (PAM) JNJ-42153605 on sleep-wake organisation in rats. In vitro, the selectivity and potency of JNJ-42153605 were characterized. In vivo, effects on sleep measures were investigated in rats after once daily oral repeated treatment for 7 days, withdrawal and consecutive re-administration of LY354740 (1–10 mg/kg) and JNJ-42153605 (3–30 mg/kg). JNJ-42153605 showed high affinity, potency and selectivity at mGluR2. Binding site analyses and knowledge-based docking confirmed the specificity of JNJ-42153605 at the mGluR2 allosteric binding site. Acute LY354740 and JNJ-42153605 dose-dependently decreased rapid eye movement (REM) sleep time and prolonged its onset latency. Sub chronic effects of LY354740 on REM sleep measures disappeared from day 3 onwards, whereas those of JNJ-42153605 were maintained after repeated exposure. LY354740 attenuated REM sleep homeostatic recovery, while this was preserved after JNJ-42153605 administration. JNJ-42153605 enhanced sleep continuity and efficiency, suggesting its potential as an add-on medication for impaired sleep quality during early stages of treatment. Abrupt cessation of JNJ-42153605 did not induce withdrawal phenomena and sleep disturbances, while the initial drug effect was fully reinstated after re-administration. Collectively, long-term treatment with JNJ-42153605 did not induce tolerance phenomena to its primary functional effects on sleep measures, nor adverse effects at withdrawal, while it promoted homeostatic recovery sleep. From the translational perspective, the present rodent findings suggest that mGluR2 positive allosteric modulation has therapeutic potential based on its superior long term efficacy over agonists in psychiatric disorders, particularly of those commonly occurring with REM sleep overdrive.
Collapse
|
29
|
Wang X, Pinto-Duarte A, Behrens MM, Zhou X, Sejnowski TJ. Characterization of spatio-temporal epidural event-related potentials for mouse models of psychiatric disorders. Sci Rep 2015; 5:14964. [PMID: 26459883 PMCID: PMC4602219 DOI: 10.1038/srep14964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/11/2015] [Indexed: 11/30/2022] Open
Abstract
Distinctive features in sensory event-related potentials (ERPs) are endophenotypic biomarkers of psychiatric disorders, widely studied using electroencephalographic (EEG) methods in humans and model animals. Despite the popularity and unique significance of the mouse as a model species in basic research, existing EEG methods applicable to mice are far less powerful than those available for humans and large animals. We developed a new method for multi-channel epidural ERP characterization in behaving mice with high precision, reliability and convenience and report an application to time-domain ERP feature characterization of the Sp4 hypomorphic mouse model for schizophrenia. Compared to previous methods, our spatio-temporal ERP measurement robustly improved the resolving power of key signatures characteristic of the disease model. The high performance and low cost of this technique makes it suitable for high-throughput behavioral and pharmacological studies.
Collapse
Affiliation(s)
- Xin Wang
- Howard Hughes Medical Institute and the Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - António Pinto-Duarte
- Howard Hughes Medical Institute and the Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - M Margarita Behrens
- Howard Hughes Medical Institute and the Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xianjin Zhou
- Department of Psychiatry, University of California at San Diego, La Jolla, CA 92093, USA
| | - Terrence J Sejnowski
- Howard Hughes Medical Institute and the Salk Institute for Biological Studies, La Jolla, CA 92037, USA.,Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
30
|
Pritchett D, Jagannath A, Brown LA, Tam SKE, Hasan S, Gatti S, Harrison PJ, Bannerman DM, Foster RG, Peirson SN. Deletion of Metabotropic Glutamate Receptors 2 and 3 (mGlu2 & mGlu3) in Mice Disrupts Sleep and Wheel-Running Activity, and Increases the Sensitivity of the Circadian System to Light. PLoS One 2015; 10:e0125523. [PMID: 25950516 PMCID: PMC4423919 DOI: 10.1371/journal.pone.0125523] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/14/2015] [Indexed: 12/22/2022] Open
Abstract
Sleep and/or circadian rhythm disruption (SCRD) is seen in up to 80% of schizophrenia patients. The co-morbidity of schizophrenia and SCRD may in part stem from dysfunction in common brain mechanisms, which include the glutamate system, and in particular, the group II metabotropic glutamate receptors mGlu2 and mGlu3 (encoded by the genes Grm2 and Grm3). These receptors are relevant to the pathophysiology and potential treatment of schizophrenia, and have also been implicated in sleep and circadian function. In the present study, we characterised the sleep and circadian rhythms of Grm2/3 double knockout (Grm2/3-/-) mice, to provide further evidence for the involvement of group II metabotropic glutamate receptors in the regulation of sleep and circadian rhythms. We report several novel findings. Firstly, Grm2/3-/- mice demonstrated a decrease in immobility-determined sleep time and an increase in immobility-determined sleep fragmentation. Secondly, Grm2/3-/- mice showed heightened sensitivity to the circadian effects of light, manifested as increased period lengthening in constant light, and greater phase delays in response to nocturnal light pulses. Greater light-induced phase delays were also exhibited by wildtype C57Bl/6J mice following administration of the mGlu2/3 negative allosteric modulator RO4432717. These results confirm the involvement of group II metabotropic glutamate receptors in photic entrainment and sleep regulation pathways. Finally, the diurnal wheel-running rhythms of Grm2/3-/- mice were perturbed under a standard light/dark cycle, but their diurnal rest-activity rhythms were unaltered in cages lacking running wheels, as determined with passive infrared motion detectors. Hence, when assessing the diurnal rest-activity rhythms of mice, the choice of assay can have a major bearing on the results obtained.
Collapse
Affiliation(s)
- David Pritchett
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Aarti Jagannath
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- F.Hoffman-La Roche, Neuroscience, Ophthalmology & Rare Diseases (NORD), Pharma Research & Early Development (pRED) Innovation Centre, Basel, Switzerland
| | - Laurence A. Brown
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Shu K. E. Tam
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Sibah Hasan
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| | - Silvia Gatti
- F.Hoffman-La Roche, Neuroscience, Ophthalmology & Rare Diseases (NORD), Pharma Research & Early Development (pRED) Innovation Centre, Basel, Switzerland
| | - Paul J. Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, United Kingdom
| | - David M. Bannerman
- Department of Experimental Psychology, University of Oxford, South Parks Road, Oxford, OX1 3UD, United Kingdom
| | - Russell G. Foster
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- * E-mail: (RGF); (SNP)
| | - Stuart N. Peirson
- Nuffield Department of Clinical Neurosciences (Nuffield Laboratory of Ophthalmology), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- * E-mail: (RGF); (SNP)
| |
Collapse
|
31
|
Özel RE, Hayat A, Andreescu S. RECENT DEVELOPMENTS IN ELECTROCHEMICAL SENSORS FOR THE DETECTION OF NEUROTRANSMITTERS FOR APPLICATIONS IN BIOMEDICINE. ANAL LETT 2015; 48:1044-1069. [PMID: 26973348 PMCID: PMC4787221 DOI: 10.1080/00032719.2014.976867] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurotransmitters are important biological molecules that are essential to many neurophysiological processes including memory, cognition, and behavioral states. The development of analytical methodologies to accurately detect neurotransmitters is of great importance in neurological and biological research. Specifically designed microelectrodes or microbiosensors have demonstrated potential for rapid, real-time measurements with high spatial resolution. Such devices can facilitate study of the role and mechanism of action of neurotransmitters and can find potential uses in biomedicine. This paper reviews the current status and recent advances in the development and application of electrochemical sensors for the detection of small-molecule neurotransmitters. Measurement challenges and opportunities of electroanalytical methods to advance study and understanding of neurotransmitters in various biological models and disease conditions are discussed.
Collapse
Affiliation(s)
- Rıfat Emrah Özel
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA. Fax: 3152686610; Tel: 3152682394
| | - Akhtar Hayat
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA. Fax: 3152686610; Tel: 3152682394
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology (CIIT), Lahore, Pakistan
| | - Silvana Andreescu
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA. Fax: 3152686610; Tel: 3152682394
| |
Collapse
|
32
|
Relevance of the metabotropic glutamate receptor (mGluR5) in the regulation of NREM-REM sleep cycle and homeostasis: Evidence from mGluR5 (−/−) mice. Behav Brain Res 2015; 282:218-26. [DOI: 10.1016/j.bbr.2015.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 12/21/2014] [Accepted: 01/06/2015] [Indexed: 12/16/2022]
|
33
|
Negative versus positive allosteric modulation of metabotropic glutamate receptors (mGluR5): indices for potential pro-cognitive drug properties based on EEG network oscillations and sleep-wake organization in rats. Psychopharmacology (Berl) 2015; 232:1107-22. [PMID: 25323624 DOI: 10.1007/s00213-014-3746-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 09/15/2014] [Indexed: 12/11/2022]
Abstract
RATIONALE Evidence is emerging that positive and negative modulation of the metabotropic glutamate (mGluR5) receptors has the potential for treating cognitive deficits and neuroprotection associated with psychiatric and neurodegenerative diseases, respectively. Sleep and synchronisation of disparate neuronal networks are critically involved in neuronal plasticity, and disturbance in vigilance states and cortical network connectivity contribute significantly to cognitive deficits described in schizophrenia and Alzheimer's disease. Here, we examined the circadian changes of mGluR5 density and the functional response to modulation of mGluR5 signaling. METHODS The current study carried out in Sprague-Dawley rats quantified the density of mGluR5 across the light-dark cycle with autoradiography. The central activity of mGluR5 negative allosteric modulators (2-methyl-6-(phenylethynyl)pyridine (MPEP) and [(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) and positive allosteric modulators (S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]oxadiazol-5-yl]-piperidin-1-yl}-methanone (ADX47273) and (7S)-3-tert-butyl-7-[3-(4-fluoro-phenyl)-1,2,4-oxadiazol-5-yl]-5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine (LSN2814617) was examined on sleep-wake architecture. The functional effect of mGluR5 modulation on cortical networks communication was described in freely moving animals. RESULTS The density of mGluR5 in the striatal, cortical, hippocampal and thalamic structures was unchanged across the light-dark cycle. Allosteric blockade of mGluR5 consistently consolidated deep sleep, enhanced sleep efficiency and elicited prominent functional coherent network activity in slow theta and gamma oscillations. However, allosteric activation of mGluR5 increased waking, decreased deep sleep and reduced functional network connectivity following the activation of slow alpha oscillatory activity. CONCLUSION This functional study differentiates the pharmacology of allosteric blockade of mGluR5 from that of allosteric activation and suggests that mGluR5 blockade enhances sleep and facilitates oscillatory network connectivity, both processes being known to have relevance in cognition processes.
Collapse
|
34
|
Wakabayashi KT, Myal SE, Kiyatkin EA. Fluctuations in nucleus accumbens extracellular glutamate and glucose during motivated glucose-drinking behavior: dissecting the neurochemistry of reward. J Neurochem 2015; 132:327-41. [PMID: 25393775 DOI: 10.1111/jnc.12993] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/17/2014] [Accepted: 11/10/2014] [Indexed: 12/21/2022]
Abstract
While motivated behavior involves multiple neurochemical systems, few studies have focused on the role of glutamate, the brain's excitatory neurotransmitter, and glucose, the energetic substrate of neural activity in reward-related neural processes. Here, we used high-speed amperometry with enzyme-based substrate-sensitive and control, enzyme-free biosensors to examine second-scale fluctuations in the extracellular levels of these substances in the nucleus accumbens shell during glucose-drinking behavior in trained rats. Glutamate rose rapidly after the presentation of a glucose-containing cup and before the initiation of drinking (reward seeking), decreased more slowly to levels below baseline during consumption (sensory reward), and returned to baseline when the ingested glucose reached the brain (metabolic reward). When water was substituted for glucose, glutamate rapidly increased with cup presentation and in contrast to glucose drinking, increased above baseline after rats tasted the water and refused to drink further. Therefore, extracellular glutamate show distinct changes associated with key events of motivated drinking behavior and opposite dynamics during sensory and metabolic components of reward. In contrast to glutamate, glucose increased at each stimulus and behavioral event, showing a sustained elevation during the entire behavior and a robust post-ingestion rise that correlated with the gradual return of glutamate levels to their baseline. By comparing active drinking with passive intra-gastric glucose delivery, we revealed that fluctuations in extracellular glucose are highly dynamic, reflecting a balance between rapid delivery because of neural activity, intense metabolism, and the influence of ingested glucose reaching the brain.
Collapse
Affiliation(s)
- Ken T Wakabayashi
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, DHHS, Baltimore, Maryland, USA
| | | | | |
Collapse
|
35
|
Aluisio L, Fraser I, Berdyyeva T, Tryputsen V, Shireman BT, Shoblock J, Lovenberg T, Dugovic C, Bonaventure P. Pharmacological or genetic orexin1 receptor inhibition attenuates MK-801 induced glutamate release in mouse cortex. Front Neurosci 2014; 8:107. [PMID: 24904253 PMCID: PMC4033200 DOI: 10.3389/fnins.2014.00107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/24/2014] [Indexed: 12/13/2022] Open
Abstract
The orexin/hypocretin neuropeptides are produced by a cluster of neurons within the lateral posterior hypothalamus and participate in neuronal regulation by activating their receptors (OX1 and OX2 receptors). The orexin system projects widely through the brain and functions as an interface between multiple regulatory systems including wakefulness, energy balance, stress, reward, and emotion. Recent studies have demonstrated that orexins and glutamate interact at the synaptic level and that orexins facilitate glutamate actions. We tested the hypothesis that orexins modulate glutamate signaling via OX1 receptors by monitoring levels of glutamate in frontal cortex of freely moving mice using enzyme coated biosensors under inhibited OX1 receptor conditions. MK-801, an NMDA receptor antagonist, was administered subcutaneously (0.178 mg/kg) to indirectly disinhibit pyramidal neurons and therefore increase cortical glutamate release. In wild-type mice, pretreatment with the OX1 receptor antagonist GSK-1059865 (10 mg/kg S.C.) which had no effect by itself, significantly attenuated the cortical glutamate release elicited by MK-801. OX1 receptor knockout mice had a blunted glutamate release response to MK-801 and exhibited about half of the glutamate release observed in wild-type mice in agreement with the data obtained with transient blockade of OX1 receptors. These results indicate that pharmacological (transient) or genetic (permanent) inhibition of the OX1 receptor similarly interfere with glutamatergic function in the cortex. Selectively targeting the OX1 receptor with an antagonist may normalize hyperglutamatergic states and thus may represent a novel therapeutic strategy for the treatment of various psychiatric disorders associated with hyperactive states.
Collapse
Affiliation(s)
- Leah Aluisio
- Janssen Pharmaceutical Research and Development, LLC San Diego, USA
| | - Ian Fraser
- Janssen Pharmaceutical Research and Development, LLC San Diego, USA
| | - Tamara Berdyyeva
- Janssen Pharmaceutical Research and Development, LLC San Diego, USA
| | - Volha Tryputsen
- Janssen Pharmaceutical Research and Development, LLC San Diego, USA
| | - Brock T Shireman
- Janssen Pharmaceutical Research and Development, LLC San Diego, USA
| | - James Shoblock
- Janssen Pharmaceutical Research and Development, LLC San Diego, USA
| | | | | | | |
Collapse
|
36
|
Ahnaou A, Ver Donck L, Drinkenburg WHIM. Blockade of the metabotropic glutamate (mGluR2) modulates arousal through vigilance states transitions: evidence from sleep-wake EEG in rodents. Behav Brain Res 2014; 270:56-67. [PMID: 24821401 DOI: 10.1016/j.bbr.2014.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/15/2014] [Accepted: 05/02/2014] [Indexed: 02/03/2023]
Abstract
Accumulating data continue to support the therapeutic potential of glutamate metabotropic (mGluR2) receptors for treatment of psychiatric disorders such as depression, anxiety and schizophrenia. Glutamate neurotransmission is an integral component of sleep-wake mechanisms, which have translational relevance to assess on-target activity of drugs. Here, we investigated the influence of mGluR2 inactivation upon sleep-wake electroencephalogram (EEG) in rodents. Rats were administered with vehicle, the specific mGluR2 antagonist LY341495 (2.5, 5, 10mg/kg) or negative allosteric modulator (NAM) Ro-4491533 (2.5, 10 and 40 mg/kg) at lights onset. mGluR2 (-/-) mice were used to confirm the selectivity of functional response. Both LY341495 and Ro-4491533 induced an immediate and endured desynchronized cortical activity during 3-6h associated with enhanced theta and gamma oscillations and depressed slow oscillations during sleep. The arousal-promoting effect is consistent with the marked lengthening of sleep onset latency, an increased number of state transitions from light sleep to waking and the gradual increase in homeostatic compensatory sleep. The arousal response to mGluR2 blockade was not accompanied by sharp rebound hypersomnolence as found with the classical psycho-stimulant amphetamine. mGluR2 (-/-) mice and their WT littermates exhibited similar sleep-wake phenotype, while Ro-4491533 (40 mg/kg) enhanced waking associated with increased locomotor activity and body temperature in WT but not in mGluR2 (-/-) mice, which confirm the role of mGluR2 inactivation in the arousal response. Our results lend support for a role of mGluR2 blockade in promoting cortical arousal associated with theta/gamma oscillations as well as high thresholds transitions from sleep to waking.
Collapse
Affiliation(s)
- A Ahnaou
- Department of Neurosciences, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | - L Ver Donck
- Department of Neurosciences, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - W H I M Drinkenburg
- Department of Neurosciences, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
37
|
Lenoir M, Kiyatkin EA. Intravenous nicotine injection induces rapid, experience-dependent sensitization of glutamate release in the ventral tegmental area and nucleus accumbens. J Neurochem 2013; 127:541-51. [PMID: 24032718 DOI: 10.1111/jnc.12450] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/20/2013] [Accepted: 09/10/2013] [Indexed: 11/28/2022]
Abstract
Although numerous data suggest that glutamate (GLU) is involved in mediating the neural effects of nicotine, direct data on nicotine-induced changes in GLU release are still lacking. Here, we used high-speed amperometry with enzyme-based GLU and enzyme-free GLU-null biosensors to examine changes in extracellular GLU levels in the ventral tegmental area (VTA) and nucleus accumbens shell (NAcc) induced by intravenous nicotine in a low, behaviorally active dose (30 μg/kg) in freely moving rats. Using this approach, we found that the initial nicotine injection in drug-naive conditions induces rapid, transient, and relatively small GLU release (~ 90 nM; latency ~ 15 s, duration ~ 60 s) that is correlative in the VTA and NAcc. Following subsequent nicotine injections within the same session, this phasic GLU release was supplemented by stronger tonic increases in GLU levels (100-300 nM) that paralleled increases in drug-induced locomotor activation. GLU responses induced by repeated nicotine injections were more phasic and stronger in the NAcc than in VTA. Therefore, GLU is phasically released within the brain's reinforcement circuit following intravenous nicotine administration. Robust enhancement of nicotine-induced GLU responses following repeated injections suggests this change as an important mediator of sensitized behavioral and neural effects of nicotine. By using high-speed amperometry with glutamate (GLU) biosensors, we show that i.v. nicotine at a low, behaviorally relevant dose induces rapid GLU release in the NAcc and VTA that is enhanced following repeated drug injections. This is the first study reporting second-scale fluctuations in extracellular GLU levels induced by nicotine in two critical structures of the motivation-reinforcement circuit and rapid sensitization of GLU responses coupled with locomotor sensitization.
Collapse
Affiliation(s)
- Magalie Lenoir
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, DHHS, Baltimore, Maryland, USA
| | | |
Collapse
|
38
|
Kiyatkin EA, Wakabayashi KT, Lenoir M. Physiological fluctuations in brain temperature as a factor affecting electrochemical evaluations of extracellular glutamate and glucose in behavioral experiments. ACS Chem Neurosci 2013; 4:652-65. [PMID: 23448428 DOI: 10.1021/cn300232m] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The rate of any chemical reaction or process occurring in the brain depends on temperature. While it is commonly believed that brain temperature is a stable, tightly regulated homeostatic parameter, it fluctuates within 1-4 °C following exposure to salient arousing stimuli and neuroactive drugs, and during different behaviors. These temperature fluctuations should affect neural activity and neural functions, but the extent of this influence on neurochemical measurements in brain tissue of freely moving animals remains unclear. In this Review, we present the results of amperometric evaluations of extracellular glutamate and glucose in awake, behaving rats and discuss how naturally occurring fluctuations in brain temperature affect these measurements. While this temperature contribution appears to be insignificant for glucose because its extracellular concentrations are large, it is a serious factor for electrochemical evaluations of glutamate, which is present in brain tissue at much lower levels, showing smaller phasic fluctuations. We further discuss experimental strategies for controlling the nonspecific chemical and physical contributions to electrochemical currents detected by enzyme-based biosensors to provide greater selectivity and reliability of neurochemical measurements in behaving animals.
Collapse
Affiliation(s)
- Eugene A. Kiyatkin
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience
Branch, National Institute on Drug Abuse − Intramural Research
Program, National Institutes of Health,
DHHS, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Ken T. Wakabayashi
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience
Branch, National Institute on Drug Abuse − Intramural Research
Program, National Institutes of Health,
DHHS, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Magalie Lenoir
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience
Branch, National Institute on Drug Abuse − Intramural Research
Program, National Institutes of Health,
DHHS, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
39
|
Naylor E, Aillon DV, Barrett BS, Wilson GS, Johnson DA, Johnson DA, Harmon HP, Gabbert S, Petillo PA. Lactate as a biomarker for sleep. Sleep 2012; 35:1209-22. [PMID: 22942499 DOI: 10.5665/sleep.2072] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES An ideal biomarker for sleep should change rapidly with sleep onset, remain at a detectably differential level throughout the sleep period, and exhibit a rapid change with waking. Currently, no molecular marker has been identified that exhibits all three properties. This study examined three substances (lactate, glucose, and glutamate) for suitability as a sleep biomarker. DESIGN Using amperometric biosensor technology in conjunction with electroencephalograph (EEG) and electromyograph (EMG) monitoring, extracellular concentrations of lactate and glucose (Cohort 1) as well as lactate and glutamate (Cohort 2) were recorded over multiple sleep/wake cycles. PATIENTS OR PARTICIPANTS There were 12 C57Bl/6J male mice (3-5 mo old). INTERVENTIONS Sleep and waking transitions were identified using EEG recordings. Extracellular concentrations of lactate, glucose, and glutamate were evaluated before and during transition events as well as during extended sleep and during a 6-h sleep deprivation period. MEASUREMENTS AND RESULTS Rapid and sustained increases in cortical lactate concentration (approximately 15 μM/min) were immediately observed upon waking and during rapid eye movement sleep. Elevated lactate concentration was also maintained throughout a 6-h period of continuous waking. A persistent and sustained decline in lactate concentration was measured during nonrapid eye movement sleep. Glutamate exhibited similar patterns, but with a much slower rise and decline (approximately 0.03 μM/min). Glucose concentration changes did not demonstrate a clear correlation with either sleep or wake. CONCLUSIONS These findings indicate that extracellular lactate concentration is a reliable sleep/wake biomarker and can be used independently of the EEG signal.
Collapse
Affiliation(s)
- Erik Naylor
- Pinnacle Technology, Inc., Lawrence, KS 66046, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wisor JP, Rempe MJ, Schmidt MA, Moore ME, Clegern WC. Sleep slow-wave activity regulates cerebral glycolytic metabolism. Cereb Cortex 2012; 23:1978-87. [PMID: 22767634 DOI: 10.1093/cercor/bhs189] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Non-rapid eye movement sleep (NREMS) onset is characterized by a reduction in cerebral metabolism and an increase in slow waves, 1-4-Hz oscillations between relatively depolarized and hyperpolarized states in the cerebral cortex. The metabolic consequences of slow-wave activity (SWA) at the cellular level remain uncertain. We sought to determine whether SWA modulates the rate of glycolysis within the cerebral cortex. The real-time measurement of lactate concentration in the mouse cerebral cortex demonstrates that it increases during enforced wakefulness. In spontaneous sleep/wake cycles, lactate concentration builds during wakefulness and rapid eye movement sleep and declines during NREMS. The rate at which lactate concentration declines during NREMS is proportional to the magnitude of electroencephalographic (EEG) activity at frequencies of <10 Hz. The induction of 1-Hz oscillations, but not 10-Hz oscillations, in the electroencephalogram by optogenetic stimulation of cortical pyramidal cells during wakefulness triggers a decline in lactate concentration. We conclude that cerebral SWA promotes a decline in the rate of glycolysis in the cerebral cortex. These results demonstrate a cellular energetic function for sleep SWA, which may contribute to its restorative effects on brain function.
Collapse
Affiliation(s)
- Jonathan P Wisor
- WWAMI Medical Education Program, Department of Veterinary Comparative Anatomy, Pharmacology and Physiology, Washington State University, Spokane, WA 99210-1945, USA.
| | | | | | | | | |
Collapse
|
41
|
Wakabayashi KT, Kiyatkin EA. Rapid changes in extracellular glutamate induced by natural arousing stimuli and intravenous cocaine in the nucleus accumbens shell and core. J Neurophysiol 2012; 108:285-99. [PMID: 22496525 DOI: 10.1152/jn.01167.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Glutamate (Glu) is a major excitatory neurotransmitter, playing a crucial role in the functioning of the nucleus accumbens (NAc), a critical area implicated in somatosensory integration and regulation of motivated behavior. In this study, high-speed amperometry with enzyme-based biosensors was used in freely moving rats to examine changes in extracellular Glu in the NAc shell and core induced by a tone, tail pinch (TP), social interaction with a male conspecific (SI), and intravenous (iv) cocaine (1 mg/kg). To establish the contribution of Glu to electrochemical signal changes, similar recordings were conducted with null (Glu(0)) sensors, which were exposed to the same chemical and physical environment but were insensitive to Glu. TP, SI, and cocaine, but not a tone, induced relatively large and prolonged current increases detected by both Glu and Glu(0) sensors. However, current differentials revealed very rapid, much smaller, and transient increases in extracellular Glu levels, more predominantly in the NAc shell than core. In contrast to monophasic responses with natural stimuli, cocaine induced a biphasic Glu increase in the shell, with a transient peak during the injection and a slower postinjection peak. Therefore, Glu is phasically released in the NAc after exposure to natural arousing stimuli and cocaine; this release is rapid, stimulus dependent, and structure specific, suggesting its role in triggering neural and behavioral activation induced by these stimuli. This study also demonstrates the need for multiple in vitro and in vivo controls to reveal relatively small, highly phasic, and transient fluctuations in Glu levels occurring under behaviorally relevant conditions.
Collapse
Affiliation(s)
- Ken T Wakabayashi
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | | |
Collapse
|
42
|
Wisor JP. A metabolic-transcriptional network links sleep and cellular energetics in the brain. Pflugers Arch 2012; 463:15-22. [PMID: 21927810 PMCID: PMC4086657 DOI: 10.1007/s00424-011-1030-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 09/05/2011] [Accepted: 09/07/2011] [Indexed: 12/22/2022]
Abstract
This review proposes a mechanistic link between cellular metabolic status, transcriptional regulatory changes and sleep. Sleep loss is associated with changes in cellular metabolic status in the brain. Metabolic sensors responsive to cellular metabolic status regulate the circadian clock transcriptional network. Modifications of the transcriptional activity of circadian clock genes affect sleep/wake state changes. Changes in sleep state reverse sleep loss-induced changes in cellular metabolic status. It is thus proposed that the regulation of circadian clock genes by cellular metabolic sensors is a critical intermediate step in the link between cellular metabolic status and sleep. Studies of this regulatory relationship may offer insights into the function of sleep at the cellular level.
Collapse
Affiliation(s)
- Jonathan P Wisor
- WWAMI Medical Education Program and Department of Veterinary Comparative Anatomy, Pharmacology and Physiology, Washington State University, Spokane, WA, USA.
| |
Collapse
|
43
|
Khan R, Gorski W, Garcia CD. Nanomolar Detection of Glutamate at a Biosensor Based on Screen-Printed Electrodes Modified with Carbon Nanotubes. ELECTROANAL 2011; 23:2357-2363. [PMID: 22735259 PMCID: PMC3379819 DOI: 10.1002/elan.201100348] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Accepted: 08/15/2011] [Indexed: 11/10/2022]
Abstract
The amperometric glutamate biosensor based on screen-printed electrodes containing carbon nanotubes (CNT), and its integration in a flow injection analysis system, is described herein. The sensor was fabricated by simply adsorbing enzyme glutamate oxidase (GlutOx) on a commercial substrate containing multi-wall CNT. The resulting device displayed excellent electroanalytical properties toward the determination of L-glutamate in a wide linear range (0.01-10 μM) with low detection limit (10 nM, S/N≥3), fast response time (≤5 s), and good operational and long-term stability. The CNT modified screen-printed electrodes have a potential to be of general interest for designing of electrochemical sensors and biosensors.
Collapse
Affiliation(s)
- Raju Khan
- Analytical Chemistry Division, North East Institute of Science & Technology, Jorhat, 785006, Assam, India
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, United States of America
| | - Waldemar Gorski
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, United States of America
| | - Carlos D. Garcia
- Department of Chemistry, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, United States of America
| |
Collapse
|
44
|
Neurochemical and electrophysiological changes induced by paradoxical sleep deprivation in rats. Behav Brain Res 2011; 225:39-46. [PMID: 21729722 DOI: 10.1016/j.bbr.2011.06.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 06/12/2011] [Accepted: 06/18/2011] [Indexed: 12/26/2022]
Abstract
The present study aims to investigate the effects of paradoxical sleep deprivation (PSD) on the waking EEG and amino acid neurotransmitters in the hippocampus and cortex of rats. Animals were deprived of paradoxical sleep for 72h by using the multiple platform method. The EEG power spectral analysis was carried out to assess the brain's electrophysiological changes due to sleep deprivation. The concentrations of amino acid neurotransmitters were assessed in the hippocampus and cortex using HPLC. Control data showed slight differences from normal animals in the delta, theta and alpha waves while an increase in the beta wave was obtained. After 24h of PSD, delta relative power increased and the rest of EEG wave's power decreased with respect to control. After 48h and 72h the spectral power analysis showed non-significant changes to control. The amino acid neurotransmitter analysis revealed a significant increase in cortical glutamate, glycine and taurine levels while in the hippocampus, glutamate, aspartate, glutamine and glycine levels increased significantly. Both the waking EEG and neurotransmitter analyses suggest that PSD induced neurochemical and electrophysiological changes that may affect brain proper functionality.
Collapse
|