1
|
Bahi A. Hippocampal overexpression of tissue-type plasminogen activator "tPA" attenuates social defeat-induced depression and ethanol related behavior in mice. Alcohol 2025; 125:1-15. [PMID: 39938666 DOI: 10.1016/j.alcohol.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/01/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Depression and anxiety disorders are often exacerbated by social stress, necessitating the exploration of molecular mechanisms underlying stress resilience. Tissue plasminogen activator (tPA), a serine protease with pleiotropic effects in the brain, plays a critical role in modulating neuroplasticity and stress responses. This study investigates the behavioral and molecular effects of tPA gain-of-function in a social stress paradigm in male C57BL/6 mice using lentiviral vectors. Behaviorally, hippocampal tPA gain-of-function mitigated depression-like responses in the novelty-suppressed feeding, sucrose splash, tail suspension, and forced swim tests following exposure to chronic social stress. Additionally, in a two-bottle choice drinking paradigm, tPA overexpression reduced social stress-induced ethanol intake and preference, suggesting a role in dampening maladaptive coping behaviors. However, analysis of tastants' intake and preference revealed no significant effects of tPA overexpression, indicating that it does not influence hedonic responses under stress conditions. Molecularly, tPA overexpression preserved hippocampal tPA mRNA expression and maintained levels of mature brain-derived neurotrophic factor in the hippocampus despite chronic stress exposure. These findings highlight the potential neuroprotective effects of tPA in maintaining hippocampal plasticity and mitigating stress-induced dysregulation of critical neurotrophic pathways. Collectively, this study underscores the potential of tPA as a therapeutic target for stress-induced mood and substance use disorders by modulating behavioral and neurobiological responses to chronic social stress.
Collapse
Affiliation(s)
- Amine Bahi
- Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman, United Arab Emirates; Center of Medical & Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; College of Medicine & Health Sciences, UAEU, Al Ain, United Arab Emirates.
| |
Collapse
|
2
|
Ma S, Skarica M, Li Q, Xu C, Risgaard RD, Tebbenkamp AT, Mato-Blanco X, Kovner R, Krsnik Ž, de Martin X, Luria V, Martí-Pérez X, Liang D, Karger A, Schmidt DK, Gomez-Sanchez Z, Qi C, Gobeske KT, Pochareddy S, Debnath A, Hottman CJ, Spurrier J, Teo L, Boghdadi AG, Homman-Ludiye J, Ely JJ, Daadi EW, Mi D, Daadi M, Marín O, Hof PR, Rasin MR, Bourne J, Sherwood CC, Santpere G, Girgenti MJ, Strittmatter SM, Sousa AM, Sestan N. Molecular and cellular evolution of the primate dorsolateral prefrontal cortex. Science 2022; 377:eabo7257. [PMID: 36007006 PMCID: PMC9614553 DOI: 10.1126/science.abo7257] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The granular dorsolateral prefrontal cortex (dlPFC) is an evolutionary specialization of primates that is centrally involved in cognition. We assessed more than 600,000 single-nucleus transcriptomes from adult human, chimpanzee, macaque, and marmoset dlPFC. Although most cell subtypes defined transcriptomically are conserved, we detected several that exist only in a subset of species as well as substantial species-specific molecular differences across homologous neuronal, glial, and non-neural subtypes. The latter are exemplified by human-specific switching between expression of the neuropeptide somatostatin and tyrosine hydroxylase, the rate-limiting enzyme in dopamine production in certain interneurons. The above molecular differences are also illustrated by expression of the neuropsychiatric risk gene FOXP2, which is human-specific in microglia and primate-specific in layer 4 granular neurons. We generated a comprehensive survey of the dlPFC cellular repertoire and its shared and divergent features in anthropoid primates.
Collapse
Affiliation(s)
- Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mario Skarica
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Qian Li
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Chuan Xu
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ryan D. Risgaard
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Xoel Mato-Blanco
- Neurogenomics Group, Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), MELIS, Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Rothem Kovner
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Željka Krsnik
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Xabier de Martin
- Neurogenomics Group, Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), MELIS, Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Victor Luria
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Xavier Martí-Pérez
- Neurogenomics Group, Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), MELIS, Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Dan Liang
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Amir Karger
- IT-Research Computing, Harvard Medical School, Boston, MA, USA
| | - Danielle K. Schmidt
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary Gomez-Sanchez
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cai Qi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kevin T. Gobeske
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sirisha Pochareddy
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ashwin Debnath
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cade J. Hottman
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joshua Spurrier
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neurology, Yale School of Medicine, New Haven, CT 06536, USA
| | - Leon Teo
- Australian Regenerative Medicine Institute, 15 Innovation Walk, Monash University, Clayton VIC, 3800, Australia
| | - Anthony G. Boghdadi
- Australian Regenerative Medicine Institute, 15 Innovation Walk, Monash University, Clayton VIC, 3800, Australia
| | - Jihane Homman-Ludiye
- Australian Regenerative Medicine Institute, 15 Innovation Walk, Monash University, Clayton VIC, 3800, Australia
| | - John J. Ely
- MAEBIOS, Alamogordo, NM 88310, USA
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Etienne W. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Da Mi
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Marcel Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Cell Systems & Anatomy, Radiology, Long School of Medicine, UT Health San Antonio
- NeoNeuron LLC, Palo Alto, CA 94306, USA
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mladen-Roko Rasin
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - James Bourne
- Australian Regenerative Medicine Institute, 15 Innovation Walk, Monash University, Clayton VIC, 3800, Australia
| | - Chet C. Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Gabriel Santpere
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Neurogenomics Group, Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), MELIS, Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Matthew J. Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA
- National Center for PTSD, US Department of Veterans Affairs, White River Junction, VT, USA
| | - Stephen M. Strittmatter
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neurology, Yale School of Medicine, New Haven, CT 06536, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - André M.M. Sousa
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Departments of Genetics and Comparative Medicine, Program in Cellular Neuroscience, Neurodegeneration and Repair, and Yale Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
3
|
Gonias SL. Plasminogen activator receptor assemblies in cell signaling, innate immunity, and inflammation. Am J Physiol Cell Physiol 2021; 321:C721-C734. [PMID: 34406905 DOI: 10.1152/ajpcell.00269.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) are serine proteases and major activators of fibrinolysis in mammalian systems. Because fibrinolysis is an essential component of the response to tissue injury, diverse cells, including cells that participate in the response to injury, have evolved receptor systems to detect tPA and uPA and initiate appropriate cell-signaling responses. Formation of functional receptor systems for the plasminogen activators requires assembly of diverse plasma membrane proteins, including but not limited to: the urokinase receptor (uPAR); integrins; N-formyl peptide receptor-2 (FPR2), receptor tyrosine kinases (RTKs), the N-methyl-d-aspartate receptor (NMDA-R), and low-density lipoprotein receptor-related protein-1 (LRP1). The cell-signaling responses elicited by tPA and uPA impact diverse aspects of cell physiology. This review describes rapidly evolving knowledge regarding the structure and function of plasminogen activator receptor assemblies. How these receptor assemblies regulate innate immunity and inflammation is then considered.
Collapse
Affiliation(s)
- Steven L Gonias
- Department of Pathology, University of California, San Diego, California
| |
Collapse
|
4
|
Mizoguchi A, Banno R, Sun R, Yaginuma H, Taki K, Kobayashi T, Sugiyama M, Tsunekawa T, Onoue T, Takagi H, Hagiwara D, Ito Y, Iwama S, Suga H, Nagai T, Yamada K, Arima H. Glucocorticoid receptor signaling in ventral tegmental area neurons increases the rewarding value of a high-fat diet in mice. Sci Rep 2021; 11:12873. [PMID: 34145364 PMCID: PMC8213822 DOI: 10.1038/s41598-021-92386-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/07/2021] [Indexed: 11/09/2022] Open
Abstract
The reward system, which consists of dopaminergic neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens and caudate-putamen in the striatum, has an important role in the pathogenesis of not only drug addiction but also diet-induced obesity. In the present study, we examined whether signaling through glucocorticoid receptors (GRs) in the reward system affects the rewarding value of a high-fat diet (HFD). To do so, we generated mice that lack functional GRs specifically in dopaminergic neurons (D-KO mice) or corticostriatal neurons (CS-KO mice), subjected the mice to caloric restriction stress conditions, and evaluated the rewarding value of a HFD by conditioned place preference (CPP) test. Caloric restriction induced increases in serum corticosterone to similar levels in all genotypes. While CS-KO as well as WT mice exhibited a significant preference for HFD in the CPP test, D-KO mice exhibited no such preference. There were no differences between WT and D-KO mice in consumption of HFD after fasting or cognitive function evaluated by a novel object recognition test. These data suggest that glucocorticoid signaling in the VTA increases the rewarding value of a HFD under restricted caloric stress.
Collapse
Affiliation(s)
- Akira Mizoguchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan. .,Physical Fitness and Sports, Research Center of Health, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-0814, Japan.
| | - Runan Sun
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Hiroshi Yaginuma
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Keigo Taki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan.,Department of CKD Initiatives/Nephrology, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan.,Division of Behavioral Neuropharmacology, Project Office for Neuropsychological Research Center, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan.
| |
Collapse
|
5
|
Medcalf RL, Keragala CB. The Fibrinolytic System: Mysteries and Opportunities. Hemasphere 2021; 5:e570. [PMID: 34095754 PMCID: PMC8171360 DOI: 10.1097/hs9.0000000000000570] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023] Open
Abstract
The deposition and removal of fibrin has been the primary role of coagulation and fibrinolysis, respectively. There is also little doubt that these 2 enzyme cascades influence each other given they share the same serine protease family ancestry and changes to 1 arm of the hemostatic pathway would influence the other. The fibrinolytic system in particular has also been known for its capacity to clear various non-fibrin proteins and to activate other enzyme systems, including complement and the contact pathway. Furthermore, it can also convert a number of growth factors into their mature, active forms. More recent findings have extended the reach of this system even further. Here we will review some of these developments and also provide an account of the influence of individual players of the fibrinolytic (plasminogen activating) pathway in relation to physiological and pathophysiological events, including aging and metabolism.
Collapse
Affiliation(s)
- Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Victoria, Australia
| | - Charithani B. Keragala
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Victoria, Australia
| |
Collapse
|
6
|
Price R, Ferrari E, Gardoni F, Mercuri NB, Ledonne A. Protease-activated receptor 1 (PAR1) inhibits synaptic NMDARs in mouse nigral dopaminergic neurons. Pharmacol Res 2020; 160:105185. [PMID: 32891865 DOI: 10.1016/j.phrs.2020.105185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022]
Abstract
Protease-activated receptor 1 (PAR1) is a G protein-coupled receptor (GPCR), whose activation requires a proteolytic cleavage in the extracellular domain exposing a tethered ligand, which binds to the same receptor thus stimulating Gαq/11-, Gαi/o- and Gα12-13 proteins. PAR1, activated by serine proteases and matrix metalloproteases, plays multifaceted roles in neuroinflammation and neurodegeneration, in stroke, brain trauma, Alzheimer's diseases, and Parkinson's disease (PD). Substantia nigra pars compacta (SNpc) is among areas with highest PAR1 expression, but current evidence on its roles herein is restricted to mechanisms controlling dopaminergic (DAergic) neurons survival, with controversial data showing PAR1 either fostering or counteracting degeneration in PD models. Since PAR1 functions on SNpc DAergic neurons activity are unknown, we investigated if PAR1 affects glutamatergic transmission in this neuronal population. We analyzed PAR1's effects on NMDARs and AMPARs by patch-clamp recordings from DAergic neurons from mouse midbrain slices. Then, we explored subunit composition of PAR1-sensitive NMDARs, with selective antagonists, and mechanisms underlying PAR1-induced NMDARs modulation, by quantifying NMDARs surface expression. PAR1 activation inhibits synaptic NMDARs in SNpc DAergic neurons, without affecting AMPARs. PAR1-sensitive NMDARs contain GluN2B/GluN2D subunits. Moreover, PAR1-mediated NMDARs hypofunction is reliant on NMDARs internalization, as PAR1 stimulation increases NMDARs intracellular levels and pharmacological limitation of NMDARs endocytosis prevents PAR1-induced NMDARs inhibition. We reveal that PAR1 regulates glutamatergic transmission in midbrain DAergic cells. This might have implications in brain's DA-dependent functions and in neurological/psychiatric diseases linked to DAergic dysfunctions.
Collapse
Affiliation(s)
- Rachel Price
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, Università di Roma Tor Vergata, Rome, Italy
| | - Elena Ferrari
- Department of Pharmacological and Biomolecolar Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecolar Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, Università di Roma Tor Vergata, Rome, Italy
| | - Ada Ledonne
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
7
|
Role of tissue plasminogen activator and plasminogen activator inhibitor as potential biomarkers in psychosis. Asian J Psychiatr 2019; 43:105-110. [PMID: 31125952 DOI: 10.1016/j.ajp.2019.05.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/03/2019] [Accepted: 05/13/2019] [Indexed: 11/23/2022]
Abstract
The identification of biological markers for psychosis has an impact on its diagnosis, prognosis, and likelihood of treatment response. Tissue plasminogen activator (tPA) is involved in important functions such as synaptic plasticity, long-term potentiation and neurogenesis. Plasminogen activator inhibitor (PAI-1) is the most important inhibitor of tPA. Preliminary studies have shown that schizophrenia patients have lower tPA and higher PAI-1 levels than the general population. The association of tPA and PAI-1 abnormalities with psychotic spectrum disorders, however, remains elusive. Our primary objective was to assess the plasma levels of tPA and PAI-1 in patients experiencing acute psychotic episodes as compared to those in healthy controls. In this prospective case-control study, we collected peripheral blood samples from psychiatric inpatients and healthy age, gender and race-matched subjects and determined plasma levels of tPA and PAI-1 by enzyme-linked immune-absorbent assays. Plasma levels of PAI-1 in patients with schizoaffective disorder were significantly lower as compared to those in control subjects (P = 0.03). tPA was lower in cases as compared to controls although it did not reach statistical significance. Asian patients and controls had lower PAI-1 levels. Further, Asian patients with schizoaffective disorder had significantly lower PAI-1 level compared to Asian patients with schizophrenia. Our results indicate that patients with schizoaffective disorder have lower PAI-1 levels than those with schizophrenia, affective psychosis, and healthy controls. Further studies are warranted to explore the potential of PAI-1 as a biomarker for diagnosing schizoaffective disorder.
Collapse
|
8
|
Mizoguchi H, Wang T, Kusaba M, Fukumoto K, Yamada K. Nicotine and varenicline ameliorate changes in reward-based choice strategy and altered decision-making in methamphetamine-treated rats. Behav Brain Res 2019; 359:935-941. [DOI: 10.1016/j.bbr.2018.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/29/2022]
|
9
|
Zalfa C, Azmoon P, Mantuano E, Gonias SL. Tissue-type plasminogen activator neutralizes LPS but not protease-activated receptor-mediated inflammatory responses to plasmin. J Leukoc Biol 2019; 105:729-740. [PMID: 30690783 DOI: 10.1002/jlb.3a0818-329rrr] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/28/2018] [Accepted: 12/29/2018] [Indexed: 12/18/2022] Open
Abstract
Tissue-type plasminogen activator (tPA) activates fibrinolysis and also suppresses innate immune system responses to LPS in bone marrow-derived macrophages (BMDMs) and in vivo in mice. The objective of this study was to assess the activity of tPA as a regulator of macrophage physiology in the presence of plasmin. Enzymatically active and enzymatically inactive (EI) tPA appeared to comprehensively block the response to LPS in BMDMs, including expression of proinflammatory cytokines such as TNF-α and IL-1β and anti-inflammatory cytokines such as IL-10 and IL-1 receptor antagonist. The activity of EI-tPA as an LPS response modifier was conserved in the presence of plasminogen. By contrast, in BMDMs treated with tPA and plasminogen or preactivated plasmin, in the presence or absence of LPS, increased proinflammatory cytokine expression was observed and tPA failed to reverse the response. Plasmin independently activated NF-κB, ERK1/2, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase in BMDMs, which is characteristic of proinflammatory stimuli. Plasmin-induced cytokine expression was blocked by ε-aminocaproic acid, aprotinin, and inhibitors of the known plasmin substrate, Protease-activated receptor-1 (PAR-1), but not by N-methyl-d-aspartate receptor inhibitor, which blocks the effects of tPA on macrophages. Cytokine expression by BMDMs treated with the PAR-1 agonist, TFLLR, was not inhibited by EI-tPA, possibly explaining why EI-tPA does not inhibit macrophage responses to plasmin and providing evidence for specificity in the ability of tPA to oppose proinflammatory stimuli. Regulation of innate immunity by the fibrinolysis system may reflect the nature of the stimulus and a balance between the potentially opposing activities of tPA and plasmin.
Collapse
Affiliation(s)
- Cristina Zalfa
- The Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Pardis Azmoon
- The Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Elisabetta Mantuano
- The Department of Pathology, University of California San Diego, La Jolla, California, USA.,The Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Steven L Gonias
- The Department of Pathology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
10
|
Lasek AW. Effects of Ethanol on Brain Extracellular Matrix: Implications for Alcohol Use Disorder. Alcohol Clin Exp Res 2016; 40:2030-2042. [PMID: 27581478 DOI: 10.1111/acer.13200] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/24/2016] [Indexed: 12/20/2022]
Abstract
The brain extracellular matrix (ECM) occupies the space between cells and is involved in cell-matrix and cell-cell adhesion. However, in addition to providing structural support to brain tissue, the ECM activates cell signaling and controls synaptic transmission. The expression and activity of brain ECM components are regulated by alcohol exposure. This review will discuss what is currently known about the effects of alcohol on the activity and expression of brain ECM components. An interpretation of how these changes might promote alcohol use disorder (AUD) will be also provided. Ethanol (EtOH) exposure decreases levels of structural proteins involved in the interstitial matrix and basement membrane, with a concomitant increase in proteolytic enzymes that degrade these components. In contrast, EtOH exposure generally increases perineuronal net components. Because the ECM has been shown to regulate both synaptic plasticity and behavioral responses to drugs of abuse, regulation of the brain ECM by alcohol may be relevant to the development of alcoholism. Although investigation of the function of brain ECM in alcohol abuse is still in early stages, a greater understanding of the interplay between ECM and alcohol might lead to novel therapeutic strategies for treating AUD.
Collapse
Affiliation(s)
- Amy W Lasek
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
11
|
Schuldt G, Galanis C, Strehl A, Hick M, Schiener S, Lenz M, Deller T, Maggio N, Vlachos A. Inhibition of Protease-Activated Receptor 1 Does not Affect Dendritic Homeostasis of Cultured Mouse Dentate Granule Cells. Front Neuroanat 2016; 10:64. [PMID: 27378862 PMCID: PMC4904007 DOI: 10.3389/fnana.2016.00064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/27/2016] [Indexed: 12/25/2022] Open
Abstract
Protease-activated receptors (PARs) are widely expressed in the central nervous system (CNS). While a firm link between PAR1-activation and functional synaptic and intrinsic neuronal properties exists, studies on the role of PAR1 in neural structural plasticity are scarce. The physiological function of PAR1 in the brain remains not well understood. We here sought to determine whether prolonged pharmacologic PAR1-inhibition affects dendritic morphologies of hippocampal neurons. To address this question we employed live-cell microscopy of mouse dentate granule cell dendrites in 3-week old entorhino-hippocampal slice cultures prepared from Thy1-GFP mice. A subset of cultures were treated with the PAR1-inhibitor SCH79797 (1 μM; up to 3 weeks). No major effects of PAR1-inhibition on static and dynamic parameters of dentate granule cell dendrites were detected under control conditions. Granule cells of PAR1-deficient slice cultures showed unaltered dendritic morphologies, dendritic spine densities and excitatory synaptic strength. Furthermore, we report that PAR1-inhibition does not prevent dendritic retraction following partial deafferentation in vitro. Consistent with this finding, no major changes in PAR1-mRNA levels were detected in the denervated dentate gyrus (DG). We conclude that neural PAR1 is not involved in regulating the steady-state dynamics or deafferentation-induced adaptive changes of cultured dentate granule cell dendrites. These results indicate that drugs targeting neural PAR1-signals may not affect the stability and structural integrity of neuronal networks in healthy brain regions.
Collapse
Affiliation(s)
- Gerlind Schuldt
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Christos Galanis
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Andreas Strehl
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Meike Hick
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Sabine Schiener
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Maximilian Lenz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University FrankfurtFrankfurt, Germany; Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University DüsseldorfDüsseldorf, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Nicola Maggio
- Department of Neurology, The Sagol Center for Neurosciences, Sheba Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel; Talpiot Medical Leadership Program, Department of Neurology and J. Sagol Neuroscience Center, The Chaim Sheba Medical CenterTel HaShomer, Israel; Sagol School of Neuroscience, Tel Aviv UniversityTel Aviv, Israel
| | - Andreas Vlachos
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University FrankfurtFrankfurt, Germany; Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University DüsseldorfDüsseldorf, Germany
| |
Collapse
|
12
|
Nagai T, Nakamuta S, Kuroda K, Nakauchi S, Nishioka T, Takano T, Zhang X, Tsuboi D, Funahashi Y, Nakano T, Yoshimoto J, Kobayashi K, Uchigashima M, Watanabe M, Miura M, Nishi A, Kobayashi K, Yamada K, Amano M, Kaibuchi K. Phosphoproteomics of the Dopamine Pathway Enables Discovery of Rap1 Activation as a Reward Signal In Vivo. Neuron 2016; 89:550-65. [DOI: 10.1016/j.neuron.2015.12.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/17/2015] [Accepted: 12/10/2015] [Indexed: 12/21/2022]
|
13
|
Chen CC, Chu P, Brumberg JC. Experience-dependent regulation of tissue-type plasminogen activator in the mouse barrel cortex. Neurosci Lett 2015; 599:152-7. [PMID: 26021877 DOI: 10.1016/j.neulet.2015.05.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 05/14/2015] [Accepted: 05/22/2015] [Indexed: 11/30/2022]
Abstract
It has been suggested that tissue-type plasminogen activator (tPA), a serine protease, plays a key role in regulating the extracellular matrix core proteins, thereby impacting the structural plasticity in the cerebral cortex. Much is known about its role in regulating plasticity in the visual cortex. However, its permissive role has not been demonstrated to generalize to other cerebral cortical areas. By utilizing a combination of immunofluorescent histochemistry and confocal microscopy, we demonstrate that endogenous tPA is indeed present in the somatosensory cortex, and its expression is experience-dependent. Chronic sensory deprivation induced by whisker trimming from birth for one month leads to increased tPA immunoreactivity in all layers of the barrel cortex. Furthermore, tPA immunoreactivity remains high even after sensation has been restored to the mystacial pad (by allowing whiskers to grow back to full length for one month). Our results suggest that tPA levels in the cerebral cortex are regulated by sensory experience, and play a key role in regulating structural remodeling in the cerebral cortex.
Collapse
Affiliation(s)
- Chia-Chien Chen
- Neuropsychology Subprogram, Psychology, 365th 5th Avenue, New York, NY 10016, USA
| | - Philip Chu
- Neuropsychology Subprogram, Psychology, 365th 5th Avenue, New York, NY 10016, USA
| | - Joshua C Brumberg
- Neuropsychology Subprogram, Psychology, 365th 5th Avenue, New York, NY 10016, USA; Biology-Neuroscience Subprogram, The Graduate Center, CUNY, 365th 5th Avenue, New York, NY 10016, USA; Department of Psychology, Queens College, CUNY, 65-30 Kissena Boulevard, Flushing, NY 11367, USA.
| |
Collapse
|
14
|
Hoirisch-Clapauch S, Mezzasalma MAU, Nardi AE. Pivotal role of tissue plasminogen activator in the mechanism of action of electroconvulsive therapy. J Psychopharmacol 2014; 28:99-105. [PMID: 24113086 DOI: 10.1177/0269881113507639] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Electroconvulsive therapy is an important treatment option for major depressive disorders, acute mania, mood disorders with psychotic features, and catatonia. Several hypotheses have been proposed as electroconvulsive therapy's mechanism of action. Our hypothesis involves many converging pathways facilitated by increased synthesis and release of tissue-plasminogen activator. Human and animal experiments have shown that tissue-plasminogen activator participates in many mechanisms of action of electroconvulsive therapy or its animal variant, electroconvulsive stimulus, including improved N-methyl-D-aspartate receptor-mediated signaling, activation of both brain-derived neurotrophic factor and vascular endothelial growth factor, increased bioavailability of zinc, purinergic release, and increased mobility of dendritic spines. As a result, tissue-plasminogen activator helps promote neurogenesis in limbic structures, modulates synaptic transmission and plasticity, improves cognitive function, and mediates antidepressant effects. Notably, electroconvulsive therapy seems to influence tissue-plasminogen activator metabolism. For example, electroconvulsive stimulus increases the expression of glutamate decarboxylase 65 isoform in γ-aminobutyric acid-releasing neurons, which enhances the release of tissue-plasminogen activator, and the expression of p11, a protein involved in plasminogen and tissue-plasminogen activator assembling. This paper reviews how electroconvulsive therapy correlates with tissue-plasminogen activator. We suggest that interventions aiming at increasing tissue-plasminogen activator levels or its bioavailability - such as daily aerobic exercises together with a carbohydrate-restricted diet, or normalization of homocysteine levels - be evaluated in controlled studies assessing response and remission duration in patients who undergo electroconvulsive therapy.
Collapse
Affiliation(s)
- Silvia Hoirisch-Clapauch
- 1Department of Hematology, Hospital Federal dos Servidores do Estado, Ministry of Health, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
15
|
Abstract
The extracellular matrix (ECM) has a prominent role in brain development, maturation of neural circuits, and adult neuroplasticity. This multifactorial role of the ECM suggests that processes that affect composition or turnover of ECM in the brain could lead to altered brain function, possibly underlying conditions of impaired mental health, such as neuropsychiatric or neurodegenerative disease. In support of this, in the last two decades, clinical and preclinical research provided evidence of correlations and to some degree causal links, between aberrant ECM function and neuropsychiatric disorders, the most prominent being addiction and schizophrenia. Based on these initial observations of involvement of different classes of ECM molecules (laminin, reelin, and their integrin receptors, as well as tenascins and chondroitin sulfate proteoglycans), ECM targets have been suggested as a novel entry point in the treatment of neuropsychiatric disorders. Hence, understanding how ECM molecules contribute to proper neuronal functioning and how this is dysregulated in conditions of mental illness is of pivotal importance. In this chapter, we will review available literature that implicates the different classes of brain ECM molecules in psychiatric disorders, with a primary focus on addiction (opiates, psychostimulants, and alcohol), and we will compare these ECM adaptations with those implicated in schizophrenia and mood disorders.
Collapse
|
16
|
Gieseler F, Ungefroren H, Settmacher U, Hollenberg MD, Kaufmann R. Proteinase-activated receptors (PARs) - focus on receptor-receptor-interactions and their physiological and pathophysiological impact. Cell Commun Signal 2013; 11:86. [PMID: 24215724 PMCID: PMC3842752 DOI: 10.1186/1478-811x-11-86] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/25/2013] [Indexed: 02/07/2023] Open
Abstract
Proteinase-activated receptors (PARs) are a subfamily of G protein-coupled receptors (GPCRs) with four members, PAR1, PAR2, PAR3 and PAR4, playing critical functions in hemostasis, thrombosis, embryonic development, wound healing, inflammation and cancer progression. PARs are characterized by a unique activation mechanism involving receptor cleavage by different proteinases at specific sites within the extracellular amino-terminus and the exposure of amino-terminal “tethered ligand“ domains that bind to and activate the cleaved receptors. After activation, the PAR family members are able to stimulate complex intracellular signalling networks via classical G protein-mediated pathways and beta-arrestin signalling. In addition, different receptor crosstalk mechanisms critically contribute to a high diversity of PAR signal transduction and receptor-trafficking processes that result in multiple physiological effects. In this review, we summarize current information about PAR-initiated physical and functional receptor interactions and their physiological and pathological roles. We focus especially on PAR homo- and heterodimerization, transactivation of receptor tyrosine kinases (RTKs) and receptor serine/threonine kinases (RSTKs), communication with other GPCRs, toll-like receptors and NOD-like receptors, ion channel receptors, and on PAR association with cargo receptors. In addition, we discuss the suitability of these receptor interaction mechanisms as targets for modulating PAR signalling in disease.
Collapse
Affiliation(s)
| | | | | | | | - Roland Kaufmann
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Drackendorfer Str, 1, D-07747, Jena, Germany.
| |
Collapse
|
17
|
Hourai A, Miyata S. Neurogenesis in the circumventricular organs of adult mouse brains. J Neurosci Res 2013; 91:757-70. [PMID: 23526379 DOI: 10.1002/jnr.23206] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 12/10/2012] [Accepted: 12/21/2012] [Indexed: 12/20/2022]
Abstract
The circumventricular organs (CVOs), including the organum vasculosum of the lamina terminalis (OVLT), subfornical organ (SFO), median eminence (ME), and area postrema (AP), allow parenchyma cells to sense a variety of blood-derived substances and/or secreted peptides into blood circulation. In the present study, we examined continuous neurogenesis in the CVOs of adult mice. The immunohistochemistry of neural progenitor cell (NPC) marker proteins revealed that Math1- and Mash1-positive cells were observed in the discrete regions of CVOs, including the capillary plexus in the OVLT, the internal zone of the ME, and the lateral zone in the AP. A few Mash1- and Math1-positive cells were seen throughout the SFO, and many Math1- but not Mash1-positive cells were observed at the arcuate nucleus. Math-positive cells were often seen to localize in close proximity to the vasculature. Bromodeoxyuridine (BrdU) immunohistochemistry revealed the incorporation of BrdU in a subpopulation of Mash1-, Math1-, HuC/D-, and microtubule-associated protein 2 (MAP2)-positive cells. Mash1- and Math1-positive cells expressed exclusively high level of plasminogen, whereas a subpopulation of HuC/D- and MAP2-positive neurons expressed low or undetectable level of plasminogen. Thus, the present study demonstrates that newborn cells express NPC marker proteins and plasminogen to localize closely at vascular matrix and moreover differentiate into neurons expressing mature neuron marker proteins, indicating that new neurons are possibly generated to integrate into new neural circuits.
Collapse
Affiliation(s)
- Atsushi Hourai
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | | |
Collapse
|
18
|
Almonte AG, Qadri LH, Sultan FA, Watson JA, Mount DJ, Rumbaugh G, Sweatt JD. Protease-activated receptor-1 modulates hippocampal memory formation and synaptic plasticity. J Neurochem 2012; 124:109-22. [PMID: 23113835 DOI: 10.1111/jnc.12075] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 09/21/2012] [Accepted: 10/22/2012] [Indexed: 11/28/2022]
Abstract
Protease-activated receptor-1 (PAR1) is an unusual G-protein coupled receptor (GPCR) that is activated through proteolytic cleavage by extracellular serine proteases. Although previous work has shown that inhibiting PAR1 activation is neuroprotective in models of ischemia, traumatic injury, and neurotoxicity, surprisingly little is known about PAR1's contribution to normal brain function. Here, we used PAR1-/- mice to investigate the contribution of PAR1 function to memory formation and synaptic function. We demonstrate that PAR1-/- mice have deficits in hippocampus-dependent memory. We also show that while PAR1-/- mice have normal baseline synaptic transmission at Schaffer collateral-CA1 synapses, they exhibit severe deficits in N-methyl-d-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP). Mounting evidence indicates that activation of PAR1 leads to potentiation of NMDAR-mediated responses in CA1 pyramidal cells. Taken together, this evidence and our data suggest an important role for PAR1 function in NMDAR-dependent processes subserving memory formation and synaptic plasticity.
Collapse
Affiliation(s)
- Antoine G Almonte
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Nakajima A, Kinugasa Y, Torii J, Hishinuma T, Tomioka Y, Yamada K, Yamakuni T. Repeated treatment with nicotine induces phosphorylation of NMDA receptor NR2B subunit in the brain regions involved in behavioral sensitization. Neurosci Lett 2012; 524:133-8. [PMID: 22819974 DOI: 10.1016/j.neulet.2012.07.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/04/2012] [Accepted: 07/11/2012] [Indexed: 10/28/2022]
Abstract
Protein phosphorylation is an important mechanism for the post-translational modulation of N-methyl-d-aspartate (NMDA) receptor functions. In the present study, we investigated the levels of NR2B phosphorylation at Tyr1472 and Ser1303 in the nucleus accumbens, striatum, frontal cortex, and hippocampus of rats that exhibit behavioral sensitization to nicotine. Repeated treatment of rats with nicotine (0.6mg/kg, s.c., for 7 days) produced locomotor sensitization accompanied by increased NR2B phosphorylation at Tyr1472 in the nucleus accumbens and striatum, brain regions involved in behavioral sensitization. In contrast, no changes in NR2B phosphorylation were observed after a single treatment with nicotine in these brain regions. In addition, no changes in NR2B phosphorylation at Ser1303 were observed after repeated treatment with nicotine in any examined brain regions. These results suggest that repeated treatment with nicotine induces NR2B phosphorylation at Tyr1472 in the nucleus accumbens and striatum, which might contribute to the development of synaptic and behavioral plasticity in response to nicotine.
Collapse
Affiliation(s)
- Akira Nakajima
- Department of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Takuma K, Mizoguchi H, Funatsu Y, Hoshina Y, Himeno Y, Fukuzaki E, Kitahara Y, Arai S, Ibi D, Kamei H, Matsuda T, Koike K, Inoue M, Nagai T, Yamada K. Combination of chronic stress and ovariectomy causes conditioned fear memory deficits and hippocampal cholinergic neuronal loss in mice. Neuroscience 2012; 207:261-73. [DOI: 10.1016/j.neuroscience.2012.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 12/28/2011] [Accepted: 01/17/2012] [Indexed: 11/28/2022]
|
21
|
McCoy KL, Gyoneva S, Vellano CP, Smrcka AV, Traynelis SF, Hepler JR. Protease-activated receptor 1 (PAR1) coupling to G(q/11) but not to G(i/o) or G(12/13) is mediated by discrete amino acids within the receptor second intracellular loop. Cell Signal 2012; 24:1351-60. [PMID: 22306780 DOI: 10.1016/j.cellsig.2012.01.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 12/27/2011] [Accepted: 01/19/2012] [Indexed: 12/29/2022]
Abstract
Protease-activated receptor 1 (PAR1) is an unusual GPCR that interacts with multiple G protein subfamilies (G(q/11), G(i/o), and G(12/13)) and their linked signaling pathways to regulate a broad range of pathophysiological processes. However, the molecular mechanisms whereby PAR1 interacts with multiple G proteins are not well understood. Whether PAR1 interacts with various G proteins at the same, different, or overlapping binding sites is not known. Here we investigated the functional and specific binding interactions between PAR1 and representative members of the G(q/11), G(i/o), and G(12/13) subfamilies. We report that G(q/11) physically and functionally interacts with specific amino acids within the second intracellular (i2) loop of PAR1. We identified five amino acids within the PAR1 i2 loop that, when mutated individually, each markedly reduced PAR1 activation of linked inositol phosphate formation in transfected COS-7 cells (functional PAR1-null cells). Among these mutations, only R205A completely abolished direct G(q/11) binding to PAR1 and also PAR1-directed inositol phosphate and calcium mobilization in COS-7 cells and PAR1-/- primary astrocytes. In stark contrast, none of the PAR1 i2 loop mutations disrupted direct PAR1 binding to either G(o) or G(12), or their functional coupling to linked pertussis toxin-sensitive ERK phosphorylation and C3 toxin-sensitive Rho activation, respectively. In astrocytes, our findings suggest that PAR1-directed calcium signaling involves a newly appreciated G(q/11)-PLCε pathway. In summary, we have identified key molecular determinants for PAR1 interactions with G(q/11), and our findings support a model where G(q/11), G(i/o) or G(12/13) each bind to distinct sites within the cytoplasmic regions of PAR1.
Collapse
Affiliation(s)
- Kelly L McCoy
- Department of Pharmacology, O. Wayne Rollins Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
22
|
Bahi A, Dreyer JL. Involvement of tissue plasminogen activator “tPA” in ethanol-induced locomotor sensitization and conditioned-place preference. Behav Brain Res 2012; 226:250-8. [DOI: 10.1016/j.bbr.2011.09.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 08/25/2011] [Accepted: 09/12/2011] [Indexed: 12/26/2022]
|
23
|
Niwa M, Nabeshima T. Roles of a novel molecule 'shati' in the development of methamphetamine-induced dependence. Curr Neuropharmacol 2011; 9:104-8. [PMID: 21886572 PMCID: PMC3137161 DOI: 10.2174/157015911795017362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 04/17/2010] [Accepted: 05/26/2010] [Indexed: 11/22/2022] Open
Abstract
The ability of drugs of abuse to cause dependence can be viewed as a form of neural plasticity. Recently, we have demonstrated that tumor necrosis factor-α (TNF-α) increases dopamine uptake and inhibits methamphetamine-induced dependence. Moreover, we have identified a novel molecule ‘shati’ in the nucleus accumbens of mice treated with methamphetamine using the PCR-select cDNA subtraction method and clarified that it is involved in the development of methamphetamine dependence: Treatment with the shati antisense oligonucleotide (shati-AS), which inhibits the expression of shati mRNA, enhanced the methamphetamine-induced hyperlocomotion, sensitization, and conditioned place preference. Further, blockage of shati mRNA by shati-AS potentiated the methamphetamine-induced increase of dopamine overflow and the methamphetamine-induced decrease in dopamine uptake in the nucleus accumbens. Interestingly, treatment with shati-AS also inhibited expression of TNF-α. Transfection of the vector containing shati cDNA into PC12 cells, dramatically induced the expression of shati and TNF-α mRNA, accelerated dopamine uptake, and inhibited the methamphetamine-induced decrease in dopamine uptake. These effects were blocked by neutralizing TNF-α. These results suggest that the functional roles of shati in methamphetamine-induced behavioral changes are mediated through the induction of TNF-α expression which inhibits the methamphetamine-induced increase of dopamine overflow and decrease in dopamine uptake.
Collapse
Affiliation(s)
- Minae Niwa
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, Nagoya 468-8503, Japan
| | | |
Collapse
|
24
|
Han KS, Mannaioni G, Hamill CE, Lee J, Junge CE, Lee CJ, Traynelis SF. Activation of protease activated receptor 1 increases the excitability of the dentate granule neurons of hippocampus. Mol Brain 2011; 4:32. [PMID: 21827709 PMCID: PMC3170262 DOI: 10.1186/1756-6606-4-32] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 08/10/2011] [Indexed: 11/25/2022] Open
Abstract
Protease activated receptor-1 (PAR1) is expressed in multiple cell types in the CNS, with the most prominent expression in glial cells. PAR1 activation enhances excitatory synaptic transmission secondary to the release of glutamate from astrocytes following activation of astrocytically-expressed PAR1. In addition, PAR1 activation exacerbates neuronal damage in multiple in vivo models of brain injury in a manner that is dependent on NMDA receptors. In the hippocampal formation, PAR1 mRNA appears to be expressed by a subset of neurons, including granule cells in the dentate gyrus. In this study we investigate the role of PAR activation in controlling neuronal excitability of dentate granule cells. We confirm that PAR1 protein is expressed in neurons of the dentate cell body layer as well as in astrocytes throughout the dentate. Activation of PAR1 receptors by the selective peptide agonist TFLLR increased the intracellular Ca2+ concentration in a subset of acutely dissociated dentate neurons as well as non-neuronal cells. Bath application of TFLLR in acute hippocampal slices depolarized the dentate gyrus, including the hilar region in wild type but not in the PAR1-/- mice. PAR1 activation increased the frequency of action potential generation in a subset of dentate granule neurons; cells in which PAR1 activation triggered action potentials showed a significant depolarization. The activation of PAR1 by thrombin increased the amplitude of NMDA receptor-mediated component of EPSPs. These data suggest that activation of PAR1 during normal function or pathological conditions, such as during ischemia or hemorrhage, can increase the excitability of dentate granule cells.
Collapse
Affiliation(s)
- Kyung-Seok Han
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Ito M, Nagai T, Mizoguchi H, Sato K, Hayase M, Otsuka N, Fukakusa A, Kumagai N, Kim HC, Nabeshima T, Takuma K, Yamada K. Activation of post-synaptic dopamine D₁ receptors promotes the release of tissue plasminogen activator in the nucleus accumbens via PKA signaling. J Neurochem 2011; 103:2589-96. [PMID: 17944865 DOI: 10.1111/j.1471-4159.2007.04946.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We have previously demonstrated that tissue plasminogen activator (tPA) plays an important role through the conversion of plasminogen to plasmin in the release of dopamine in the nucleus accumbens (NAc) evoked by depolarization or the systemic administration of drugs of abuse such as morphine and nicotine. In the present study, we examined the mechanisms by which drugs of abuse increase extracellular tPA activity in the NAc in vivo using in situ zymography. The dopamine D(1) receptor (D(1) R) agonist SKF38393, but not D(2) receptor agonist quinpirole, significantly increased extracellular tPA activity in the NAc. The effect of SKF38393 was blocked by pre-treatment with the dopamine D(1) R antagonist SCH23390. Microinjection of Rp-cAMPs, a protein kinase A inhibitor, into the NAc completely blocked the effect of SKF38393. Systemic administration of morphine and methamphetamine increased extracellular tPA activity in the NAc, and these effects were completely blocked by pre-treatment with SCH23390 and raclopride. The results suggest that activation of post-synaptic dopamine D(1) Rs in the NAc leads to an increase in extracellular tPA activity via protein kinase A signaling. Furthermore, dopamine D(2) receptors are also involved in the release of tPA induced by morphine and methamphetamine.
Collapse
Affiliation(s)
- Mina Ito
- Laboratory of Neuropsychopharmacology, Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Almonte AG, Sweatt JD. Serine proteases, serine protease inhibitors, and protease-activated receptors: roles in synaptic function and behavior. Brain Res 2011; 1407:107-22. [PMID: 21782155 DOI: 10.1016/j.brainres.2011.06.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 06/03/2011] [Accepted: 06/16/2011] [Indexed: 12/11/2022]
Abstract
Serine proteases, serine protease inhibitors, and protease-activated receptors have been intensively investigated in the periphery and their roles in a wide range of processes-coagulation, inflammation, and digestion, for example-have been well characterized (see Coughlin, 2000; Macfarlane et al., 2001; Molinari et al., 2003; Wang et al., 2008; Di Cera, 2009 for reviews). A growing number of studies demonstrate that these protein systems are widely expressed in many cell types and regions in mammalian brains. Accumulating lines of evidence suggest that the brain has co-opted the activities of these interesting proteins to regulate various processes underlying synaptic activity and behavior. In this review, we discuss emerging roles for serine proteases in the regulation of mechanisms underlying synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Antoine G Almonte
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
27
|
Neuronal protease-activated receptor 1 drives synaptic retrograde signaling mediated by the endocannabinoid 2-arachidonoylglycerol. J Neurosci 2011; 31:3104-9. [PMID: 21414931 DOI: 10.1523/jneurosci.6000-10.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Protease-activated receptor 1 (PAR1) is a member of the G-protein coupled receptors that are proteolytically activated by serine proteases. Recent studies suggest a definite contribution of PAR1 to brain functions, including learning and memory. However, cellular mechanisms by which PAR1 activation influences neuronal activity are not well understood. Here we show that PAR1 activation drives retrograde endocannabinoid signaling and thereby regulates synaptic transmission. In cultured hippocampal neurons from rat, PAR1 activation by thrombin or PAR1-specific peptide agonists transiently suppressed inhibitory transmission at cannabinoid-sensitive, but not cannabinoid-insensitive, synapses. The PAR1-induced suppression of synaptic transmission was accompanied by an increase in paired-pulse ratio, and was blocked by a cannabinoid CB(1) receptor antagonist. The PAR1-induced suppression was blocked by pharmacological inhibition of postsynaptic diacylglycerol lipase (DGL), a key enzyme for biosynthesis of the major endocannabinoid 2-arachidonoylglycerol (2-AG), and was absent in knock-out mice lacking the α isoform of DGL. The PAR1-induced IPSC suppression remained intact under the blockade of metabotropic glutamate receptors and was largely resistant to the treatment that blocked Ca(2+) elevation in glial cells following PAR1 activation, which excludes the major contribution of glial PAR1 in IPSC suppression. We conclude that activation of neuronal PAR1 triggers retrograde signaling mediated by 2-AG, which activates presynaptic CB(1) receptors and suppresses transmitter release at hippocampal inhibitory synapses.
Collapse
|
28
|
Adams MN, Ramachandran R, Yau MK, Suen JY, Fairlie DP, Hollenberg MD, Hooper JD. Structure, function and pathophysiology of protease activated receptors. Pharmacol Ther 2011; 130:248-82. [PMID: 21277892 DOI: 10.1016/j.pharmthera.2011.01.003] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/03/2011] [Indexed: 12/18/2022]
Abstract
Discovered in the 1990s, protease activated receptors(1) (PARs) are membrane-spanning cell surface proteins that belong to the G protein coupled receptor (GPCR) family. A defining feature of these receptors is their irreversible activation by proteases; mainly serine. Proteolytic agonists remove the PAR extracellular amino terminal pro-domain to expose a new amino terminus, or tethered ligand, that binds intramolecularly to induce intracellular signal transduction via a number of molecular pathways that regulate a variety of cellular responses. By these mechanisms PARs function as cell surface sensors of extracellular and cell surface associated proteases, contributing extensively to regulation of homeostasis, as well as to dysfunctional responses required for progression of a number of diseases. This review examines common and distinguishing structural features of PARs, mechanisms of receptor activation, trafficking and signal termination, and discusses the physiological and pathological roles of these receptors and emerging approaches for modulating PAR-mediated signaling in disease.
Collapse
Affiliation(s)
- Mark N Adams
- Mater Medical Research Institute, Aubigny Place, Raymond Terrace, South Brisbane Qld 4101, Australia
| | | | | | | | | | | | | |
Collapse
|
29
|
Taniguchi Y, Inoue N, Morita S, Nikaido Y, Nakashima T, Nagai N, Okada K, Matsuo O, Miyata S. Localization of plasminogen in mouse hippocampus, cerebral cortex, and hypothalamus. Cell Tissue Res 2010; 343:303-17. [PMID: 21190118 DOI: 10.1007/s00441-010-1110-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 11/29/2010] [Indexed: 01/15/2023]
Abstract
Although the tissue plasminogen activator/plasminogen system contributes to numerous brain functions, such as learning, memory, and anxiety behavior, little attention has as yet been given to the localization of plasminogen in the brain. We have investigated the localization of plasminogen in the adult mouse brain by using immunohistochemistry. In the hippocampus, plasminogen immunoreactivity was seen in the pyramidal cell layer as numerous punctate structures in neuronal somata. An electron-microscopic study further demonstrated that the plasminogen-immunoreactive punctate structures represented secretory vesicles and/or vesicle clusters. In the cerebral cortex, plasminogen immunoreactivity was evident in the somata of the layer II/III and V neurons. A quantitative analysis revealed that parvalbumin (PV)-positive neurons had more plasminogen-immunoreactive puncta compared with those of PV-negative neurons in the hippocampus and cerebral cortex. Plasminogen immunoreactivity was present throughout the hypothalamus, being particularly prominent in the neuronal somata of the organum vasculosum laminae terminalis, ventromedial preoptic nucleus, supraoptic nucleus, subfornical organ, medial part of the paraventricular nucleus (PVN), posterior part of the PVN, and arcuate hypothalamic nucleus. Thus, plasminogen is highly expressed in specific populations of hippocampal, cortical, and hypothalamic neurons, and plasminogen-containing vesicles are mainly observed at neuronal somata.
Collapse
Affiliation(s)
- Yuki Taniguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Biphasic regulation of tissue plasminogen activator activity in ischemic rat brain and in cultured neural cells: essential role of astrocyte-derived plasminogen activator inhibitor-1. Neurochem Int 2010; 58:423-33. [PMID: 21193004 DOI: 10.1016/j.neuint.2010.12.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/21/2010] [Accepted: 12/21/2010] [Indexed: 11/20/2022]
Abstract
In brain, the serine protease tissue plasminogen activator (tPA) and its endogenous inhibitor plasminogen activator inhibitor-1 (PAI-1) have been implicated in the regulation of various neurophysiological and pathological responses. In this study, we investigated the differential role of neurons and astrocytes in the regulation of tPA/PAI-1 activity in ischemic brain. The activity of tPA peaked transiently and then decreased in cortex and striatum along with delayed induction of PAI-1 in the inflammatory stage after MCAO/reperfusion injury. In cultured primary cells, glutamate stimulation increased tPA activity in neurons but not in other cells such as microglia and astrocytes. With LPS stimulation, a model of neuroinflammatory insults, robust PAI-1 induction was observed in astrocytes but not in neurons and microglia. The upregulation of PAI-1 by LPS in astrocytes was also verified by RT-PCR analysis as well as PAI-1 promoter reporter assay. Lastly, we checked the effects of hypoxia on tPA/PAI-1 activity. Hypoxia increased tPA release from neurons without effects on microglia, while the activity of tPA in astrocyte was decreased consistent with increased PAI-1 activity in astrocyte. Taken together, the results from the present study suggest that neurons are the major source of tPA and that the glutamate-induced stimulated release is mainly governed by neurons in the acute phase. In contrast, the massive up-regulation of PAI-1 in astrocytes during subchronic and chronic inflammatory conditions, leads to decreased tPA activity in the later stages of MCAO. Differential regulation of tPA and PAI-1 in neurons, astrocytes and microglia suggest more attention is required to understand the role of local tPA activity in the vicinity of individual cell types.
Collapse
|
31
|
Imamura Y, Morita S, Nakatani Y, Okada K, Ueshima S, Matsuo O, Miyata S. Tissue plasminogen activator and plasminogen are critical for osmotic homeostasis by regulating vasopressin secretion. J Neurosci Res 2010; 88:1995-2006. [PMID: 20175210 DOI: 10.1002/jnr.22370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Systemic osmotic homeostasis is regulated mainly by neuroendocrine system of arginine-vasopressin (AVP) in mammalians. In the present study, we demonstrated that the immunoreactivity of tissue plasminogen activator (tPA) was observed specifically at neurosecretory granules of AVP-positive magnocellular terminals and that of plasminogen was seen at astrocytes in the neurohypophysis (NH). Both tPA and plasminogen knockout (KO) mice revealed higher plasma osmolarity upon water deprivation, a chronic osmotic stimulation, as compared with their wild-type (WT) animals, indicating abnormal osmotic control in these KO mice. tPA KO mice but not plasminogen ones revealed lower ability in secreting AVP into the blood circulation upon an acute osmotic stimulation. Both tPA and plasminogen KO animals showed lower ability in secreting AVP into the blood circulation upon a chronic osmotic stimulation. The recombinant tPA was able to promote the release of AVP from isolated NH. Chronic osmotic stimulation decreased the laminin expression level of neurohypophysial microvessel in WT mice but not in plasminogen KO ones. We suggest that AVP secretion is critically regulated by tPA-dependent facilitation of AVP release from terminals and plasminogen-dependent increase of AVP permeability across microvessels possibly via laminin degradation.
Collapse
Affiliation(s)
- Yuhki Imamura
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
The tissue plasminogen activator/plasmin system may act through cleavage of pro-BDNF to increase risk of substance abuse. CNS Spectr 2010; 15:350. [PMID: 20629271 DOI: 10.1017/s1092852900000328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
33
|
Nagai N, Matsuo O. Roles of fibrinolytic system components in the nervous system. PATHOPHYSIOLOGY 2010; 17:141-7. [DOI: 10.1016/j.pathophys.2009.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 03/10/2009] [Accepted: 03/23/2009] [Indexed: 10/20/2022] Open
|
34
|
Park GH, Jeon SJ, Ko HM, Ryu JR, Lee JM, Kim HY, Han SH, Kang YS, Park SH, Shin CY, Ko KH. Activation of microglial cells via protease-activated receptor 2 mediates neuronal cell death in cultured rat primary neuron. Nitric Oxide 2010; 22:18-29. [DOI: 10.1016/j.niox.2009.10.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 10/06/2009] [Accepted: 10/22/2009] [Indexed: 11/30/2022]
|
35
|
Lohman RJ, Jones NC, O’Brien TJ, Cocks TM. A regulatory role for protease-activated receptor-2 in motivational learning in rats. Neurobiol Learn Mem 2009; 92:301-9. [DOI: 10.1016/j.nlm.2009.03.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 02/19/2009] [Accepted: 03/25/2009] [Indexed: 11/25/2022]
|
36
|
Essential role of mitogen-activated protein kinase pathways in protease activated receptor 2-mediated nitric-oxide production from rat primary astrocytes. Nitric Oxide 2009; 21:110-9. [PMID: 19527794 DOI: 10.1016/j.niox.2009.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Revised: 05/20/2009] [Accepted: 05/26/2009] [Indexed: 11/24/2022]
Abstract
Protease-activated receptors (PARs) play important roles in the regulation of brain function such as neuroinflammation by transmitting the signal from proteolytic enzymes such as thrombin and trypsin. We and others have reported that a member of the family, PAR-2 is activated by trypsin, whose involvement in the neurophysiological process is increasingly evident, and is involved in the neuroinflammatory processes including morphological changes of astrocytes. In this study, we investigated the role of PAR-2 in the production of nitric oxide (NO) in rat primary astrocytes. Treatment of PAR-2 agonist trypsin increased NO production in a dose-dependent manner, which was mediated by the induction of inducible nitric-oxide synthase. The trypsin-mediated production of NO was mimicked by PAR-2 agonist peptide and reduced by either pharmacological PAR-2 antagonist peptide or by siRNA-mediated inhibition of PAR-2 expression, which suggests the critical role of PAR-2 in this process. NO production by PAR-2 was mimicked by PMA, a PKC activator, and was attenuated by Go6976, a protein kinase C (PKC) inhibitor. PAR-2 stimulation activated three subtypes of mitogen-activated protein kinases (MAPKs): extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 MAPK. NO production by PAR-2 was blocked by inhibition of ERK, p38, and JNK pathways. PAR-2 stimulation also activated nuclear factor-kappaB (NF-kappaB) DNA binding and transcriptional activity as well as IkappaBalpha phosphorylation. Inhibitors of NF-kappaB pathway inhibited PAR-2-mediated NO production. In addition, inhibitors of MAPK pathways prevented transcriptional activation of NF-kappaB reporter constructs. These results suggest that PAR-2 activation-mediated NO production in astrocytes is transduced by the activation of MAPKs followed by NF-kappaB pathways.
Collapse
|
37
|
Tissue plasminogen activator modulates the cellular and behavioral response to cocaine. Proc Natl Acad Sci U S A 2009; 106:1983-8. [PMID: 19181855 DOI: 10.1073/pnas.0812491106] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cocaine exposure induces long-lasting molecular and structural adaptations in the brain. In this study, we show that tissue plasminogen activator (tPA), an extracellular protease involved in neuronal plasticity, modulates the biochemical and behavioral response to cocaine. When injected in the acute binge paradigm, cocaine enhanced tPA activity in the amygdala, which required activation of corticotropin-releasing factor type-1 (CRF-R1) receptors. Compared with WT mice, tPA-/- mice injected with cocaine displayed attenuated phosphorylation of ERK, cAMP response element binding protein (CREB), and dopamine and cAMP-regulated phosphoprotein 32 kDa (DARPP-32) and blunted induction of immediate early genes (IEGs) c-Fos, Egr-1, and Homer 1a in the amygdala and the nucleus accumbens (NAc). tPA-/- mice also displayed significantly higher basal preprodynorphin (ppDyn) mRNA levels in the NAc in comparison to WT mice, and cocaine decreased ppDyn mRNA levels in tPA-/- mice only. Cocaine-induced locomotor sensitization and conditioned place preference (CPP) were attenuated in tPA-/- mice. Cocaine exposure also had an anxiolytic effect in tPA-/- but not WT mice. These results identify tPA as an important and novel component of the signaling pathway that modulates cocaine-induced changes in neuroadaptation and behavior.
Collapse
|
38
|
Abstract
Drug addiction/dependence is defined as a chronically relapsing disorder that is characterized by compulsive drug taking, inability to limit intake, and intense drug cravings. The positive reinforcing/rewarding effects of drugs primarily depend on the mesocorticolimbic dopamine system innervating the nucleus accumbens while the craving for drugs is associated with activation of the prefrontal cortex. The chronic intake of drugs causes homeostatic molecular and functional changes in synapses, which may be critically associated with the development of drug dependence. Recent studies have demonstrated that various cytokines and proteinases are produced in the brain on treatment with drugs of abuse, and play a role in drug dependence. These endogenous modulators of drug dependence are classified into two groups, pro-addictive and anti-addictive factors. The former including basic fibroblast growth factor, brain-derived neurotrophic factor, tissue plasminogen activator, matrix metalloproteinase (MMP)-2 and MMP-9 act to potentiate the rewarding effects of drugs, while the latter such as tumor necrosis factor-alpha and glial cell line-derived neurotrophic factor reduce the reward. These findings suggest that an imbalance between pro-addictive and anti-addictive factors contributes to the development and relapse of drug dependence. Furthermore, targeting these endogenous modulators would provide new therapeutic approaches to the treatment of drug dependence.
Collapse
Affiliation(s)
- Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan.
| |
Collapse
|
39
|
The role of tissue-type plasminogen activator system in amphetamine-induced conditional place preference extinction and reinstatement. Neuropsychopharmacology 2008; 33:2726-34. [PMID: 18256596 DOI: 10.1038/sj.npp.1301668] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Extracellular serine proteases of the plasminogen activator family (tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) may modulate synaptic adhesion and associate with learning behavior. Psychostimulants strongly induce their expression in the mesolimbic dopaminergic pathway, but cocaine preferentially induces uPA, whereas morphine and amphetamine preferentially induce tPA. tPA-expressing animals displayed enhanced conditional place preference (CPP) for amphetamine compared with uPA-overexpressing animals. Thus, modulation of the plasminogen system in the brain might be a potential target against drugs of abuse. In the present study, we aim to identify whether tPA is involved in the acquisition/learning phase or in the expression/retrieval phase of conditioned drug preference. For this purpose, animals were injected with lentiviruses expressing or silencing tPA in the NAc and place preference was assessed. We found that tPA expression is associated with acquisition of place preference and animals overexpressing tPA spend >87% of the time in the drug-associated compartment, compared with 60% for control animals. When ectopic expression of tPA has been inhibited by doxycycline during acquisition, animals do no more associate the environment with the drug. Suppression of endogenous tPA expression in animals treated with LV-siRNA fully suppresses place preference, and these animals appear to avoid the drug-associated box. tPA overexpression delays extinction, but priming with low doses of amphetamine reinstates place preference even after full extinction. Together, these data clearly indicate that tPA plays an important role in acquisition of amphetamine-induced CPP, but its role in CPP expression does not seem important.
Collapse
|
40
|
Bahi A, Kusnecov AW, Dreyer JL. Effects of urokinase-type plasminogen activator in the acquisition, expression and reinstatement of cocaine-induced conditioned-place preference. Behav Brain Res 2008; 191:17-25. [DOI: 10.1016/j.bbr.2008.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Revised: 02/22/2008] [Accepted: 03/03/2008] [Indexed: 10/22/2022]
|
41
|
Mannaioni G, Orr AG, Hamill CE, Yuan H, Pedone KH, McCoy KL, Berlinguer Palmini R, Junge CE, Lee CJ, Yepes M, Hepler JR, Traynelis SF. Plasmin potentiates synaptic N-methyl-D-aspartate receptor function in hippocampal neurons through activation of protease-activated receptor-1. J Biol Chem 2008; 283:20600-11. [PMID: 18474593 DOI: 10.1074/jbc.m803015200] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protease-activated receptor-1 (PAR1) is activated by a number of serine proteases, including plasmin. Both PAR1 and plasminogen, the precursor of plasmin, are expressed in the central nervous system. In this study we examined the effects of plasmin in astrocyte and neuronal cultures as well as in hippocampal slices. We find that plasmin evokes an increase in both phosphoinositide hydrolysis (EC(50) 64 nm) and Fura-2/AM fluorescence (195 +/- 6.7% above base line, EC(50) 65 nm) in cortical cultured murine astrocytes. Plasmin also activates extracellular signal-regulated kinase (ERK1/2) within cultured astrocytes. The plasmin-induced rise in intracellular Ca(2+) concentration ([Ca(2+)](i)) and the increase in phospho-ERK1/2 levels were diminished in PAR1(-/-) astrocytes and were blocked by 1 microm BMS-200261, a selective PAR1 antagonist. However, plasmin had no detectable effect on ERK1/2 or [Ca(2+)](i) signaling in primary cultured hippocampal neurons or in CA1 pyramidal cells in hippocampal slices. Plasmin (100-200 nm) application potentiated the N-methyl-D-aspartate (NMDA) receptor-dependent component of miniature excitatory postsynaptic currents recorded from CA1 pyramidal neurons but had no effect on alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate- or gamma-aminobutyric acid receptor-mediated synaptic currents. Plasmin also increased NMDA-induced whole cell receptor currents recorded from CA1 pyramidal cells (2.5 +/- 0.3-fold potentiation over control). This effect was blocked by BMS-200261 (1 microm; 1.02 +/- 0.09-fold potentiation over control). These data suggest that plasmin may serve as an endogenous PAR1 activator that can increase [Ca(2+)](i) in astrocytes and potentiate NMDA receptor synaptic currents in CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Guido Mannaioni
- Dipartimento di Farmacologia, Università degli Studi di Firenze, Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mizoguchi H, Yamada K, Nabeshima T. Neuropsychotoxicity of abused drugs: involvement of matrix metalloproteinase-2 and -9 and tissue inhibitor of matrix metalloproteinase-2 in methamphetamine-induced behavioral sensitization and reward in rodents. J Pharmacol Sci 2008; 106:9-14. [PMID: 18198472 DOI: 10.1254/jphs.fm0070139] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) function to remodel the pericellular environment. We have investigated the role of the MMP/TIMP system in methamphetamine (METH) dependence in rodents, in which the remodeling of neural circuits may be crucial. Repeated METH treatment induced behavioral sensitization, which was accompanied by an increase in MMP-2/-9/TIMP-2 activity in the brain. An antisense TIMP-2 oligonucleotide enhanced the sensitization, which was associated with a potentiation of the METH-induced release of dopamine in the nucleus accumbens (NAc). MMP-2/-9 inhibitors blocked the METH-induced behavioral sensitization and conditioned place preference (CPP), a measure of the rewarding effect of a drug, and reduced the METH-increased dopamine release in the NAc. In MMP-2- and MMP-9-deficient mice, METH-induced behavioral sensitization and CPP as well as dopamine release were attenuated. The MMP/TIMP system may be involved in METH-induced sensitization and reward by regulating extracellular dopamine levels.
Collapse
Affiliation(s)
- Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | |
Collapse
|
43
|
Fukakusa A, Mizoguchi H, Koike H, Nabeshima T, Takuma K, Yamada K. Tissue Plasminogen Activator Is Not Involved in Methamphetamine-Induced Neurotoxicity. J Pharmacol Sci 2008; 106:321-4. [DOI: 10.1254/jphs.sc0070328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
44
|
Nagai T, Nabeshima T, Yamada K. Basic and Translational Research on Proteinase-Activated Receptors: Regulation of Nicotine Reward by the Tissue Plasminogen Activator (tPA) – Plasmin System via Proteinase-Activated Receptor 1. J Pharmacol Sci 2008; 108:408-14. [DOI: 10.1254/jphs.08r04fm] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
45
|
Proteinases and signalling: pathophysiological and therapeutic implications via PARs and more. Br J Pharmacol 2007; 153 Suppl 1:S263-82. [PMID: 18059329 DOI: 10.1038/sj.bjp.0707507] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Proteinases like thrombin, trypsin and tissue kallikreins are now known to regulate cell signaling by cleaving and activating a novel family of G-protein-coupled proteinase-activated receptors (PARs 1-4) via exposure of a tethered receptor-triggering ligand. On their own, short synthetic PAR-selective PAR-activating peptides (PAR-APs) mimicking the tethered ligand sequences can activate PARs 1, 2 and 4 and cause physiological responses both in vitro and in vivo. Using the PAR-APs as sentinel probes in vivo, it has been found that PAR activation can affect the vascular, renal, respiratory, gastrointestinal, musculoskeletal and nervous systems (both central and peripheral nervous system) and can promote cancer metastasis and invasion. In general, responses triggered by PARs 1, 2 and 4 are in keeping with an innate immune inflammatory response, ranging from vasodilatation to intestinal inflammation, increased cytokine production and increased or decreased nociception. Further, PARs have been implicated in a number of disease states, including cancer and inflammation of the cardiovascular, respiratory, musculoskeletal, gastrointestinal and nervous systems. In addition to activating PARs, proteinases can cause hormone-like effects by other signalling mechanisms, like growth factor receptor activation, that may be as important as the activation of PARs. We, therefore, propose that the PARs themselves, their activating serine proteinases and their associated signalling pathways can be considered as attractive targets for therapeutic drug development. Thus, proteinases in general must now be considered as 'hormone-like' messengers that can signal either via PARs or other mechanisms.
Collapse
|
46
|
Fukakusa A, Nagai T, Mizoguchi H, Otsuka N, Kimura H, Kamei H, Kim HC, Nabeshima T, Takuma K, Yamada K. Role of tissue plasminogen activator in the sensitization of methamphetamine-induced dopamine release in the nucleus accumbens. J Neurochem 2007; 105:436-44. [PMID: 18036193 DOI: 10.1111/j.1471-4159.2007.05142.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have previously demonstrated that repeated, but not acute, methamphetamine (METH) treatment increases tissue plasminogen activator (tPA) activity in the brain, which is associated with the development of behavioral sensitization to METH. In this study, we investigated whether the tPA-plasmin system is involved in the development of sensitization in METH-induced dopamine release in the nucleus accumbens (NAc). There was no difference in acute METH-induced increase in extracellular dopamine levels in the NAc between wild-type and tPA-deficient (tPA-/-) mice. Repeated METH treatment resulted in a significant enhancement of METH- induced dopamine release in wild-type mice, but not tPA-/- mice. Microinjection of exogenous tPA or plasmin into the NAc of wild-type mice significantly potentiated acute METH- induced dopamine release. Degradation of laminin was evident in brain tissues incubated with tPA plus plasminogen or plasmin in vitro although tPA or plasminogen alone had no effect. Immunohistochemical analysis revealed that microinjection of plasmin into the NAc reduced laminin immunoreactivity without neuronal damage. Our findings suggest that the tPA-plasmin system participates in the development of behavioral sensitization induced by repeated METH treatment, by regulating the processes underlying the sensitization of METH-induced dopamine release in the NAc, in which degradation of laminin by plasmin may play a role.
Collapse
Affiliation(s)
- Ayumi Fukakusa
- Laboratory of Neuropsychopharmacology, Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Almonte AG, Hamill CE, Chhatwal JP, Wingo TS, Barber JA, Lyuboslavsky PN, David Sweatt J, Ressler KJ, White DA, Traynelis SF. Learning and memory deficits in mice lacking protease activated receptor-1. Neurobiol Learn Mem 2007; 88:295-304. [PMID: 17544303 PMCID: PMC2040495 DOI: 10.1016/j.nlm.2007.04.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 03/30/2007] [Accepted: 04/02/2007] [Indexed: 11/18/2022]
Abstract
The roles of serine proteases and protease activated receptors have been extensively studied in coagulation, wound healing, inflammation, and neurodegeneration. More recently, serine proteases have been suggested to influence synaptic plasticity. In this context, we examined the role of protease activated receptor 1 (PAR1), which is activated following proteolytic cleavage by thrombin and plasmin, in emotionally motivated learning. We were particularly interested in PAR1 because its activation enhances the function of NMDA receptors, which are required for some forms of synaptic plasticity. We examined several baseline behavioral measures, including locomotor activity, expression of anxiety-like behavior, motor task acquisition, nociceptive responses, and startle responses in C57Bl/6 mice in which the PAR1 receptor has been genetically deleted. In addition, we evaluated learning and memory in these mice using two memory tasks, passive avoidance and cued fear-conditioning. Whereas locomotion, pain response, startle, and measures of baseline anxiety were largely unaffected by PAR1 removal, PAR1-/- animals showed significant deficits in a passive avoidance task and in cued fear conditioning. These data suggest that PAR1 may play an important role in emotionally motivated learning.
Collapse
Affiliation(s)
- Antoine G Almonte
- Department of Pharmacology, Emory University, School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tzschentke TM. Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol 2007; 12:227-462. [PMID: 17678505 DOI: 10.1111/j.1369-1600.2007.00070.x] [Citation(s) in RCA: 1031] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conditioned place preference (CPP) continues to be one of the most popular models to study the motivational effects of drugs and non-drug treatments in experimental animals. This is obvious from a steady year-to-year increase in the number of publications reporting the use this model. Since the compilation of the preceding review in 1998, more than 1000 new studies using place conditioning have been published, and the aim of the present review is to provide an overview of these recent publications. There are a number of trends and developments that are obvious in the literature of the last decade. First, as more and more knockout and transgenic animals become available, place conditioning is increasingly used to assess the motivational effects of drugs or non-drug rewards in genetically modified animals. Second, there is a still small but growing literature on the use of place conditioning to study the motivational aspects of pain, a field of pre-clinical research that has so far received little attention, because of the lack of appropriate animal models. Third, place conditioning continues to be widely used to study tolerance and sensitization to the rewarding effects of drugs induced by pre-treatment regimens. Fourth, extinction/reinstatement procedures in place conditioning are becoming increasingly popular. This interesting approach is thought to model certain aspects of relapse to addictive behavior and has previously almost exclusively been studied in drug self-administration paradigms. It has now also become established in the place conditioning literature and provides an additional and technically easy approach to this important phenomenon. The enormous number of studies to be covered in this review prevented in-depth discussion of many methodological, pharmacological or neurobiological aspects; to a large extent, the presentation of data had to be limited to a short and condensed summary of the most relevant findings.
Collapse
Affiliation(s)
- Thomas M Tzschentke
- Grünenthal GmbH, Preclinical Research and Development, Department of Pharmacology, Aachen, Germany.
| |
Collapse
|
49
|
Niwa M, Nitta A, Mizoguchi H, Ito Y, Noda Y, Nagai T, Nabeshima T. A novel molecule "shati" is involved in methamphetamine-induced hyperlocomotion, sensitization, and conditioned place preference. J Neurosci 2007; 27:7604-15. [PMID: 17626222 PMCID: PMC6672622 DOI: 10.1523/jneurosci.1575-07.2007] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Drug addiction places an enormous burden on society through its repercussions on crime rate and healthcare. Repeated exposure to drugs of abuse causes cellular adaptations in specific neuronal populations that ultimately can lead to a state of addiction. In the present study, we have identified a novel molecule "shati" from the nucleus accumbens (NAc) of mice treated with methamphetamine (METH) using the PCR-select complementary DNA subtraction method. Moreover, we investigated whether shati is involved in METH-induced hyperlocomotion, sensitization, and conditioned place preference (CPP). METH induced expression of shati mRNA dose dependently via dopamine (DA) receptors. We prepared antibodies against shati and, using them, found shati to be expressed in neuronal cells of the mouse brain. Treatment with the shati antisense oligonucleotide (shati-AS), which significantly inhibited the expression of shati mRNA, enhanced the acute METH response, METH-induced behavioral sensitization, and CPP. Blockage of shati mRNA by shati-AS potentiated the METH-induced increase of DA overflow in the NAc and the METH-induced decrease in synaptosomal and vesicular DA uptake in the midbrain. These results suggest that a novel molecule shati is involved in the development of METH-induced hyperlocomotion, sensitization, and CPP. The functional roles of shati in METH-regulated behavioral alternations are likely to be mediated by its inhibitory effects on the METH-induced increase of DA overflow in the NAc and the METH-induced decrease in DA uptake in the midbrain.
Collapse
Affiliation(s)
- Minae Niwa
- Department of Neuropsychopharmacology and Hospital Pharmacy and
- Department of Chemical Pharmacology, Meijo University Graduate School of Pharmaceutical Sciences, Nagoya 468-8503, Japan
| | - Atsumi Nitta
- Department of Neuropsychopharmacology and Hospital Pharmacy and
| | | | - Yasutomo Ito
- Equipment Center for Research and Education, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan, and
| | - Yukihiro Noda
- Department of Neuropsychopharmacology and Hospital Pharmacy and
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy and
| | - Toshitaka Nabeshima
- Department of Neuropsychopharmacology and Hospital Pharmacy and
- Department of Chemical Pharmacology, Meijo University Graduate School of Pharmaceutical Sciences, Nagoya 468-8503, Japan
| |
Collapse
|
50
|
Ito M, Nagai T, Mizoguchi H, Fukakusa A, Nakanishi Y, Kamei H, Nabeshima T, Takuma K, Yamada K. Possible involvement of protease-activated receptor-1 in the regulation of morphine-induced dopamine release and hyperlocomotion by the tissue plasminogen activator-plasmin system. J Neurochem 2007; 101:1392-9. [PMID: 17286591 DOI: 10.1111/j.1471-4159.2006.04423.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We have previously demonstrated that tissue plasminogen activator (tPA)-plasmin system participates in the rewarding effect of morphine, by regulating dopamine release in the nucleus accumbens (NAc). However, it is unclear how plasmin increases the morphine-induced release of dopamine and hyperlocomotion. In the present study we investigated whether protease activated receptor-1 (PAR-1) is involved in the regulation of acute morphine-induced dopamine release by the tPA-plasmin system. Morphine significantly but transiently increased extracellular tPA activity in the NAc, which was completely blocked by naloxone. Microinjection of a PAR-1 antagonist, (tyr(-1))-thrombin receptor activating peptide 7, into the NAc significantly reduced morphine-induced dopamine release in the NAc and hyperlocomotion although the treatment had no effect on basal dopamine release and spontaneous locomotor activity. Furthermore, the PAR-1 antagonist blocked the ameliorating effect of plasmin on the defect of morphine-induced dopamine release in the NAc of tPA-deficient mice. In contrast, intracerebroventricular injection of the PAR-1 antagonist had no effect on the antinociceptive effects of morphine in mice. These results suggest that PAR-1 is a target for the tPA-plasmin system in the regulation of acute morphine-induced dopamine release in the NAc.
Collapse
Affiliation(s)
- Mina Ito
- Laboratory of Neuropsychopharmacology, Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|