1
|
Lei L, Wang YF, Chen CY, Wang YT, Zhang Y. Novel insight into astrocyte-mediated gliotransmission modulates the synaptic plasticity in major depressive disorder. Life Sci 2024; 355:122988. [PMID: 39153595 DOI: 10.1016/j.lfs.2024.122988] [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: 04/25/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Major depressive disorder (MDD) is a form of glial cell-based synaptic dysfunction disease in which glial cells interact closely with neuronal synapses and perform synaptic information processing. Glial cells, particularly astrocytes, are active components of the brain and are responsible for synaptic activity through the release gliotransmitters. A reduced density of astrocytes and astrocyte dysfunction have both been identified the brains of patients with MDD. Furthermore, gliotransmission, i.e., active information transfer mediated by gliotransmitters between astrocytes and neurons, is thought to be involved in the pathogenesis of MDD. However, the mechanism by which astrocyte-mediated gliotransmission contributes to depression remains unknown. This review therefore summarizes the alterations in astrocytes in MDD, including astrocyte marker, connexin 43 (Cx43) expression, Cx43 gap junctions, and Cx43 hemichannels, and describes the regulatory mechanisms of astrocytes involved in synaptic plasticity. Additionally, we investigate the mechanisms acting of the glutamatergic, gamma-aminobutyric acidergic, and purinergic systems that modulate synaptic function and the antidepressant mechanisms of the related receptor antagonists. Further, we summarize the roles of glutamate, gamma-aminobutyric acid, d-serine, and adenosine triphosphate in depression, providing a basis for the identification of diagnostic and therapeutic targets for MDD.
Collapse
Affiliation(s)
- Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
2
|
Donoso MV, Catalán-Salas V, Pulgar-Sepúlveda R, Eugenín J, Huidobro-Toro JP. Physiology, Pathophysiology and Clinical Relevance of D-Amino Acids Dynamics: From Neurochemistry to Pharmacotherapy. CHEM REC 2024; 24:e202400013. [PMID: 39318079 DOI: 10.1002/tcr.202400013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/18/2024] [Indexed: 09/26/2024]
Abstract
Over three decades ago, two independent groups of investigators identified free D-aspartic and later D-serine in specific brain nuclei and endocrine glands. This finding revealed a novel, non-proteinogenic role of these molecules. Moreover, the finding that aged proteins from the human eye crystallin, teeth, bone, blood vessels or the brain incorporate D-aspartic acids to specific primary protein sequences fostered the hypothesis that aging might be related to D-amino acid isomerization of body proteins. The experimental confirmation that schizophrenia and neurodegenerative diseases modify plasma free D-amino acids or tissue levelsnurtured the opportunity of using D-amino acids as therapeutic agents for several disease treatments, a strategy that prompted the successful current application of D-amino acids to human medicine.
Collapse
Affiliation(s)
- M Verónica Donoso
- Pharmacology Laboratory, Department Biology, Faculty of Chemistry and Biology, Centro Desarrollo de Nanociencias y Nanotecnología (CEDENNA), Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile
| | - Vicente Catalán-Salas
- Pharmacology Laboratory, Department Biology, Faculty of Chemistry and Biology, Centro Desarrollo de Nanociencias y Nanotecnología (CEDENNA), Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile
| | - Raúl Pulgar-Sepúlveda
- Neural System Laboratory, Department Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile
| | - Jaime Eugenín
- Neural System Laboratory, Department Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile
| | - J Pablo Huidobro-Toro
- Pharmacology Laboratory, Department Biology, Faculty of Chemistry and Biology, Centro Desarrollo de Nanociencias y Nanotecnología (CEDENNA), Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile
| |
Collapse
|
3
|
Quinones C, Wilson JP, Kumbhare D, Guthikonda B, Hoang S. Clinical Assessment and Management of Acute Spinal Cord Injury. J Clin Med 2024; 13:5719. [PMID: 39407779 PMCID: PMC11477398 DOI: 10.3390/jcm13195719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/09/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
The information contained in this article is suitable for clinicians practicing in the United States desiring a general overview of the assessment and management of spinal cord injury (SCI), focusing on initial care, assessment, acute management, complications, prognostication, and future research directions. SCI presents significant challenges, affecting patients physically, emotionally, and financially, with variable recovery outcomes ranging from full functionality to lifelong dependence on caregivers. Initial care aims to minimize secondary injury through thorough neurological evaluations and imaging studies to assess the severity of the injury. Acute management prioritizes stabilizing respiratory and cardiovascular functions and maintaining proper spinal cord perfusion. Patients with unstable or progressive neurological decline benefit from timely surgical intervention to optimize neurological recovery. Subacute management focuses on addressing common complications affecting the respiratory, gastrointestinal, and genitourinary systems, emphasizing a holistic, multidisciplinary approach. Prognostication is currently based on neurological assessments and imaging findings, but emerging biomarkers offer the potential to refine outcome predictions further. Additionally, novel therapeutic interventions, such as hypothermia therapy and neuroprotective medications are being explored to mitigate secondary damage and enhance recovery. This paper serves as a high-yield refresher for clinicians for the assessment and management of acute spinal cord injury during index admission.
Collapse
Affiliation(s)
| | | | | | | | - Stanley Hoang
- Department of Neurosurgery, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA; (C.Q.); (J.P.W.J.); (D.K.); (B.G.)
| |
Collapse
|
4
|
Castillo-Vazquez SK, Massieu L, Rincón-Heredia R, García-de la Torre P, Quiroz-Baez R, Gomez-Verjan JC, Rivero-Segura NA. Glutamatergic Neurotransmission in Aging and Neurodegenerative Diseases: A Potential Target to Improve Cognitive Impairment in Aging. Arch Med Res 2024; 55:103039. [PMID: 38981341 DOI: 10.1016/j.arcmed.2024.103039] [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: 01/11/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
Aging is characterized by the decline in many of the individual's capabilities. It has been recognized that the brain undergoes structural and functional changes during aging that are occasionally associated with the development of neurodegenerative diseases. In this sense, altered glutamatergic neurotransmission, which involves the release, binding, reuptake, and degradation of glutamate (Glu) in the brain, has been widely studied in physiological and pathophysiological aging. In particular, changes in glutamatergic neurotransmission are exacerbated during neurodegenerative diseases and are associated with cognitive impairment, characterized by difficulties in memory, learning, concentration, and decision-making. Thus, in the present manuscript, we aim to highlight the relevance of glutamatergic neurotransmission during cognitive impairment to develop novel strategies to prevent, ameliorate, or delay cognitive decline. To achieve this goal, we provide a comprehensive review of the changes reported in glutamatergic neurotransmission components, such as Glu transporters and receptors during physiological aging and in the most studied neurodegenerative diseases. Finally, we describe the current therapeutic strategies developed to target glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Selma Karime Castillo-Vazquez
- Dirección de Investigación, Instituto Nacional de Geriatría, Mexico City, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lourdes Massieu
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ruth Rincón-Heredia
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Paola García-de la Torre
- 4 Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City Mexico
| | - Ricardo Quiroz-Baez
- Dirección de Investigación, Instituto Nacional de Geriatría, Mexico City, Mexico
| | | | | |
Collapse
|
5
|
Onder C, Onder C, Akesen S, Yumusak E, Akesen B. Riluzole is Effective on Spinal Decompression for Treating Acute Spinal Injury When Compared With Methylprednisolone and the Combination of Two Drugs: In Vivo Rat Model. Global Spine J 2024; 14:1899-1908. [PMID: 36812057 PMCID: PMC11418726 DOI: 10.1177/21925682231159068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
STUDY DESIGN Randomized controlled animal experiment. OBJECTIVES To determine and compare the efficacy of riluzole, MPS and the combination of two drugs in a rat model with acute spinal trauma, electrophysiologically and histopathologically. METHODS 59 rats were divided into 4 groups as control, riluzole (6 mg/kg, every 12 hours for 7 days), MPS (30 mg/kg, 2nd and 4th hours after injury) and riluzole + MPS. Spinal trauma was created and the subjects were followed for 7 days. Electrophysiological recordings were made via neuromonitoring. The subjects were sacrificed and histopathological examination was made. RESULTS For the amplitude values, mean alteration in the period from the spinal cord injury to the end of the 7th day is 15.89 ± 20.00%, 210.93 ± 199.44%, 24.75% ± 10.13% increase and 18.91 ± 30.01% decrease for the control, riluzole, riluzole + MPS and MPS groups, respectively. Although the riluzole treatment group produced the greatest increase in amplitude, it was observed that no treatment provided a significant improvement compared to the control group, in terms of latency and amplitude. It was observed that there was significantly less cavitation area in the riluzole treatment group compared to the control group (P = .020). (P < .05). CONCLUSIONS Electrophysiologically, no treatment was found to provide significant improvement. Histopathologically, it was observed that riluzole provided significant neural tissue protection.
Collapse
Affiliation(s)
- Cem Onder
- Faculty of Medicine, Department of Orthopaedics and Traumatology, Uludağ University, Bursa, Turkey
| | - Cigdem Onder
- Department of Physical Therapy and Rehabilitation, Sehitkamil Hospital, Gaziantep, Turkey
| | - Selcan Akesen
- Faculty of Medicine, Department of Anesthesiology and Reanimation, Uludağ University, Bursa, Turkey
| | - Ezgi Yumusak
- Faculty of Veterinary Medicine, Department of Pathology, Uludağ University, Bursa, Turkey
| | - Burak Akesen
- Faculty of Medicine, Department of Orthopaedics and Traumatology, Uludağ University, Bursa, Turkey
| |
Collapse
|
6
|
Levett JJ, Georgiopoulos M, Martel S, Mugheiry WA, Stavropoulos NA, Vega-Arroyo M, Santaguida C, Weber MH, Golan JD, Jarzem P, Ouellet JA, Klironomos G, Demetriades AK. Pharmacological Treatment of Degenerative Cervical Myelopathy: A Critical Review of Current Evidence. Neurospine 2024; 21:375-400. [PMID: 38955515 PMCID: PMC11224758 DOI: 10.14245/ns.2448140.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 07/04/2024] Open
Abstract
Degenerative cervical myelopathy (DCM) is the leading cause of spinal cord dysfunction in adults, representing substantial morbidity and significant financial and resource burdens. Typically, patients with progressive DCM will eventually receive surgical treatment. Nonetheless, despite advancements in pharmacotherapeutics, evidence for pharmacological therapy remains limited. Health professionals from various fields would find interest in pharmacological agents that could benefit patients with mild DCM or enhance surgical outcomes. This review aims to consolidate all clinical and experimental evidence on the pharmacological treatment of DCM. We conducted a comprehensive narrative review that presents all pharmacological agents that have been investigated for DCM treatment in both humans and animal models. Riluzole exhibits effectiveness solely in rat models, but not in treating mild DCM in humans. Cerebrolysin emerges as a potential neuroprotective agent for myelopathy in animals but had contradictory results in clinical trials. Limaprost alfadex demonstrates motor function improvement in animal models and exhibits promising outcomes in a small clinical trial. Glucocorticoids not only fail to provide clinical benefits but may also lead to adverse events. Cilostazol, anti-Fas ligand antibody, and Jingshu Keli display promise in animal studies, while erythropoietin, granulocyte colony-stimulating factor and limaprost alfadex exhibit potential in both animal and human research. Existing evidence mainly rests on weak clinical data and animal experimentation. Current pharmacological efforts target ion channels, stem cell differentiation, inflammatory, vascular, and apoptotic pathways. The inherent nature and pathogenesis of DCM offer substantial prospects for developing neurodegenerative or neuroprotective therapies capable of altering disease progression, potentially delaying surgical intervention, and optimizing outcomes for those undergoing surgical decompression.
Collapse
Affiliation(s)
- Jordan J Levett
- Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Miltiadis Georgiopoulos
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Spinal Surgery Unit, Swansea Bay University Health Board, Swansea, UK
| | - Simon Martel
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Wissam Al Mugheiry
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Nikolaos A. Stavropoulos
- First Department of Orthopaedic Surgery NKUA, “ATTIKON” University General Hospital, Athens, Greece
| | - Miguel Vega-Arroyo
- Winnipeg Spine Program, University of Manitoba, Winnipeg, MB, Canada
- Neurosurgery Department, Sanford Brain & Spine Center, Fargo, ND, USA
| | - Carlo Santaguida
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Michael H. Weber
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Jeff D. Golan
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Peter Jarzem
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Jean A. Ouellet
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Georgios Klironomos
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Bay Shore, NY, USA
| | - Andreas K. Demetriades
- Edinburgh Spinal Surgery Outcomes Study Group, Department of Neurosurgery, Royal Infirmary, Edinburgh, UK
| |
Collapse
|
7
|
Tsekrekou M, Giannakou M, Papanikolopoulou K, Skretas G. Protein aggregation and therapeutic strategies in SOD1- and TDP-43- linked ALS. Front Mol Biosci 2024; 11:1383453. [PMID: 38855322 PMCID: PMC11157337 DOI: 10.3389/fmolb.2024.1383453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 06/11/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with severe socio-economic impact. A hallmark of ALS pathology is the presence of aberrant cytoplasmic inclusions composed of misfolded and aggregated proteins, including both wild-type and mutant forms. This review highlights the critical role of misfolded protein species in ALS pathogenesis, particularly focusing on Cu/Zn superoxide dismutase (SOD1) and TAR DNA-binding protein 43 (TDP-43), and emphasizes the urgent need for innovative therapeutic strategies targeting these misfolded proteins directly. Despite significant advancements in understanding ALS mechanisms, the disease remains incurable, with current treatments offering limited clinical benefits. Through a comprehensive analysis, the review focuses on the direct modulation of the misfolded proteins and presents recent discoveries in small molecules and peptides that inhibit SOD1 and TDP-43 aggregation, underscoring their potential as effective treatments to modify disease progression and improve clinical outcomes.
Collapse
Affiliation(s)
- Maria Tsekrekou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Maria Giannakou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Katerina Papanikolopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, Vari, Greece
- ResQ Biotech, Patras Science Park, Rio, Greece
| | - Georgios Skretas
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- ResQ Biotech, Patras Science Park, Rio, Greece
- Institute for Bio-innovation, Biomedical Sciences Research Centre “Alexander Fleming”, Vari, Greece
| |
Collapse
|
8
|
Genge A, Wainwright S, Vande Velde C. Amyotrophic lateral sclerosis: exploring pathophysiology in the context of treatment. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:225-236. [PMID: 38001557 DOI: 10.1080/21678421.2023.2278503] [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: 08/02/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex, neurodegenerative disorder in which alterations in structural, physiological, and metabolic parameters act synergistically. Over the last decade there has been a considerable focus on developing drugs to slow the progression of the disease. Despite this, only four disease-modifying therapies are approved in North America. Although additional research is required for a thorough understanding of ALS, we have accumulated a large amount of knowledge that could be better integrated into future clinical trials to accelerate drug development and provide patients with improved treatment options. It is likely that future, successful ALS treatments will take a multi-pronged therapeutic approach, targeting different pathways, akin to personalized medicine in oncology. In this review, we discuss the link between ALS pathophysiology and treatments, looking at the therapeutic failures as learning opportunities that can help us refine and optimize drug development.
Collapse
Affiliation(s)
- Angela Genge
- Clinical Research Unit Director, ALS Clinic, Montreal, Quebec, Canada
| | - Steven Wainwright
- Amylyx Pharmaceuticals, Inc, Vancouver, British Columbia, Canada, and
| | - Christine Vande Velde
- CHUM Research Center, Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Dewa KI, Arimura N, Kakegawa W, Itoh M, Adachi T, Miyashita S, Inoue YU, Hizawa K, Hori K, Honjoya N, Yagishita H, Taya S, Miyazaki T, Usui C, Tatsumoto S, Tsuzuki A, Uetake H, Sakai K, Yamakawa K, Sasaki T, Nagai J, Kawaguchi Y, Sone M, Inoue T, Go Y, Ichinohe N, Kaibuchi K, Watanabe M, Koizumi S, Yuzaki M, Hoshino M. Neuronal DSCAM regulates the peri-synaptic localization of GLAST in Bergmann glia for functional synapse formation. Nat Commun 2024; 15:458. [PMID: 38302444 PMCID: PMC10834496 DOI: 10.1038/s41467-023-44579-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 12/19/2023] [Indexed: 02/03/2024] Open
Abstract
In the central nervous system, astrocytes enable appropriate synapse function through glutamate clearance from the synaptic cleft; however, it remains unclear how astrocytic glutamate transporters function at peri-synaptic contact. Here, we report that Down syndrome cell adhesion molecule (DSCAM) in Purkinje cells controls synapse formation and function in the developing cerebellum. Dscam-mutant mice show defects in CF synapse translocation as is observed in loss of function mutations in the astrocytic glutamate transporter GLAST expressed in Bergmann glia. These mice show impaired glutamate clearance and the delocalization of GLAST away from the cleft of parallel fibre (PF) synapse. GLAST complexes with the extracellular domain of DSCAM. Riluzole, as an activator of GLAST-mediated uptake, rescues the proximal impairment in CF synapse formation in Purkinje cell-selective Dscam-deficient mice. DSCAM is required for motor learning, but not gross motor coordination. In conclusion, the intercellular association of synaptic and astrocyte proteins is important for synapse formation and function in neural transmission.
Collapse
Affiliation(s)
- Ken-Ichi Dewa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, 409-3898, Japan
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Nariko Arimura
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan.
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Tohoku, Japan.
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Masayuki Itoh
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Toma Adachi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Satoshi Miyashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Yukiko U Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Kento Hizawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Tohoku, Japan
| | - Kei Hori
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Natsumi Honjoya
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Tohoku, Japan
| | - Haruya Yagishita
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Tohoku, Japan
| | - Shinichiro Taya
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
- Division of Behavioural Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Taisuke Miyazaki
- Department of Health Sciences, School of Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan
| | - Chika Usui
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan
| | - Shoji Tatsumoto
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan
| | - Akiko Tsuzuki
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Hirotomo Uetake
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Saitama, 274-8510, Japan
| | - Kazuhisa Sakai
- Department of Ultrastructural Research, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Kazuhiro Yamakawa
- Department of Neurodevelopmental Disorder Genetics, Nagoya City University Graduate School of Medicine, Nagoya, Aichi, 467-8601, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Tohoku, Japan
| | - Jun Nagai
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Yoshiya Kawaguchi
- Department of Life Science Frontiers, Center for iPS cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Masaki Sone
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Saitama, 274-8510, Japan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Yasuhiro Go
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan
- Department of System Neuroscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan
- Graduate School of Information Science, University of Hyogo, Kobe, Hyogo, 650-0047, Japan
| | - Noritaka Ichinohe
- Department of Ultrastructural Research, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, 060-8638, Japan
- The University of Texas at Austin, Austin, Texas, 78712-0805, USA
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, 409-3898, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan.
| |
Collapse
|
10
|
Lu HL, Jin JH, Liang SC, Feng CW, Li ZM, Zhao FG, Liu X, Shen YM. Photocatalytic Three-Component Reaction for the Synthesis of Multifunctional Diaryl Sulfides. J Org Chem 2023; 88:16547-16555. [PMID: 37971809 DOI: 10.1021/acs.joc.3c02048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
A photocatalytic three-component reaction of a nitroarene, a thiophenol, and a ketone for the synthesis of multifunctional diaryl sulfides was reported using a nitro group as the nitrogen source and thiophenol as the sulfur source. Thiophenol also serves as a proton donor to reduce nitroarene to arylamine as a key intermediate for the formation of C-N and C-S bonds. Good functional group tolerance and mild reaction conditions make this method have practical synthetic value for diversified multifunctional diaryl sulfides.
Collapse
Affiliation(s)
- Hui-Ling Lu
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Jia-Hui Jin
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Shang-Chuang Liang
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Chuan-Wei Feng
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Zhi-Ming Li
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Fu-Gang Zhao
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
- Zhejiang Sci-Tech University Shengzhou Innovation Research Institute, Shengzhou 312400, PR China
| | - Xunshan Liu
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
- Zhejiang Sci-Tech University Shengzhou Innovation Research Institute, Shengzhou 312400, PR China
| | - Yong-Miao Shen
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
- Zhejiang Sci-Tech University Shengzhou Innovation Research Institute, Shengzhou 312400, PR China
| |
Collapse
|
11
|
Citraro R, Bosco F, Di Gennaro G, Tallarico M, Guarnieri L, Gallelli L, Rania V, Siniscalchi A, De Sarro G, Leo A. An In Vivo Electroencephalographic Analysis of the Effect of Riluzole against Limbic and Absence Seizure and Comparison with Glutamate Antagonists. Pharmaceutics 2023; 15:2006. [PMID: 37514193 PMCID: PMC10386681 DOI: 10.3390/pharmaceutics15072006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/20/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Riluzole (RLZ) has demonstrated neuroprotective effects in several neurological disorders. These neuroprotective effects seem to be mainly due to its ability to inhibit the excitatory glutamatergic neurotransmission, acting on different targets located both at the presynaptic and postsynaptic levels. METHODS In the present study, we evaluated the effects of Riluzole (RLZ) against limbic seizures, induced by AMPA, kainate, and NMDA receptor agonists in Sprague-Dawley rats, and in a well-validated genetic model of absence epilepsy, the WAG/Rij rat. Furthermore, in this latter model, we also studied the effect of RLZ in co-administration with the competitive NMDA receptor antagonist, CPP, or the non-competitive AMPA receptor antagonist, THIQ-10c, on spike-wave discharges (SWDs) in WAG/Rij rats, to understand the potential involvement of AMPA and NMDA receptors in the anti-absence effect of RLZ. RESULTS In Sprague-Dawley rats, RLZ pretreatment significantly reduced the limbic seizure severity induced by glutamatergic agonists, suggesting an antagonism of RLZ mainly on NMDA rather than non-NMDA receptors. RLZ also reduced SWD parameters in WAG/Rij rats. Interestingly, the co-administration of RLZ with CPP did not increase the anti-absence activity of RLZ in this model, advocating a competitive effect on the NMDA receptor. In contrast, the co-administration of RLZ with THIQ-10c induced an additive effect against absence seizure in WAG/Rij rats. CONCLUSIONS these results suggest that the antiepileptic effects of RLZ, in both seizure models, can be mainly due to the antagonism of the NMDA glutamatergic receptors.
Collapse
Affiliation(s)
- Rita Citraro
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
- Research Center FAS@UMG, Department of Health Science, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Bosco
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Gianfranco Di Gennaro
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Martina Tallarico
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Lorenza Guarnieri
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Luca Gallelli
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
- Research Center FAS@UMG, Department of Health Science, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Rania
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Antonio Siniscalchi
- Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, 87100 Cosenza, Italy
| | - Giovambattista De Sarro
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
- Research Center FAS@UMG, Department of Health Science, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Antonio Leo
- Section of Pharmacology, Science of Health Department, School of Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
- Research Center FAS@UMG, Department of Health Science, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
12
|
Pardo-Moreno T, Mohamed-Mohamed H, Suleiman-Martos S, Ramos-Rodriguez JJ, Rivas-Dominguez A, Melguizo-Rodríguez L, Gómez-Urquiza JL, Bermudez-Pulgarin B, Garcia-Morales V. Amyotrophic Lateral Sclerosis and Serum Lipid Level Association: A Systematic Review and Meta-Analytic Study. Int J Mol Sci 2023; 24:ijms24108675. [PMID: 37240018 DOI: 10.3390/ijms24108675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/16/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with unknown etiology. Many metabolic alterations occur during ALS progress and can be used as a method of pre-diagnostic and early diagnosis. Dyslipidemia is one of the physiological changes observed in numerous ALS patients. The aim of this study is to analyze the possible relationship between the rate of disease progression (functional rating scale (ALS-FRS)) and the plasma lipid levels at the early stage of ALS. A systematic review was carried out in July 2022. The search equation was "Triglycerides AND amyotrophic lateral sclerosis" and its variants. Four meta-analyses were performed. Four studies were included in the meta-analysis. No significant differences were observed between the lipid levels (total cholesterol, triglycerides, HDL cholesterol, and LDL cholesterol) and the ALS-FRS score at the onset of the disease. Although the number of studies included in this research was low, the results of this meta-analytic study suggest that there is no clear relationship between the symptoms observed in ALS patients and the plasma lipid levels. An increase in research, as well as an expansion of the geographical area, would be of interest.
Collapse
Affiliation(s)
- Teresa Pardo-Moreno
- Department of Physiology, Faculty of Health Sciences-Ceuta, University of Granada, 51001 Ceuta, Spain
| | - Himan Mohamed-Mohamed
- Department of Physiology, Faculty of Health Sciences-Ceuta, University of Granada, 51001 Ceuta, Spain
| | | | - Juan José Ramos-Rodriguez
- Department of Physiology, Faculty of Health Sciences-Ceuta, University of Granada, 51001 Ceuta, Spain
| | | | - Lucía Melguizo-Rodríguez
- Department of Nursery, Faculty of Health Sciences-Ceuta, University of Granada, 51001 Ceuta, Spain
| | - José L Gómez-Urquiza
- Department of Nursery, Faculty of Health Sciences-Ceuta, University of Granada, 51001 Ceuta, Spain
| | | | - Victoria Garcia-Morales
- Physiology Area, Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cádiz, 11003 Cádiz, Spain
| |
Collapse
|
13
|
Hsu SK, Lu CW, Chiu KM, Lee MY, Lin TY, Wang SJ. Mangiferin depresses vesicular glutamate release in synaptosomes from the rat cerebral cortex by decreasing synapsin I phosphorylation. Eur J Pharmacol 2023; 950:175772. [PMID: 37146708 DOI: 10.1016/j.ejphar.2023.175772] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
Mangiferin is a glucosyl xanthone that has been shown to be a neuroprotective agent against brain disorders involving excess glutamate. However, the effect of mangiferin on the function of the glutamatergic system has not been investigated. In this study, we used synaptosomes from the rat cerebral cortex to investigate the effect of mangiferin on glutamate release and identify the possible underlying mechanism. We observed that mangiferin produced a concentration-dependent reduction in the release of glutamate elicited by 4-aminopyridine with an IC50 value of 25 μM. Inhibition of glutamate release was blocked by removing extracellular calcium and by treatment with the vacuolar-type H+-ATPase inhibitor bafilomycin A1, which prevents the uptake and storage of glutamate in vesicles. Moreover, we showed that mangiferin decreased the 4-aminopyridine-elicited FM1-43 release and synaptotagmin 1 luminal domain antibody (syt1-L ab) uptake from synaptosomes, which correlated with decreased synaptic vesicle exocytosis. Transmission electron microscopy in synaptosomes also showed that mangiferin attenuated the 4-aminopyridine-elicited decrease in the number of synaptic vesicles. In addition, antagonism of Ca2+/calmodulin-dependent kinase II (CaMKII) and protein kinase A (PKA) counteracted mangiferin's effect on glutamate release. Mangiferin also decreased the phosphorylation of CaMKII, PKA, and synapsin I elicited by 4-aminopyridine treatment. Our data suggest that mangiferin reduces PKA and CaMKII activation and synapsin I phosphorylation, which could decrease synaptic vesicle availability and lead to a subsequent reduction in vesicular glutamate release from synaptosomes.
Collapse
Affiliation(s)
- Szu-Kai Hsu
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Department of Neurosurgery, Cathay General Hospital, Taipei, 106438, Taiwan
| | - Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Electrical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Ming-Yi Lee
- Department of Medical Research, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan.
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, 33303, Taiwan.
| |
Collapse
|
14
|
Mead RJ, Shan N, Reiser HJ, Marshall F, Shaw PJ. Amyotrophic lateral sclerosis: a neurodegenerative disorder poised for successful therapeutic translation. Nat Rev Drug Discov 2023; 22:185-212. [PMID: 36543887 PMCID: PMC9768794 DOI: 10.1038/s41573-022-00612-2] [Citation(s) in RCA: 131] [Impact Index Per Article: 131.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2022] [Indexed: 12/24/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating disease caused by degeneration of motor neurons. As with all major neurodegenerative disorders, development of disease-modifying therapies has proven challenging for multiple reasons. Nevertheless, ALS is one of the few neurodegenerative diseases for which disease-modifying therapies are approved. Significant discoveries and advances have been made in ALS preclinical models, genetics, pathology, biomarkers, imaging and clinical readouts over the last 10-15 years. At the same time, novel therapeutic paradigms are being applied in areas of high unmet medical need, including neurodegenerative disorders. These developments have evolved our knowledge base, allowing identification of targeted candidate therapies for ALS with diverse mechanisms of action. In this Review, we discuss how this advanced knowledge, aligned with new approaches, can enable effective translation of therapeutic agents from preclinical studies through to clinical benefit for patients with ALS. We anticipate that this approach in ALS will also positively impact the field of drug discovery for neurodegenerative disorders more broadly.
Collapse
Affiliation(s)
- Richard J Mead
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Keapstone Therapeutics, The Innovation Centre, Broomhall, Sheffield, UK
| | - Ning Shan
- Aclipse Therapeutics, Radnor, PA, US
| | | | - Fiona Marshall
- MSD UK Discovery Centre, Merck, Sharp and Dohme (UK) Limited, London, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, UK.
- Keapstone Therapeutics, The Innovation Centre, Broomhall, Sheffield, UK.
| |
Collapse
|
15
|
Riluzole and novel naphthalenyl substituted aminothiazole derivatives prevent acute neural excitotoxic injury in a rat model of temporal lobe epilepsy. Neuropharmacology 2023; 224:109349. [PMID: 36436594 PMCID: PMC9843824 DOI: 10.1016/j.neuropharm.2022.109349] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022]
Abstract
Epileptogenic seizures, or status epilepticus (SE), leads to excitotoxic injury in hippocampal and limbic neurons in the kainic acid (KA) animal model of temporal lobe epilepsy (TLE). Here, we have further characterized neural activity regulated methylaminoisobutryic acid (MeAIB)/glutamine transport activity in mature rat hippocampal neurons in vitro that is inhibited by riluzole (IC50 = 1 μM), an anti-convulsant benzothiazole agent. We screened a library of riluzole derivatives and identified SKA-41 followed by a second screen and synthesized several novel chlorinated aminothiazoles (SKA-377, SKA-378, SKA-379) that are also potent MeAIB transport inhibitors in vitro, and brain penetrant following systemic administration. When administered before KA, SKA-378 did not prevent seizures but still protected the hippocampus and several other limbic areas against SE-induced neurodegeneration at 3d. When SKA-377 - 379, (30 mg/kg) were administered after KA-induced SE, acute neural injury in the CA3, CA1 and CA4/hilus was also largely attenuated. Riluzole (10 mg/kg) blocks acute neural injury. Kinetic analysis of SKA-378 and riluzoles' blockade of Ca2+-regulated MeAIB transport in neurons in vitro indicates that inhibition occurs via a non-competitive, indirect mechanism. Sodium channel NaV1.6 antagonism blocks neural activity regulated MeAIB/Gln transport in vitro (IC50 = 60 nM) and SKA-378 is the most potent inhibitor of NaV1.6 (IC50 = 28 μM) compared to NaV1.2 (IC50 = 118 μM) in heterologous cells. However, pharmacokinetic analysis suggests that sodium channel blockade may not be the predominant mechanism of neuroprotection here. Riluzole and our novel aminothiazoles are agents that attenuate acute neural hippocampal injury following KA-induced SE and may help to understand mechanisms involved in the progression of epileptic disease.
Collapse
|
16
|
Alijanpour S, Miryounesi M, Ghafouri-Fard S. The role of excitatory amino acid transporter 2 (EAAT2) in epilepsy and other neurological disorders. Metab Brain Dis 2023; 38:1-16. [PMID: 36173507 DOI: 10.1007/s11011-022-01091-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/15/2022] [Indexed: 02/03/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS). Excitatory amino acid transporters (EAATs) have important roles in the uptake of glutamate and termination of glutamatergic transmission. Up to now, five EAAT isoforms (EAAT1-5) have been identified in mammals. The main focus of this review is EAAT2. This protein has an important role in the pathoetiology of epilepsy. De novo dominant mutations, as well as inherited recessive mutation in this gene, have been associated with epilepsy. Moreover, dysregulation of this protein is implicated in a range of neurological diseases, namely amyotrophic lateral sclerosis, alzheimer's disease, parkinson's disease, schizophrenia, epilepsy, and autism. In this review, we summarize the role of EAAT2 in epilepsy and other neurological disorders, then provide an overview of the therapeutic modulation of this protein.
Collapse
Affiliation(s)
- Sahar Alijanpour
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Miryounesi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| |
Collapse
|
18
|
Saba K, Patel AB. Riluzole restores memory and brain energy metabolism in AβPP-PS1 mouse model of Alzheimer's disease. Biochem Biophys Res Commun 2022; 610:140-146. [DOI: 10.1016/j.bbrc.2022.04.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 11/02/2022]
|
19
|
Inhibition of Glutamate Release from Rat Cortical Nerve Terminals by Dehydrocorydaline, an Alkaloid from Corydalis yanhusuo. Molecules 2022; 27:molecules27030960. [PMID: 35164225 PMCID: PMC8838318 DOI: 10.3390/molecules27030960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 02/01/2023] Open
Abstract
Excessive release of glutamate induces excitotoxicity and causes neuronal damage in several neurodegenerative diseases. Natural products have emerged as potential neuroprotective agents for preventing and treating neurological disorders. Dehydrocorydaline (DHC), an active alkaloid compound isolated from Corydalis yanhusuo, possesses neuroprotective capacity. The present study investigated the effect of DHC on glutamate release using a rat brain cortical synaptosome model. Our results indicate that DHC inhibited 4-aminopyridine (4-AP)-evoked glutamate release and elevated intrasynaptosomal calcium levels. The inhibitory effect of DHC on 4-AP-evoked glutamate release was prevented in the presence of the vesicular transporter inhibitor bafilomycin A1 and the N- and P/Q-type Ca2+ channel blocker ω-conotoxin MVIIC but not the intracellular inhibitor of Ca2+ release dantrolene or the mitochondrial Na+/Ca2+ exchanger inhibitor CGP37157. Moreover, the inhibitory effect of DHC on evoked glutamate release was prevented by the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) inhibitor PD98059. Western blotting data in synaptosomes also showed that DHC significantly decreased the level of ERK1/2 phosphorylation and synaptic vesicle-associated protein synapsin I, the main presynaptic target of ERK. Together, these results suggest that DHC inhibits presynaptic glutamate release from cerebrocortical synaptosomes by suppressing presynaptic voltage-dependent Ca2+ entry and the MAPK/ERK/synapsin I signaling pathway.
Collapse
|
20
|
Taiji R, Yamanaka M, Taniguchi W, Nishio N, Tsutsui S, Nakatsuka T, Yamada H. Anti-allodynic and promotive effect on inhibitory synaptic transmission of riluzole in rat spinal dorsal horn. Biochem Biophys Rep 2021; 28:101130. [PMID: 34541342 PMCID: PMC8435917 DOI: 10.1016/j.bbrep.2021.101130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/21/2021] [Accepted: 09/03/2021] [Indexed: 10/29/2022] Open
Abstract
Riluzole (2-amino-6-(trifluoromethoxy)benzothiazole) is a drug known for its inhibitory effect on glutamatergic transmission and its anti-nociceptive and anti-allodynic effects in neuropathic pain rat models. Riluzole also has an enhancing effect on GABAergic synaptic transmission. However, the effect on the spinal dorsal horn, which plays an important role in modulating nociceptive transmission, remains unknown. We investigated the ameliorating effect of riluzole on mechanical allodynia using the von Frey test in a rat model of neuropathic pain and analyzed the synaptic action of riluzole on inhibitory synaptic transmission in substantia gelatinosa (SG) neurons using whole-cell patch clamp recordings. We found that single-dose intraperitoneal riluzole (4 mg/kg) administration effectively attenuated mechanical allodynia in the short term in a rat model of neuropathic pain. Moreover, 300 μM riluzole induced an outward current in rat SG neurons. The outward current induced by riluzole was not suppressed in the presence of tetrodotoxin. Furthermore, we found that the outward current was suppressed by simultaneous bicuculline and strychnine application, but not by strychnine alone. Altogether, these results suggest that riluzole enhances inhibitory synaptic transmission monosynaptically by potentiating GABAergic synaptic transmission in the rat spinal dorsal horn.
Collapse
Affiliation(s)
- Ryo Taiji
- Department of Orthopaedic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Manabu Yamanaka
- Department of Orthopaedic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Wataru Taniguchi
- Department of Orthopaedic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Naoko Nishio
- Department of Orthopaedic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Shunji Tsutsui
- Department of Orthopaedic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Terumasa Nakatsuka
- Pain Research Center, Kansai University of Health Sciences, 2-11-1 Wakaba, Kumatori, Osaka, 590-0482, Japan
| | - Hiroshi Yamada
- Department of Orthopaedic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| |
Collapse
|
21
|
Jichkar AA, Opai IA, Karade NN. N-Iodosuccinimide mediated intramolecular oxidative C(sp2)-S bond formation for the synthesis of 2-aminobenzothiazole derivatives. J Sulphur Chem 2021. [DOI: 10.1080/17415993.2021.1989436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Atul A. Jichkar
- Department of Chemistry, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, India
| | - Imran A. Opai
- Department of Chemistry, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, India
| | - Nandkishor N. Karade
- Department of Chemistry, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, India
| |
Collapse
|
22
|
Green JL, Dos Santos WF, Fontana ACK. Role of glutamate excitotoxicity and glutamate transporter EAAT2 in epilepsy: Opportunities for novel therapeutics development. Biochem Pharmacol 2021; 193:114786. [PMID: 34571003 DOI: 10.1016/j.bcp.2021.114786] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022]
Abstract
Epilepsy is a complex neurological syndrome characterized by seizures resulting from neuronal hyperexcitability and sudden and synchronized bursts of electrical discharges. Impaired astrocyte function that results in glutamate excitotoxicity has been recognized to play a key role in the pathogenesis of epilepsy. While there are 26 drugs marketed as anti-epileptic drugs no current treatments are disease modifying as they only suppress seizures rather than the development and progression of epilepsy. Excitatory amino acid transporters (EAATs) are critical for maintaining low extracellular glutamate concentrations and preventing excitotoxicity. When extracellular glutamate concentrations rise to abnormal levels, glutamate receptor overactivation and the subsequent excessive influx of calcium into the post-synaptic neuron can trigger cell death pathways. In this review we discuss targeting EAAT2, the predominant glutamate transporter in the CNS, as a promising approach for developing therapies for epilepsy. EAAT2 upregulation via transcriptional and translational regulation has proven successful in vivo in reducing spontaneous recurrent seizures and offering neuroprotective effects. Another approach to regulate EAAT2 activity is through positive allosteric modulation (PAM). Novel PAMs of EAAT2 have recently been identified and are under development, representing a promising approach for the advance of novel therapeutics for epilepsy.
Collapse
Affiliation(s)
- Jennifer Leigh Green
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, United States
| | | | | |
Collapse
|
23
|
Tefera TW, Steyn FJ, Ngo ST, Borges K. CNS glucose metabolism in Amyotrophic Lateral Sclerosis: a therapeutic target? Cell Biosci 2021; 11:14. [PMID: 33431046 PMCID: PMC7798275 DOI: 10.1186/s13578-020-00511-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal progressive neurodegenerative disorder primarily characterized by selective degeneration of both the upper motor neurons in the brain and lower motor neurons in the brain stem and the spinal cord. The exact mechanism for the selective death of neurons is unknown. A growing body of evidence demonstrates abnormalities in energy metabolism at the cellular and whole-body level in animal models and in people living with ALS. Many patients with ALS exhibit metabolic changes such as hypermetabolism and body weight loss. Despite these whole-body metabolic changes being observed in patients with ALS, the origin of metabolic dysregulation remains to be fully elucidated. A number of pre-clinical studies indicate that underlying bioenergetic impairments at the cellular level may contribute to metabolic dysfunctions in ALS. In particular, defects in CNS glucose transport and metabolism appear to lead to reduced mitochondrial energy generation and increased oxidative stress, which seem to contribute to disease progression in ALS. Here, we review the current knowledge and understanding regarding dysfunctions in CNS glucose metabolism in ALS focusing on metabolic impairments in glucose transport, glycolysis, pentose phosphate pathway, TCA cycle and oxidative phosphorylation. We also summarize disturbances found in glycogen metabolism and neuroglial metabolic interactions. Finally, we discuss options for future investigations into how metabolic impairments can be modified to slow disease progression in ALS. These investigations are imperative for understanding the underlying causes of metabolic dysfunction and subsequent neurodegeneration, and to also reveal new therapeutic strategies in ALS.
Collapse
Affiliation(s)
- Tesfaye Wolde Tefera
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Frederik J Steyn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.,Center for Clinical Research, The University of Queensland, Brisbane, Australia
| | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.,Center for Clinical Research, The University of Queensland, Brisbane, Australia
| | - Karin Borges
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
24
|
Jørgensen HS, Jensen DB, Dimintiyanova KP, Bonnevie VS, Hedegaard A, Lehnhoff J, Moldovan M, Grondahl L, Meehan CF. Increased Axon Initial Segment Length Results in Increased Na + Currents in Spinal Motoneurones at Symptom Onset in the G127X SOD1 Mouse Model of Amyotrophic Lateral Sclerosis. Neuroscience 2020; 468:247-264. [PMID: 33246068 DOI: 10.1016/j.neuroscience.2020.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/22/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease preferentially affecting motoneurones. Transgenic mouse models have been used to investigate the role of abnormal motoneurone excitability in this disease. Whilst an increased excitability has repeatedly been demonstrated in vitro in neonatal and embryonic preparations from SOD1 mouse models, the results from the only studies to record in vivo from spinal motoneurones in adult SOD1 models have produced conflicting findings. Deficits in repetitive firing have been reported in G93A SOD1(high copy number) mice but not in presymptomatic G127X SOD1 mice despite shorter motoneurone axon initial segments (AISs) in these mice. These discrepancies may be due to the earlier disease onset and prolonged disease progression in G93A SOD1 mice with recordings potentially performed at a later sub-clinical stage of the disease in this mouse. To test this, and to explore how the evolution of excitability changes with symptom onset we performed in vivo intracellular recording and AIS labelling in G127X SOD1 mice immediately after symptom onset. No reductions in repetitive firing were observed showing that this is not a common feature across all ALS models. Immunohistochemistry for the Na+ channel Nav1.6 showed that motoneurone AISs increase in length in G127X SOD1 mice at symptom onset. Consistent with this, the rate of rise of AIS components of antidromic action potentials were significantly faster confirming that this increase in length represents an increase in AIS Na+ channels occurring at symptom onset in this model.
Collapse
Affiliation(s)
- H S Jørgensen
- Department of Neuroscience, University of Copenhagen, Denmark
| | - D B Jensen
- Department of Neuroscience, University of Copenhagen, Denmark
| | | | - V S Bonnevie
- Department of Neuroscience, University of Copenhagen, Denmark
| | - A Hedegaard
- Department of Neuroscience, University of Copenhagen, Denmark
| | - J Lehnhoff
- Department of Neuroscience, University of Copenhagen, Denmark
| | - M Moldovan
- Department of Neuroscience, University of Copenhagen, Denmark
| | - L Grondahl
- Department of Neuroscience, University of Copenhagen, Denmark
| | - C F Meehan
- Department of Neuroscience, University of Copenhagen, Denmark.
| |
Collapse
|
25
|
Hascup KN, Findley CA, Britz J, Esperant-Hilaire N, Broderick SO, Delfino K, Tischkau S, Bartke A, Hascup ER. Riluzole attenuates glutamatergic tone and cognitive decline in AβPP/PS1 mice. J Neurochem 2020; 156:513-523. [PMID: 33107040 DOI: 10.1111/jnc.15224] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/17/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022]
Abstract
We have previously demonstrated hippocampal hyperglutamatergic signaling occurs prior to plaque accumulation in AβPP/PS1 mice. Here, we evaluate 2-Amino-6-(trifluoromethoxy) benzothiazole (riluzole) as an early intervention strategy for Alzheimer's disease (AD), aimed at restoring glutamate neurotransmission prior to substantial Beta amyloid (Aβ) plaque accumulation and cognitive decline. Male AβPP/PS1 mice, a model of progressive cerebral amyloidosis, were treated with riluzole from 2-6 months of age. Morris water maze, in vivo electrochemistry, and immunofluorescence were performed to assess cognition, glutamatergic neurotransmission, and pathology, respectively, at 12 months. Four months of prodromal riluzole treatment in AβPP/PS1 mice resulted in long-lasting procognitive effects and attenuated glutamatergic tone that was observed six months after discontinuing riluzole treatment. Riluzole-treated AβPP/PS1 mice had significant improvement in long-term memory compared to vehicle-treated AβPP/PS1 mice that was similar to normal aging C57BL/6J control mice. Furthermore, basal glutamate concentration and evoked-glutamate release levels, which were elevated in vehicle-treated AβPP/PS1 mice, were restored to levels observed in age-matched C57BL/6J mice in AβPP/PS1 mice receiving prodromal riluzole treatment. Aβ plaque accumulation was not altered with riluzole treatment. This study supports that interventions targeting the glutamatergic system during the early stages of AD progression have long-term effects on disease outcome, and importantly may prevent cognitive decline. Our observations provide preclinical support for targeting glutamate neurotransmission in patients at risk for developing AD. Read the Editorial Highlight for this article on page 399.
Collapse
Affiliation(s)
- Kevin N Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Caleigh A Findley
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Jesse Britz
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Nahayo Esperant-Hilaire
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Sarah O Broderick
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kristin Delfino
- Department of Surgery, Center for Clinical Research, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Shelley Tischkau
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
26
|
Chen S, Liao Q, Lu K, Zhou J, Huang C, Bi F. Riluzole Exhibits No Therapeutic Efficacy on a Transgenic Rat model of Amyotrophic Lateral Sclerosis. Curr Neurovasc Res 2020; 17:275-285. [PMID: 32271694 DOI: 10.2174/1567202617666200409125227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurological disorder clinically characterized by motor system dysfunction, with intraneuronal accumulation of the TAR DNAbinding protein 43 (TDP-43) being a pathological hallmark. Riluzole is a primarily prescribed medicine for ALS patients, while its therapeutical efficacy appears limited. TDP-43 transgenic mice are existing animal models for mechanistic/translational research into ALS. METHODS We developed a transgenic rat model of ALS expressing a mutant human TDP-43 transgene (TDP-43M337V) and evaluated the therapeutic effect of Riluzole on this model. Relative to control, rats with TDP-43M337V expression promoted by the neurofilament heavy subunit (NEF) gene or specifically in motor neurons promoted by the choline acetyltransferase (ChAT) gene showed progressive worsening of mobility and grip strength, along with loss of motor neurons, microglial activation, and intraneuronal accumulation of TDP-43 and ubiquitin aggregations in the spinal cord. RESULTS Compared to vehicle control, intragastric administration of Riluzole (30 mg/kg/d) did not mitigate the behavioral deficits nor alter the neuropathologies in the transgenics. CONCLUSION These findings indicate that transgenic rats recapitulate the basic neurological and neuropathological characteristics of human ALS, while Riluzole treatment can not halt the development of the behavioral and histopathological phenotypes in this new transgenic rodent model of ALS.
Collapse
Affiliation(s)
- Si Chen
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Qiao Liao
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Ke Lu
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Jinxia Zhou
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Cao Huang
- Department of Pathology Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Fangfang Bi
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
27
|
Obrador E, Salvador R, López-Blanch R, Jihad-Jebbar A, Vallés SL, Estrela JM. Oxidative Stress, Neuroinflammation and Mitochondria in the Pathophysiology of Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2020; 9:E901. [PMID: 32971909 PMCID: PMC7555310 DOI: 10.3390/antiox9090901] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron (MN) disease. Its primary cause remains elusive, although a combination of different causal factors cannot be ruled out. There is no cure, and prognosis is poor. Most patients with ALS die due to disease-related complications, such as respiratory failure, within three years of diagnosis. While the underlying mechanisms are unclear, different cell types (microglia, astrocytes, macrophages and T cell subsets) appear to play key roles in the pathophysiology of the disease. Neuroinflammation and oxidative stress pave the way leading to neurodegeneration and MN death. ALS-associated mitochondrial dysfunction occurs at different levels, and these organelles are involved in the mechanism of MN death. Molecular and cellular interactions are presented here as a sequential cascade of events. Based on our present knowledge, the discussion leads to the idea that feasible therapeutic strategies should focus in interfering with the pathophysiology of the disease at different steps.
Collapse
Affiliation(s)
| | | | | | | | | | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 4601016 Valencia, Spain; (E.O.); (R.S.); (R.L.-B.); (A.J.-J.); (S.L.V.)
| |
Collapse
|
28
|
Qi Z, Yang X, Sang Y, Liu Y, Li J, Xu B, Liu W, He M, Xu Z, Deng Y, Zhu J. Fluoxetine and Riluzole Mitigates Manganese-Induced Disruption of Glutamate Transporters and Excitotoxicity via Ephrin-A3/GLAST-GLT-1/Glu Signaling Pathway in Striatum of Mice. Neurotox Res 2020; 38:508-523. [PMID: 32472497 DOI: 10.1007/s12640-020-00209-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 01/05/2023]
Abstract
Manganese (Mn) is an essential element required for many biological processes and systems in the human body. Mn intoxication increases brain glutamate (Glu) levels causing neuronal damage. Recent studies have reported that ephrin-A3 regulates this glutamate transporter. However, none has explored the role of this crucial molecule in Mn-induced excitotoxicity. The present study investigated whether ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity using astrocytes and Kunming mice. The mechanisms were explored using fluoxetine (ephrin-A3 inhibitor) and riluzole (a Glu release inhibitor). Firstly, we demonstrated that Mn exposure (500 μM or 50 mg/kg MnCl2) significantly increased Mn, ephrin-A3, and Glu levels, and inhibited Na+-K+ ATPase activity, as well as mRNA and protein levels of GLAST and GLT-1. Secondly, we found that astrocytes and mice pretreated with fluoxetine (100 μM or 15 mg/kg) and riluzole (100 μM or 32 μmol/kg) prior to Mn exposure had lower ephrin-A3 and Glu levels, but higher Na+-K+ ATPase activity, expression levels of GLAST and GLT-1 than those exposed to 500 μM or 50 mg/kg MnCl2. Moreover, the morphology of cells and the histomorphology of mice striatum were injured. Results showed that pretreatment with fluoxetine and riluzole attenuated the Mn-induced motor dysfunctions. Together, these results suggest that the ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity, and fluoxetine and riluzole can mitigate the Mn-induced excitotoxicity in mice brain.
Collapse
Affiliation(s)
- Zhipeng Qi
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Xinxin Yang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yanqi Sang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yanan Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Miao He
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| | - Jinghai Zhu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| |
Collapse
|
29
|
Effect of Riluzole, a Glutamate Release Inhibitor, on Synaptic Plasticity in the Intrahippocampal Aβ Rat Model of Alzheimer’s Disease. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09820-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
30
|
Updates on Preclinical and Translational Neuroscience of Mood Disorders: A Brief Historical Focus on Ketamine for the Clinician. J Clin Psychopharmacol 2019; 39:665-672. [PMID: 31688400 DOI: 10.1097/jcp.0000000000001132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The development of new-generation antidepressants comes at a time of great clinical need when the global burden of depression, suicide, and other psychiatric conditions continues to increase. Our current treatment armamentarium is limited by the time delay needed for antidepressant effects and the significant number of patients who do not show an adequate response to antidepressants. The past 2 decades of psychiatric research has revealed that ketamine, known to be used only as an anesthetic and drug of abuse and to produce experimental models of psychosis, is effective at subanesthetic doses to ameliorate clinical depression. METHODS We performed a systematic search of PubMed/MEDLINE indexed reports to identify clinical and translational research done with ketamine for purposes of treating depression. RESULTS We will first present the rationale for investigating ketamine and other N-methyl-D-aspartate receptor antagonists as a novel class of glutamate system targeting antidepressants. We will summarize putative molecular pathways underlying mood disorders and outline a brief history of investigation into ketamine as a treatment for depression. Recent clinical/translational evidence of ketamine's rapid-acting antidepressant mechanism will be critically reviewed in detail. CONCLUSIONS At the end of this review, we will opine on the role of ketamine and derivatives in clinical practice.
Collapse
|
31
|
Sakurai H, Dording C, Yeung A, Foster S, Jain F, Chang T, Trinh NH, Bernard R, Boyden S, Iqbal SZ, Wilkinson ST, Mathew SJ, Mischoulon D, Fava M, Cusin C. Longer-term open-label study of adjunctive riluzole in treatment-resistant depression. J Affect Disord 2019; 258:102-108. [PMID: 31400624 PMCID: PMC6710149 DOI: 10.1016/j.jad.2019.06.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/03/2019] [Accepted: 06/30/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND While riluzole has been investigated for the treatment of depression, little is known about its longer-term efficacy and optimal treatment duration in treatment-resistant depression (TRD). The objective of this study is to characterize the longer-term outcome of adjunctive riluzole therapy for TRD in an open-label extension of an 8-week acute treatment trial. METHODS The data from 66 patients with TRD who received adjunctive riluzole in a 12-week open-label extension phase were analyzed. Response rates (⩾50% reduction in the Mongomery-Asberg Depression Rating Scale [MADRS] score), relapse rates (a MADRS score of ⩾22 in patients who had previously achieved response), and adverse events were examined in patients who had achieved response at the end of the acute phase and those who had not. RESULTS Among acute phase responders, the maintained response rate was 66.7% (8/12) and the relapse rate was 8.3% (1/12). In acute phase non-responders, the response rate was 24.1% (13/54). The most commonly reported adverse event was fatigue (9.1%). Three cases were considered serious adverse events; vomiting (n = 1), shortness of breath (n = 1), and aborted suicide attempt (n = 1). LIMITATIONS This longer-term study was open-label and uncontrolled. The sample size was relatively small. CONCLUSIONS Longer-term adjunctive riluzole appears relatively well tolerated and beneficial for maintaining previous response. Additionally, approximately one fourth of patients who did not respond to 8-week antidepressant treatment might respond if treated with riluzole for 12 weeks. Those findings warrant further investigation because adjunctive riluzole could represent an option for treatment of depression when standard antidepressants have failed.
Collapse
Affiliation(s)
- Hitoshi Sakurai
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA,Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Christina Dording
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Albert Yeung
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Simmie Foster
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Felipe Jain
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Trina Chang
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Nhi-Ha Trinh
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Richard Bernard
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Sean Boyden
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Syed Z Iqbal
- Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA,Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Samuel T Wilkinson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Sanjay J Mathew
- Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA,Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - David Mischoulon
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Maurizio Fava
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Cristina Cusin
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, 1 Bowdoin Square, 6th Floor, Boston, MA, USA.
| |
Collapse
|
32
|
Marazziti D, Albert U, Mucci F, Piccinni A. The Glutamate and the Immune Systems: New Targets for the Pharmacological Treatment of OCD. Curr Med Chem 2019; 25:5731-5738. [PMID: 29119912 DOI: 10.2174/0929867324666171108152035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/06/2017] [Accepted: 12/25/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND In the last decades the pharmacological treatment of obsessivecompulsive disorder (OCD) has been significantly promoted by the effectiveness of selective serotonin (5-HT) reuptake inhibitors (SSRIs) and the subsequent development of the 5-HT hypothesis of OCD. However, since a large majority of patients (between 40% and 60 %) do not respond to SSRIs or strategies based on the modulation of the 5-HT system, it is now essential to search for other possible therapeutic targets. AIMS The aim of this paper was to review current literature through a PubMed and Google Scholar search of novel hypotheses and related compounds for the treatment of OCD, with a special focus on the glutammate and the immune systems. DISCUSSION The literature indicates that glutamate, the main excitatory neurotransmitter, might play an important role in the pathophysiology of OCD. In addition, a series of clinical studies also supports the potential efficacy of drugs modulating the glutamate system. The role of the immune system alterations in OCD in both children and adults needs to be more deeply elucidated. In children, a subtype of OCD has been widely described resulting from infections driven by group A streptococcus β-hemolitic and belonging to the so-called "pediatric autoimmune neuropsychiatric disorders associated with streptococcus" (PANDAS). In adults, available findings are meager and controversial, although interesting. CONCLUSION The glutamate and the immune systems represent two intriguing topics of research that hold promise for the development of open novel treatment strategies in OCD.
Collapse
Affiliation(s)
- Donatella Marazziti
- Dipartimento di Medicina Clinica e Sperimentale, Section of Psychiatry, University of Pisa, Pisa, Italy
| | - Umberto Albert
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Italy
| | - Federico Mucci
- Dipartimento di Medicina Clinica e Sperimentale, Section of Psychiatry, University of Pisa, Pisa, Italy
| | - Armando Piccinni
- Dipartimento di Medicina Clinica e Sperimentale, Section of Psychiatry, University of Pisa, Pisa, Italy
| |
Collapse
|
33
|
The Effect of Glutamatergic Modulators on Extracellular Glutamate: How Does this Information Contribute to the Discovery of Novel Antidepressants? Curr Ther Res Clin Exp 2019; 91:25-32. [PMID: 31871505 PMCID: PMC6911922 DOI: 10.1016/j.curtheres.2019.100566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/29/2019] [Indexed: 01/19/2023] Open
Abstract
The complexity of glutamatergic signaling challenges glutamate modulator usage. Functional biomarkers are needed to understand the MOA of glutamate modulators. Evaluating drug effect on EAATs' kinetics may add to antidepressant discovery.
Background In the search for new antidepressants, clinical researchers have been using drugs that simultaneously modulate multiple targets. During preclinical and clinical trials, the glutamatergic modulators riluzole and ketamine have received particular attention. Glutamatergic agents have a modulatory effect on synaptic transmission, so they can act on both neurons and astrocytes. In addition to influencing the quantity of glutamate released, these modulators can also affect the expression, localization, and functionality of glutamate-binding sites. Objective This review discusses the complexity of the glutamatergic system, the ambiguity of data regarding glutamate levels in patients with depression, as well as the mechanisms of action for riluzole and ketamine, which includes their relation to the physiology of glutamatergic transmission. The principal aim is to contribute to the development of novel glutamatergic antidepressant medications whilst emphasizing the need for innovative approaches that evaluate their effects on extracellular glutamate. Methods Literature was obtained via PubMed by searching the term depression in combination with each of the following terms: riluzole, ketamine, and glutamate. The search was restricted to full-text articles published in English between 1985 and 2018 relating to both the modulatory mechanisms of glutamatergic-binding proteins and the antidepressant actions of these medicines. Articles about mechanisms associated with synaptic plasticity and antidepressant effects were excluded. Results Although experimental data relates glutamatergic signaling to the pathophysiology of major depression and bipolar disorder, the role of glutamate—as well as its extracellular concentration in patients with said disorders—is still unclear. Riluzole's antidepressant action is ascribed to its capacity to reduce glutamate levels in the synaptic cleft, and ketamine's effect has been associated with increased extracellular glutamate levels. Conclusions The strategy of using glutamatergic modulators as therapeutic agents requires a better understanding of the role of glutamate in the pathophysiology of depression. Gaining such understanding is a challenge because it entails evaluating different targets as well as the effects of these modulators on the kinetics of glutamate uptake. Essentially, glutamate transport is a dynamic process and, currently, it is still necessary to develop new approaches to assay glutamate in the synaptic cleft. ORCID: 0000-0002-3358-6939.
Collapse
|
34
|
Faissner S, Plemel JR, Gold R, Yong VW. Progressive multiple sclerosis: from pathophysiology to therapeutic strategies. Nat Rev Drug Discov 2019; 18:905-922. [PMID: 31399729 DOI: 10.1038/s41573-019-0035-2] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system that involves demyelination and axonal degeneration. Although substantial progress has been made in drug development for relapsing-remitting MS, treatment of the progressive forms of the disease, which are characterized clinically by the accumulation of disability in the absence of relapses, remains unsatisfactory. This unmet clinical need is related to the complexity of the pathophysiological mechanisms involved in MS progression. Chronic inflammation, which occurs behind a closed blood-brain barrier with activation of microglia and continued involvement of T cells and B cells, is a hallmark pathophysiological feature. Inflammation can enhance mitochondrial damage in neurons, which, consequently, develop an energy deficit, further reducing axonal health. The growth-inhibitory and inflammatory environment of lesions also impairs remyelination, a repair process that might protect axons from degeneration. Moreover, neurodegeneration is accelerated by the altered expression of ion channels on denuded axons. In this Review, we discuss the current understanding of these disease mechanisms and highlight emerging therapeutic strategies based on these insights, including those targeting the neuroinflammatory and degenerative aspects as well as remyelination-promoting approaches.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany. .,Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| | - Jason R Plemel
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
35
|
Pagliaroli L, Widomska J, Nespoli E, Hildebrandt T, Barta C, Glennon J, Hengerer B, Poelmans G. Riluzole Attenuates L-DOPA-Induced Abnormal Involuntary Movements Through Decreasing CREB1 Activity: Insights from a Rat Model. Mol Neurobiol 2019; 56:5111-5121. [PMID: 30484112 PMCID: PMC6647536 DOI: 10.1007/s12035-018-1433-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/15/2018] [Indexed: 12/17/2022]
Abstract
Chronic administration of L-DOPA, the first-line treatment of dystonic symptoms in childhood or in Parkinson's disease, often leads to the development of abnormal involuntary movements (AIMs), which represent an important clinical problem. Although it is known that Riluzole attenuates L-DOPA-induced AIMs, the molecular mechanisms underlying this effect are not understood. Therefore, we studied the behavior and performed RNA sequencing of the striatum in three groups of rats that all received a unilateral lesion with 6-hydroxydopamine in their medial forebrain bundle, followed by the administration of saline, L-DOPA, or L-DOPA combined with Riluzole. First, we provide evidence that Riluzole attenuates AIMs in this rat model. Subsequently, analysis of the transcriptomics data revealed that Riluzole is predicted to reduce the activity of CREB1, a transcription factor that regulates the expression of multiple proteins that interact in a molecular landscape involved in apoptosis. Although this mechanism underlying the beneficial effect of Riluzole on AIMs needs to be confirmed, it provides clues towards novel or existing compounds for the treatment of AIMs that modulate the activity of CREB1 and, hence, its downstream targets.
Collapse
Affiliation(s)
- Luca Pagliaroli
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Joanna Widomska
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ester Nespoli
- CNS Department, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- Department of Child and Adolescent Psychiatry/Psychotherapy, University of Ulm, Ulm, Germany
| | - Tobias Hildebrandt
- Target Discovery, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Csaba Barta
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Jeffrey Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bastian Hengerer
- CNS Department, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
36
|
Zhou X, Li G, Yang B, Wu J. Quercetin Enhances Inhibitory Synaptic Inputs and Reduces Excitatory Synaptic Inputs to OFF- and ON-Type Retinal Ganglion Cells in a Chronic Glaucoma Rat Model. Front Neurosci 2019; 13:672. [PMID: 31293381 PMCID: PMC6604910 DOI: 10.3389/fnins.2019.00672] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/12/2019] [Indexed: 01/28/2023] Open
Abstract
Background Glaucoma is a neurodegenerative disease caused by excitotoxic injury of retinal ganglion cells (RGCs). In our previous model of high intraocular pressure, prepared by injecting magnetic beads into the anterior chamber, we demonstrated that an important natural dietary flavonoid compound (quercetin) can improve RGC function. However, it is unclear whether quercetin can improve the synaptic function of RGCs and how quercetin regulates synaptic transmission in rat models of chronic glaucoma. Methods A rat model of chronic glaucoma was prepared by electrocoagulation of the superior scleral vein. Electrophysiological electroretinography was used to detect the photopic negative response (PhNR). The whole-cell patch-clamp technique was used to clamp ON- and OFF- type RGCs in sections from normal retinas and from retinas that had been subjected to glaucoma for 4 weeks. Results Quercetin can reverse the decrease in PhNR amplitude caused by chronic glaucoma. The baseline frequency of miniature GABAergic inhibitory postsynaptic currents (mIPSCs) in the RGCs of glaucomatous retinal slices was lower than that of the control group. The frequencies of miniature excitatory postsynaptic currents (mEPSCs) were not significantly different between control and glaucomatous RGCs. The baseline frequencies of GABAergic mIPSCs and mEPSCs in OFF-type glaucomatous RGCs were greater than those in ON-type glaucomatous RGCs. Quercetin increased miniature GABAergic inhibitory neurotransmission to RGCs and decreased miniature glutamatergic excitatory neurotransmission, reducing the excitability of the RGCs themselves, thus alleviating the excitability of RGCs in glaucomatous slices. Conclusion Quercetin may be a promising therapeutic agent for improving RGC survival and function in glaucomatous neurodegeneration. Quercetin exerted direct protective effects on RGCs by increasing inhibitory neurotransmission and decreasing excitatory neurotransmission to RGCs, thus reducing excitotoxic damage to those cells in glaucoma.
Collapse
Affiliation(s)
- Xujiao Zhou
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai, China
| | - Gang Li
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai, China
| | - Boqi Yang
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai, China
| | - Jihong Wu
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai, China.,Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Carpanini SM, Torvell M, Morgan BP. Therapeutic Inhibition of the Complement System in Diseases of the Central Nervous System. Front Immunol 2019; 10:362. [PMID: 30886620 PMCID: PMC6409326 DOI: 10.3389/fimmu.2019.00362] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system plays critical roles in development, homeostasis, and regeneration in the central nervous system (CNS) throughout life; however, complement dysregulation in the CNS can lead to damage and disease. Complement proteins, regulators, and receptors are widely expressed throughout the CNS and, in many cases, are upregulated in disease. Genetic and epidemiological studies, cerebrospinal fluid (CSF) and plasma biomarker measurements and pathological analysis of post-mortem tissues have all implicated complement in multiple CNS diseases including multiple sclerosis (MS), neuromyelitis optica (NMO), neurotrauma, stroke, amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Given this body of evidence implicating complement in diverse brain diseases, manipulating complement in the brain is an attractive prospect; however, the blood-brain barrier (BBB), critical to protect the brain from potentially harmful agents in the circulation, is also impermeable to current complement-targeting therapeutics, making drug design much more challenging. For example, antibody therapeutics administered systemically are essentially excluded from the brain. Recent protocols have utilized "Trojan horse" techniques to transport therapeutics across the BBB or used osmotic shock or ultrasound to temporarily disrupt the BBB. Most research to date exploring the impact of complement inhibition on CNS diseases has been in animal models, and some of these studies have generated convincing data; for example, in models of MS, NMO, and stroke. There have been a few recent clinical trials of available anti-complement drugs in CNS diseases associated with BBB impairment, for example the use of the anti-C5 monoclonal antibody (mAb) eculizumab in NMO, but for most CNS diseases there have been no human trials of anti-complement therapies. Here we will review the evidence implicating complement in diverse CNS disorders, from acute, such as traumatic brain or spine injury, to chronic, including demyelinating, neuroinflammatory, and neurodegenerative diseases. We will discuss the particular problems of drug access into the CNS and explore ways in which anti-complement therapies might be tailored for CNS disease.
Collapse
Affiliation(s)
- Sarah M Carpanini
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Megan Torvell
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Bryan Paul Morgan
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom.,Division of Infection and Immunity, School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
38
|
Gaykar RN, Bhattacharjee S, Biju AT. Transition-Metal-Free Thioamination of Arynes Using Sulfenamides. Org Lett 2019; 21:737-740. [PMID: 30648876 DOI: 10.1021/acs.orglett.8b03966] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The insertion of arynes into the S-N σ-bond of sulfenamides allowing the synthesis of o-sulfanylaniline derivatives with reasonable functional group compatibility is presented. The aryne generated from 2-(trimethylsilyl)aryl triflates using CsF in DME was the key for the success of this transition-metal-free thioamination reaction, which involves new C-N and C-S bond formations in a single step under mild conditions. Moreover, the synthetic potential of this method was demonstrated by the synthesis of the antidepressant drug vortioxetine.
Collapse
Affiliation(s)
- Rahul N Gaykar
- Department of Organic Chemistry , Indian Institute of Science , Bangalore 560012 , India
| | - Subrata Bhattacharjee
- Department of Organic Chemistry , Indian Institute of Science , Bangalore 560012 , India
| | - Akkattu T Biju
- Department of Organic Chemistry , Indian Institute of Science , Bangalore 560012 , India
| |
Collapse
|
39
|
Wilkinson ST, Kiselycznyk C, Banasr M, Webler RD, Haile C, Mathew SJ. Serum and plasma brain-derived neurotrophic factor and response in a randomized controlled trial of riluzole for treatment resistant depression. J Affect Disord 2018; 241:514-518. [PMID: 30153634 PMCID: PMC6140345 DOI: 10.1016/j.jad.2018.08.075] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/02/2018] [Accepted: 08/12/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Serum brain-derived neurotrophic factor (BDNF) is decreased in individuals with major depressive disorder (MDD). Pre-clinical and clinical reports suggest that the glutamate release inhibitor riluzole increases BDNF and may have antidepressant properties. Here we report serum (sBDNF) and plasma (pBDNF) levels from a randomized controlled, adjunctive, sequential parallel comparison design trial of riluzole in MDD. METHODS Serum and plasma BDNF samples were drawn at baseline and weeks 6 and 8 from 55 subjects randomized to adjunctive treatment with riluzole or placebo for 8 weeks. RESULTS Riluzole responders had lower baseline serum (19.08 ng/ml [SD 9.22] v. 28.80 ng/ml [9.63], p = 0.08) and plasma (2.72 ng/ml [1.07] v. 4.60 ng/ml [1.69], p = 0.06) BDNF compared to non-responders at a trend level. This pattern was nominally seen in placebo responders for baseline pBDNF to some degree (1.21 ng/ml [SD 1.29] v. 3.58 ng/ml [SD 1.67], p = 0.12) but not in baseline sBDNF. LIMITATIONS A number of limitations warrant comment, including the small sample size of viable BDNF samples and the small number of riluzole responders. CONCLUSIONS Preliminary evidence reported here suggests that lower baseline BDNF may be associated with better clinical response to riluzole.
Collapse
Affiliation(s)
| | | | - Mounira Banasr
- Campbell Family Mental Health Research Institute of CAMH (Centre of Addiction and Mental Health), Toronto, ON, Canada
| | | | - Colin Haile
- Baylor College of Medicine, Houston, TX,Michael E. DeBakey VA Medical Center, Houston, TX
| | - Sanjay J. Mathew
- Baylor College of Medicine, Houston, TX,Michael E. DeBakey VA Medical Center, Houston, TX
| |
Collapse
|
40
|
McGown A, Stopford MJ. High-throughput drug screens for amyotrophic lateral sclerosis drug discovery. Expert Opin Drug Discov 2018; 13:1015-1025. [PMID: 30317895 DOI: 10.1080/17460441.2018.1533953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a rapid adult-onset neurodegenerative disorder characterised by the progressive loss of upper and lower motor neurons. Current treatment options are limited for ALS, with very modest effects on survival. Therefore, there is a unmet need for novel therapeutics to treat ALS. Areas covered: This review highlights the many diverse high-throughput screening platforms that have been implemented in ALS drug discovery. The authors discuss cell free assays including in silico and protein interaction models. The review also covers classical in vitro cell studies and new cell technologies, such as patient derived cell lines. Finally, the review looks at novel in vivo models and their use in high-throughput ALS drug discovery Expert opinion: Greater use of patient-derived in vitro cell models and development of better animal models of ALS will improve translation of lead compounds into clinic. Furthermore, AI technology is being developed to digest and interpret obtained data and to make 'hidden knowledge' usable to researchers. As a result, AI will improve target selection for high-throughput drug screening (HTDS) and aid lead compound optimisation. Furthermore, with greater genetic characterisation of ALS patients recruited to clinical trials, AI may help identify responsive genetic subtypes of patients from clinical trials.
Collapse
Affiliation(s)
- Alexander McGown
- a Sheffield Institute for Translational Neuroscience (SITraN) , University of Sheffield , Sheffield , United Kingdom
| | - Matthew John Stopford
- a Sheffield Institute for Translational Neuroscience (SITraN) , University of Sheffield , Sheffield , United Kingdom
| |
Collapse
|
41
|
Lazarevic V, Yang Y, Ivanova D, Fejtova A, Svenningsson P. Riluzole attenuates the efficacy of glutamatergic transmission by interfering with the size of the readily releasable neurotransmitter pool. Neuropharmacology 2018; 143:38-48. [PMID: 30222983 DOI: 10.1016/j.neuropharm.2018.09.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/11/2018] [Accepted: 09/12/2018] [Indexed: 01/02/2023]
Abstract
Riluzole is a potent neuroprotective agent which primarily inhibits excitatory neurotransmission interfering with presynaptic release, uptake and postsynaptic actions of glutamate by mechanisms that are not well understood. Riluzole and related prodrugs with improved blood brain barrier penetrance, are shown to be effective for the treatment of amyotrophic lateral sclerosis, ataxias, epilepsy and mood disorders. Our study was undertaken to decipher molecular and subcellular mechanisms of riluzole's antiglutamatergic effect, particularly focusing on presynaptic active zone structure and function. Applying multifarious live cell imaging techniques and amperometric glutamate recordings, we measured the impact of riluzole on presynaptic activity, synaptic vesicle recycling and glutamate release. Our in vitro and in vivo data revealed a unique mechanism whereby riluzole reduces the efficacy of glutamatergic transmission by selectively lowering the size of the readily releasable pool. This effect was correlated with the inhibition of protein kinase C-dependent Munc18-1 phosphorylation which is known to interfere with neurotransmitter release.
Collapse
Affiliation(s)
- Vesna Lazarevic
- Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Yunting Yang
- Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Daniela Ivanova
- RG Presynaptic Plasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Anna Fejtova
- RG Presynaptic Plasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany; Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Per Svenningsson
- Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
42
|
Pelletier JC, Chen S, Bian H, Shah R, Smith GR, Wrobel JE, Reitz AB. Dipeptide Prodrugs of the Glutamate Modulator Riluzole. ACS Med Chem Lett 2018; 9:752-756. [PMID: 30034613 DOI: 10.1021/acsmedchemlett.8b00189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/15/2018] [Indexed: 12/13/2022] Open
Abstract
We have previously reported a prodrug strategy based on the marketed drug riluzole (2-amino-6-trifluoromethoxybenzothiazole), associated with the benefits of lower patient to patient variability of exposure and potentially once daily oral dosing, as opposed to the large variance and twice daily dosing, which is currently observed with the parent drug. Riluzole is a glutamate modulator that is currently approved by the US FDA to treat amyotrophic lateral sclerosis (ALS). Riluzole also strongly suppresses the growth of melanoma cells that express the type 1 metabotropic glutamate receptor (GRM1, mGluR1). Riluzole is a substrate for the variably expressed liver isozyme CYP1A2, which has been shown to contribute to the variance in exposure of riluzole in humans upon oral administration. In addition, an elevated Cmax following oral administration is a probable cause of increased liver enzyme levels in some patients. In order to mitigate these issues, a series of natural and unnatural dipeptide prodrugs of riluzole were prepared as products that bear lower first-pass hepatic clearance. The prodrugs were evaluated for their ability to produce riluzole in serum while remaining intact prior to absorption from the GI tract, characteristic of a type IIB prodrug. Here, we describe dipeptide conjugates of riluzole and report that the t-Bu-Gly-Sar-riluzole analog FC-3423 (6) is absorbed well and converts to riluzole in rats and mice in a regular and well-defined manner. FC-3423 strongly suppress tumor cell growth in mouse xenograft models of melanoma at a molar dose 10-fold less than that of riluzole itself.
Collapse
Affiliation(s)
- Jeffrey C. Pelletier
- Fox Chase Chemical Diversity Center, Inc., 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Suzie Chen
- Department of Chemical Biology, Ernest Mario School of Pharmacy, 164 Frelinghuysen Road, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Haiyan Bian
- Fox Chase Chemical Diversity Center, Inc., 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Raj Shah
- Department of Chemical Biology, Ernest Mario School of Pharmacy, 164 Frelinghuysen Road, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Garry R. Smith
- Fox Chase Chemical Diversity Center, Inc., 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Jay E. Wrobel
- Fox Chase Chemical Diversity Center, Inc., 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Allen B. Reitz
- Fox Chase Chemical Diversity Center, Inc., 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
- ALS Biopharma, LLC, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| |
Collapse
|
43
|
A Randomized Placebo-Controlled Cross-Over Pilot Study of Riluzole for Drug-Refractory Irritability in Autism Spectrum Disorder. J Autism Dev Disord 2018; 48:3051-3060. [DOI: 10.1007/s10803-018-3562-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
44
|
Abstract
The amygdala is a limbic brain region that plays a key role in emotional processing, neuropsychiatric disorders, and the emotional-affective dimension of pain. Preclinical and clinical studies have identified amygdala hyperactivity as well as impairment of cortical control mechanisms in pain states. Hyperactivity of basolateral amygdala (BLA) neurons generates enhanced feedforward inhibition and deactivation of the medial prefrontal cortex (mPFC), resulting in pain-related cognitive deficits. The mPFC sends excitatory projections to GABAergic neurons in the intercalated cell mass (ITC) in the amygdala, which project to the laterocapsular division of the central nucleus of the amygdala (CeLC; output nucleus) and serve gating functions for amygdala output. Impairment of these cortical control mechanisms allows the development of amygdala pain plasticity. Mechanisms of abnormal amygdala activity in pain with particular focus on loss of cortical control mechanisms as well as new strategies to correct pain-related amygdala dysfunction will be discussed in the present review.
Collapse
|
45
|
Hardiman O, Al-Chalabi A, Chio A, Corr EM, Logroscino G, Robberecht W, Shaw PJ, Simmons Z, van den Berg LH. Amyotrophic lateral sclerosis. Nat Rev Dis Primers 2017; 3:17071. [PMID: 28980624 DOI: 10.1038/nrdp.2017.71] [Citation(s) in RCA: 842] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease, is characterized by the degeneration of both upper and lower motor neurons, which leads to muscle weakness and eventual paralysis. Until recently, ALS was classified primarily within the neuromuscular domain, although new imaging and neuropathological data have indicated the involvement of the non-motor neuraxis in disease pathology. In most patients, the mechanisms underlying the development of ALS are poorly understood, although a subset of patients have familial disease and harbour mutations in genes that have various roles in neuronal function. Two possible disease-modifying therapies that can slow disease progression are available for ALS, but patient management is largely mediated by symptomatic therapies, such as the use of muscle relaxants for spasticity and speech therapy for dysarthria.
Collapse
Affiliation(s)
- Orla Hardiman
- Academic Unit of Neurology, Room 5.41 Trinity Biomedical Science Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Adriano Chio
- Rita Levi Montalcini Department of Neurosciences, University of Turin, Turin, Italy
| | - Emma M Corr
- Academic Unit of Neurology, Room 5.41 Trinity Biomedical Science Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | | | - Wim Robberecht
- KU Leuven-University of Leuven, University Hospitals Leuven, Department of Neurology, Leuven, Belgium
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Zachary Simmons
- Department of Neurology, Milton S. Hershey Medical Center, Penn State Health, Hershey, Pennsylvania, USA
| | - Leonard H van den Berg
- Department of Neurology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
46
|
Neuronal Dysfunction in iPSC-Derived Medium Spiny Neurons from Chorea-Acanthocytosis Patients Is Reversed by Src Kinase Inhibition and F-Actin Stabilization. J Neurosci 2017; 36:12027-12043. [PMID: 27881786 DOI: 10.1523/jneurosci.0456-16.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 09/07/2016] [Accepted: 09/26/2016] [Indexed: 11/21/2022] Open
Abstract
Chorea-acanthocytosis (ChAc) is a fatal neurological disorder characterized by red blood cell acanthocytes and striatal neurodegeneration. Recently, severe cell membrane disturbances based on depolymerized cortical actin and an elevated Lyn kinase activity in erythrocytes from ChAc patients were identified. How this contributes to the mechanism of neurodegeneration is still unknown. To gain insight into the pathophysiology, we established a ChAc patient-derived induced pluripotent stem cell model and an efficient differentiation protocol providing a large population of human striatal medium spiny neurons (MSNs), the main target of neurodegeneration in ChAc. Patient-derived MSNs displayed enhanced neurite outgrowth and ramification, whereas synaptic density was similar to controls. Electrophysiological analysis revealed a pathologically elevated synaptic activity in ChAc MSNs. Treatment with the F-actin stabilizer phallacidin or the Src kinase inhibitor PP2 resulted in the significant reduction of disinhibited synaptic currents to healthy control levels, suggesting a Src kinase- and actin-dependent mechanism. This was underlined by increased G/F-actin ratios and elevated Lyn kinase activity in patient-derived MSNs. These data indicate that F-actin stabilization and Src kinase inhibition represent potential therapeutic targets in ChAc that may restore neuronal function. SIGNIFICANCE STATEMENT Chorea-acanthocytosis (ChAc) is a fatal neurodegenerative disease without a known cure. To gain pathophysiological insight, we newly established a human in vitro model using skin biopsies from ChAc patients to generate disease-specific induced pluripotent stem cells (iPSCs) and developed an efficient iPSC differentiation protocol providing striatal medium spiny neurons. Using patch-clamp electrophysiology, we detected a pathologically enhanced synaptic activity in ChAc neurons. Healthy control levels of synaptic activity could be restored by treatment of ChAc neurons with the F-actin stabilizer phallacidin and the Src kinase inhibitor PP2. Because Src kinases are involved in bridging the membrane to the actin cytoskeleton by membrane protein phosphorylation, our data suggest an actin-dependent mechanism of this dysfunctional phenotype and potential treatment targets in ChAc.
Collapse
|
47
|
Abstract
Alzheimer's disease (AD) is a major form of senile dementia, characterized by progressive memory and neuronal loss combined with cognitive impairment. AD is the most common neurodegenerative disease worldwide, affecting one-fifth of those aged over 85 years. Recent therapeutic approaches have been strongly influenced by five neuropathological hallmarks of AD: acetylcholine deficiency, glutamate excitotoxicity, extracellular deposition of amyloid-β (Aβ plague), formation of intraneuronal neurofibrillary tangles (NTFs), and neuroinflammation. The lowered concentrations of acetylcholine (ACh) in AD result in a progressive and significant loss of cognitive and behavioral function. Current AD medications, memantine and acetylcholinesterase inhibitors (AChEIs) alleviate some of these symptoms by enhancing cholinergic signaling, but they are not curative. Since 2003, no new drugs have been approved for the treatment of AD. This article focuses on the current research in clinical trials targeting the neuropathological findings of AD including acetylcholine response, glutamate transmission, Aβ clearance, tau protein deposits, and neuroinflammation. These investigations include acetylcholinesterase inhibitors, agonists and antagonists of neurotransmitter receptors, β-secretase (BACE) or γ-secretase inhibitors, vaccines or antibodies targeting Aβ clearance or tau protein, as well as anti-inflammation compounds. Ongoing Phase III clinical trials via passive immunotherapy against Aβ peptides (crenezumab, gantenerumab, and aducanumab) seem to be promising. Using small molecules blocking 5-HT6 serotonin receptor (intepirdine), inhibiting BACE activity (E2609, AZD3293, and verubecestat), or reducing tau aggregation (TRx0237) are also currently in Phase III clinical trials. We here systemically review the findings from recent clinical trials to provide a comprehensive review of novel therapeutic compounds in the treatment and prevention of AD.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402 Taiwan
- Division of Colorectal Surgery, China Medical University Hospital, Taichung, 40447 Taiwan
| | - Wen-Mei Fu
- Pharmacological Institute, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei, 10051 Taiwan
| |
Collapse
|
48
|
Ren J, Ding X, Greer JJ. Mechanistic Studies of Capsaicin-Induced Apnea in Rodents. Am J Respir Cell Mol Biol 2017; 56:252-260. [PMID: 27710012 DOI: 10.1165/rcmb.2016-0228oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Inhalation of capsaicin-based sprays can cause central respiratory depression and lethal apneas. There are contradictory reports regarding the sites of capsaicin action. Furthermore, an understanding of the neurochemical mechanisms underlying capsaicin-induced apneas and the development of pharmacological interventions is lacking. The main objectives of this study were to perform a systematic study of the mechanisms of action of capsaicin-induced apneas and to provide insights relevant to pharmacological intervention. In vitro and in vivo rat and transient receptor potential vanilloid superfamily member 1 (TRPV1)-null mouse models were used to measure respiratory parameters and seizure-like activity in the presence of capsaicin and compounds that modulate glutamatergic neurotransmission. Administration of capsaicin to in vitro and in vivo rat and wild-type mouse models induced dose-dependent apneas and the production of seizure-like activity. No significant changes were observed in TRPV1-null mice or rat medullary slice preparations. The capsaicin-induced effects were inhibited by the TRPV1 antagonist capsazepine, amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptor antagonists CNQX, NBQX, perampanel, and riluzole, a drug that inhibits glutamate release and increases glutamate uptake. The capsaicin-induced effects on breathing and seizure-like activity were accentuated by positive allosteric modulators of the AMPA receptors, CX717 and cyclothiazide. To summarize, capsaicin-induced apneas and seizure-like behaviors are mediated via TRPV1 activation acting at lung afferents, spinal cord-ascending tracts, and medullary structures (including nucleus tractus solitarius). AMPA receptor-mediated conductances play an important role in capsaicin-induced apneas and seizure-like activity. A pharmaceutical strategy targeted at reducing AMPA receptor-mediated glutamatergic signaling may reduce capsaicin-induced deleterious effects.
Collapse
Affiliation(s)
- Jun Ren
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Xiuqing Ding
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - John J Greer
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
49
|
Luo D, Ma R, Wu Y, Zhang X, Liu Y, Wang L, Fu W. Mechanism Underlying Acupuncture-Ameliorated Depressive Behaviors by Enhancing Glial Glutamate Transporter in Chronic Unpredictable Mild Stress (CUMS) Rats. Med Sci Monit 2017. [PMID: 28644824 PMCID: PMC5493061 DOI: 10.12659/msm.902549] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background Major depressive disorder (MDD) is a recurrent mental illness worldwide. The glutamatergic neurotransmission system is now a target for antidepressant therapy because it takes part in synaptic plasticity and cognition in physical condition and has a potential excitatory neurotoxicity in pathological conditions. Glial glutamate transporter EAAT2 performs 90% of Glu neurotransmission. Therefore, the aim of the study was to evaluate the effect of acupuncture on depressive behaviors and EAAT2 in CUMS. Material/Methods We randomly divided 56 male SD rats into a normal group, a model group, an acupuncture group, and a riluzole group. Rats in the model group, acupuncture group, and riluzole group underwent chronic unpredictable mild stress (CUMS) exposure for 21 days. The acupuncture group received electro-acupuncture stimulation on LI4 and LR3 for 5 continuous days per week for 4 weeks, and rats in the riluzole group received 4 mg/kg of riluzole orally (Sanofi, J20140092) for 4 weeks after undergoing CUMS stimulation. Results Rats showed significantly increased sucrose consumption in the sucrose preference test paradigm, and showed elevated food intake and shortened latency in the novelty-suppressed feeding test paradigm after undergoing acupuncture therapy and riluzole treatment. The amelioration of depressive behavioral actions was consistent with increasing number of positive cells, protein, and mRNA expression of glial glutamate transporter EAAT2 in the hippocampus and PFC. Conclusions The results suggest that acupuncture and riluzole are both effective in improving sucrose consumption, latency, and food intake in CUMS rats. However, acupuncture appears to achieve an antidepressant effect later than riluzole does because it might need accumulated stimulation by enhancing EAAT2 expression. Enhance glial glutamate transporter EAAT2 in the hippocampus and PFC is a mechanism underlying the antidepressant effect of acupuncture.
Collapse
Affiliation(s)
- Ding Luo
- The Second Clinical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland).,Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland)
| | - Rui Ma
- Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland)
| | - Yanan Wu
- Department of Traditional Chinese Medicine, The Fourth Hosipital of Changsha City, Changsha, Hunan, China (mainland)
| | - Xuechun Zhang
- The Second Clinical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland)
| | - Yue Liu
- The Second Clinical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland)
| | - Lin Wang
- Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland)
| | - Wenbin Fu
- The Second Clinical College, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland).,Department of Acupuncture and Moxibustion, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
50
|
Abstract
BACKGROUND Glutamatergic system abnormalities are implicated in the pathophysiology and treatment of both major depressive disorder and bipolar depression (BDep). Subsequent to studies demonstrating the rapid and robust antidepressant effects of ketamine, an N-methyl-D-aspartate receptor antagonist, other glutamatergic modulators are now being studied in clinical trials of mood disorders. A previous open-label study found that riluzole, administered in combination with the mood stabilizer lithium, had antidepressant effects. METHODS We conducted a randomized, double-blind, placebo-controlled trial of riluzole monotherapy for the treatment of BDep. Nineteen subjects aged 18 to 70 years with bipolar disorder currently experiencing a depressive episode were tapered off of excluded medications and randomized to receive riluzole (50-200 mg/d) or placebo for 8 weeks. Rating scale scores (Montgomery-Åsberg Depression Rating Scale, Hamilton Rating Scale for Depression, Hamilton Rating Scale for Anxiety, and Young Mania Rating Scale) were obtained weekly. RESULTS No significant differences in depressive symptoms were observed between subjects treated with riluzole and those receiving placebo (P = 0.12). Anxiety scores were significantly lower in the placebo group (P = 0.046). An interim analysis was conducted that resulted in stopping the study because of futility; no subjects had achieved treatment response. CONCLUSIONS Although we found no change in severity of depressive symptoms in BDep patients receiving riluzole compared with placebo, this trial was limited by the relatively high number of subject withdrawals and the small sample size. Thus, while riluzole monotherapy did not demonstrate efficacy for BDep, further studies examining riluzole as adjunctive therapy for this disorder may be warranted.Clinical Trials Identifier NCT00054704.
Collapse
|