1
|
Sternberg Z. Neurodegenerative Etiology of Aromatic L-Amino Acid Decarboxylase Deficiency: a Novel Concept for Expanding Treatment Strategies. Mol Neurobiol 2024; 61:2996-3018. [PMID: 37953352 DOI: 10.1007/s12035-023-03684-2] [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: 02/07/2023] [Accepted: 09/29/2023] [Indexed: 11/14/2023]
Abstract
Aromatic l-amino acid decarboxylase deficiency (AADC-DY) is caused by one or more mutations in the DDC gene, resulting in the deficit in catecholamines and serotonin neurotransmitters. The disease has limited therapeutic options with relatively poor clinical outcomes. Accumulated evidence suggests the involvement of neurodegenerative mechanisms in the etiology of AADC-DY. In the absence of neurotransmitters' neuroprotective effects, the accumulation and the chronic presence of several neurotoxic metabolites including 4-dihydroxy-L-phenylalanine, 3-methyldopa, and homocysteine, in the brain of subjects with AADC-DY, promote oxidative stress and reduce the cellular antioxidant and methylation capacities, leading to glial activation and mitochondrial dysfunction, culminating to neuronal injury and death. These pathophysiological processes have the potential to hinder the clinical efficacy of treatments aimed at increasing neurotransmitters' synthesis and or function. This review describes in detail the mechanisms involved in AADC-DY neurodegenerative etiology, highlighting the close similarities with those involved in other neurodegenerative diseases. We then offer novel strategies for the treatment of the disease with the objective to either reduce the level of the metabolites or counteract their prooxidant and neurotoxic effects. These treatment modalities used singly or in combination, early in the course of the disease, will minimize neuronal injury, preserving the functional integrity of neurons, hence improving the clinical outcomes of both conventional and unconventional interventions in AADC-DY. These modalities may not be limited to AADC-DY but also to other metabolic disorders where a specific mutation leads to the accumulation of prooxidant and neurotoxic metabolites.
Collapse
Affiliation(s)
- Zohi Sternberg
- Jacobs School of Medicine and Biomedical Sciences, Buffalo Medical Center, Buffalo, NY, 14203, USA.
| |
Collapse
|
2
|
Raghib MF, Bernitsas E. From Animal Models to Clinical Trials: The Potential of Antimicrobials in Multiple Sclerosis Treatment. Biomedicines 2023; 11:3069. [PMID: 38002068 PMCID: PMC10668955 DOI: 10.3390/biomedicines11113069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, demyelinating disease of the central nervous system (CNS). Microbes, including bacteria and certain viruses, particularly Epstein-Barr virus (EBV), have been linked to the pathogenesis of MS. While there is currently no cure for MS, antibiotics and antivirals have been studied as potential treatment options due to their immunomodulatory ability that results in the regulation of the immune process. The current issue addressed in this systematic review is the effect of antimicrobials, including antibiotics, antivirals, and antiparasitic agents in animals and humans. We performed a comprehensive search of PubMed, Google Scholar, and Scopus for articles on antimicrobials in experimental autoimmune encephalomyelitis animal models of MS, as well as in people with MS (pwMS). In animal models, antibiotics tested included beta-lactams, minocycline, rapamycin, macrolides, and doxycycline. Antivirals included acyclovir, valacyclovir, and ganciclovir. Hydroxychloroquine was the only antiparasitic that was tested. In pwMS, we identified a total of 24 studies, 17 of them relevant to antibiotics, 6 to antivirals, and 1 relevant to antiparasitic hydroxychloroquine. While the effect of antimicrobials in animal models was promising, only minocycline and hydroxychloroquine improved outcome measures in pwMS. No favorable effect of the antivirals in humans has been observed yet. The number and size of clinical trials testing antimicrobials have been limited. Large, multicenter, well-designed studies are needed to further evaluate the effect of antimicrobials in MS.
Collapse
Affiliation(s)
- Muhammad Faraz Raghib
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Evanthia Bernitsas
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Sastry Neuroimaging Laboratory, Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
3
|
Degirmenci Y, Angelopoulou E, Georgakopoulou VE, Bougea A. Cognitive Impairment in Parkinson's Disease: An Updated Overview Focusing on Emerging Pharmaceutical Treatment Approaches. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1756. [PMID: 37893474 PMCID: PMC10608778 DOI: 10.3390/medicina59101756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/17/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023]
Abstract
Cognitive impairment in patients with Parkinson's disease (PD) is one of the commonest and most disabling non-motor manifestations during the course of the disease. The clinical spectrum of PD-related cognitive impairment includes subjective cognitive decline (SCD), mild cognitive impairment (MCI) and PD dementia (PDD). As the disease progresses, cognitive decline creates a significant burden for the family members and/or caregivers of patients with PD, and has a great impact on quality of life. Current pharmacological treatments have demonstrated partial efficacy and failed to halt disease progression, and novel, effective, and safe therapeutic strategies are required. Accumulating preclinical and clinical evidence shows that several agents may provide beneficial effects on patients with PD and cognitive impairment, including ceftriaxone, ambroxol, intranasal insulin, nilotinib, atomoxetine, mevidalen, blarcamesine, prasinezumab, SYN120, ENT-01, NYX-458, GRF6021, fosgonimeton, INT-777, Neuropeptide S, silibinin, osmotin, cordycepin, huperzine A, fibroblast growth factor 21, Poloxamer 188, ginsenoside Rb1, thioredoxin-1, tangeretin, istradefylline and Eugenia uniflora. Potential underlying mechanisms include the inhibition of a-synuclein aggregation, the improvement of mitochondrial function, the regulation of synaptic plasticity, an impact on the gut-brain axis, the modulation of neuroinflammation and the upregulation of neurotrophic factors, as well as cholinergic, dopaminergic, serotoninergic and norepinephrine neurotransmission. In this updated overview, we aim to cover the clinical aspects of the spectrum of PD-related cognitive impairment and discuss recent evidence on emerging treatment approaches that are under investigation at a preclinical and clinical level. Finally, we aim to provide additional insights and propose new ideas for investigation that may be feasible and effective for the spectrum of PD-related cognitive impairment.
Collapse
Affiliation(s)
- Yildiz Degirmenci
- Department of Neurology, School of Medicine, Istanbul Health and Technology University, 34093 Istanbul, Turkey;
- Parkinson’s Disease and Movement Disorders Unit, Neurology Clinic, Sisli Kolan International Hospital, 34384 Istanbul, Turkey
| | - Efthalia Angelopoulou
- 1st Department of Neurology, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece;
| | | | - Anastasia Bougea
- 1st Department of Neurology, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece;
| |
Collapse
|
4
|
Bilal W, Khawar MB, Afzal A, Naseer A, Hamid SE, Shahzaman S, Qamar F. Recent advances to Neuroprotection: repurposing drugs against neuroinflammatory disorders. Mol Biol Rep 2023:10.1007/s11033-023-08490-6. [PMID: 37231215 DOI: 10.1007/s11033-023-08490-6] [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: 12/22/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023]
Abstract
Cell death is a natural mechanism for biological clearance for the maintenance of homeostasis in a dynamic microenvironment of the central nervous system. Stress and various factors can lead to imbalance between cellular genesis and cell death leading to dysfunctionality and a number of neuropathological disorders. Drug repurposing can help bypass development time and cost. A complete understanding of drug actions and neuroinflammatory pathways can lead to effective control of neurodegenerative disorders. This review covers recent advances in various neuroinflammatory pathways understanding, biomarkers, and drug repurposing for neuroprotection.
Collapse
Affiliation(s)
- Wishah Bilal
- Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Babar Khawar
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan.
| | - Ali Afzal
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences & Technology, University of Central Punjab, Lahore, Pakistan
| | - Arshia Naseer
- Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Syeda Eisha Hamid
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences & Technology, University of Central Punjab, Lahore, Pakistan
| | - Sara Shahzaman
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences & Technology, University of Central Punjab, Lahore, Pakistan
| | | |
Collapse
|
5
|
Suárez-Rivero JM, López-Pérez J, Muela-Zarzuela I, Pastor-Maldonado C, Cilleros-Holgado P, Gómez-Fernández D, Álvarez-Córdoba M, Munuera-Cabeza M, Talaverón-Rey M, Povea-Cabello S, Suárez-Carrillo A, Piñero-Pérez R, Reche-López D, Romero-Domínguez JM, Sánchez-Alcázar JA. Neurodegeneration, Mitochondria, and Antibiotics. Metabolites 2023; 13:metabo13030416. [PMID: 36984858 PMCID: PMC10056573 DOI: 10.3390/metabo13030416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Neurodegenerative diseases are characterized by the progressive loss of neurons, synapses, dendrites, and myelin in the central and/or peripheral nervous system. Actual therapeutic options for patients are scarce and merely palliative. Although they affect millions of patients worldwide, the molecular mechanisms underlying these conditions remain unclear. Mitochondrial dysfunction is generally found in neurodegenerative diseases and is believed to be involved in the pathomechanisms of these disorders. Therefore, therapies aiming to improve mitochondrial function are promising approaches for neurodegeneration. Although mitochondrial-targeted treatments are limited, new research findings have unraveled the therapeutic potential of several groups of antibiotics. These drugs possess pleiotropic effects beyond their anti-microbial activity, such as anti-inflammatory or mitochondrial enhancer function. In this review, we will discuss the controversial use of antibiotics as potential therapies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Juan M. Suárez-Rivero
- Institute for Biomedical Researching and Innovation of Cádiz (INiBICA) University Hospital Puerta del Mar, 11009 Cádiz, Spain
| | - Juan López-Pérez
- Institute for Biomedical Researching and Innovation of Cádiz (INiBICA) University Hospital Puerta del Mar, 11009 Cádiz, Spain
| | - Inés Muela-Zarzuela
- Institute for Biomedical Researching and Innovation of Cádiz (INiBICA) University Hospital Puerta del Mar, 11009 Cádiz, Spain
| | - Carmen Pastor-Maldonado
- Department of Molecular Biology Interfaculty Institute for Cell Biology, University of Tuebingen, D-72076 Tuebingen, Germany
| | - Paula Cilleros-Holgado
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - David Gómez-Fernández
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - Mónica Álvarez-Córdoba
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - Manuel Munuera-Cabeza
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - Marta Talaverón-Rey
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - Suleva Povea-Cabello
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - Alejandra Suárez-Carrillo
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - Rocío Piñero-Pérez
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - Diana Reche-López
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - José M. Romero-Domínguez
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
| | - José Antonio Sánchez-Alcázar
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide-University), 41013 Sevilla, Spain
- Correspondence: ; Tel.: +34-954978071
| |
Collapse
|
6
|
The Interplay between Gut Microbiota and Parkinson's Disease: Implications on Diagnosis and Treatment. Int J Mol Sci 2022; 23:ijms232012289. [PMID: 36293176 PMCID: PMC9603886 DOI: 10.3390/ijms232012289] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
Abstract
The bidirectional interaction between the gut microbiota (GM) and the Central Nervous System, the so-called gut microbiota brain axis (GMBA), deeply affects brain function and has an important impact on the development of neurodegenerative diseases. In Parkinson’s disease (PD), gastrointestinal symptoms often precede the onset of motor and non-motor manifestations, and alterations in the GM composition accompany disease pathogenesis. Several studies have been conducted to unravel the role of dysbiosis and intestinal permeability in PD onset and progression, but the therapeutic and diagnostic applications of GM modifying approaches remain to be fully elucidated. After a brief introduction on the involvement of GMBA in the disease, we present evidence for GM alterations and leaky gut in PD patients. According to these data, we then review the potential of GM-based signatures to serve as disease biomarkers and we highlight the emerging role of probiotics, prebiotics, antibiotics, dietary interventions, and fecal microbiota transplantation as supportive therapeutic approaches in PD. Finally, we analyze the mutual influence between commonly prescribed PD medications and gut-microbiota, and we offer insights on the involvement also of nasal and oral microbiota in PD pathology, thus providing a comprehensive and up-to-date overview on the role of microbial features in disease diagnosis and treatment.
Collapse
|
7
|
Lin CL, Zheng TL, Tsou SH, Chang HM, Tseng LH, Yu CH, Hung CS, Ho YJ. Amitriptyline Improves Cognitive and Neuronal Function in a Rat Model that Mimics Dementia with Lewy Bodies. Behav Brain Res 2022; 435:114035. [PMID: 35926562 DOI: 10.1016/j.bbr.2022.114035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/09/2022] [Accepted: 07/28/2022] [Indexed: 11/19/2022]
Abstract
Dementia with Lewy bodies (DLB), a highly prevalent neurodegenerative disorder, causes motor and cognitive deficits. The main pathophysiologies of DLB are glutamate excitotoxicity and accumulation of Lewy bodies comprising α-synuclein (α-syn) and β-amyloid (Aβ). Amitriptyline (AMI) promotes expression of glutamate transporter-1 and glutamate reuptake. In this study, we measured the effects of AMI on behavioral and neuronal function in a DLB rat model. We used rivastigmine (RIVA) as a positive control. To establish the DLB rat model, male Wistar rats were stereotaxically injected with recombinant adenoassociated viral vector with the SNCA gene (10μg/10μL) and Aβ (5μg/2.5μL) into the left ventricle and prefrontal cortex, respectively. AMI (10mg/kg/day, i.p.), RIVA (2mg/kg/day, i.p.), or saline was injected intraperitoneally after surgery. From the 29th day, behavioral tests were performed to evaluate the motor and cognitive functions of the rats. Immunohistochemical staining was used to assess neuronal changes. We measured the α-syn level, number of newborn cells, and neuronal density in the hippocampus and in the nigrostriatal dopaminergic system. The DLB group exhibited deficit in object recognition. Both the AMI and RIVA treatments reversed these deficits. Histologically, the DLB rats exhibited cell loss in the substantia nigra pars compacta and in the hippocampal CA1 area. AMI reduced this cell loss, but RIVA did not. In addition, the DLB rats exhibited a lower number of newborn cells and higher α-syn levels in the dentate gyrus (DG). AMI did not affect α-syn accumulation but recovered neurogenesis in the DG of the rats, whereas RIVA reversed the α-syn accumulation but did not affect neurogenesis in the rats. We suggest that AMI may have potential for use in the treatment of DLB.
Collapse
Affiliation(s)
- Chih-Li Lin
- Institute of Medicine, Department of Medical Research, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Ting-Lin Zheng
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Sing-Hua Tsou
- Institute of Medicine, Department of Medical Research, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 40201, Taiwan, ROC
| | - Hung-Ming Chang
- Department of Anantomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, ROC
| | - Li-Ho Tseng
- Graduate School of Environmental Management, Tajen University, Pingtung 907, Taiwan, ROC
| | - Ching-Han Yu
- Department of Pysiology, School of Medicine, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.
| | - Ching-Sui Hung
- Occupational Safety and Health Office, Taipei City Hospital, Taipei 10581, Taiwan, ROC.
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 40201, Taiwan, ROC.
| |
Collapse
|
8
|
Tikhonova MA, Chang HM, Singh SK, Vieau D. Editorial: Experimental and Innovative Approaches to Multi-Target Treatment of Parkinson's and Alzheimer's Diseases. Front Neurosci 2022; 16:910020. [PMID: 35651630 PMCID: PMC9150500 DOI: 10.3389/fnins.2022.910020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 01/01/2023] Open
Affiliation(s)
- Maria A. Tikhonova
- Laboratory of the Experimental Models of Neurodegenerative Processes, Department of Experimental Neuroscience, Scientific Research Institute of Neurosciences and Medicine (SRINM), Novosibirsk, Russia
- *Correspondence: Maria A. Tikhonova
| | - Hung-Ming Chang
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Didier Vieau
- U1172 - LilNCog - Lille Neuroscience and Cognition, Alzheimer and Tauopathies, Université de Lille, Lille, France
| |
Collapse
|
9
|
Fan Y, Han J, Zhao L, Wu C, Wu P, Huang Z, Hao X, Ji Y, Chen D, Zhu M. Experimental Models of Cognitive Impairment for Use in Parkinson's Disease Research: The Distance Between Reality and Ideal. Front Aging Neurosci 2021; 13:745438. [PMID: 34912207 PMCID: PMC8667076 DOI: 10.3389/fnagi.2021.745438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/01/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. Cognitive impairment is one of the key non-motor symptoms of PD, affecting both mortality and quality of life. However, there are few experimental studies on the pathology and treatments of PD with mild cognitive impairment (PD-MCI) and PD dementia (PDD) due to the lack of representative models. To identify new strategies for developing representative models, we systematically summarized previous studies on PD-MCI and PDD and compared differences between existing models and diseases. Our initial search identified 5432 articles, of which 738 were duplicates. A total of 227 articles met our inclusion criteria and were included in the analysis. Models fell into three categories based on model design: neurotoxin-induced, transgenic, and combined. Although the neurotoxin-induced experimental model was the most common type that was used during every time period, transgenic and combined experimental models have gained significant recent attention. Unfortunately, there remains a big gap between ideal and actual experimental models. While each model has its own disadvantages, there have been tremendous advances in the development of PD models of cognitive impairment, and almost every model can verify a hypothesis about PD-MCI or PDD. Finally, our proposed strategies for developing novel models are as follows: a set of plans that integrate symptoms, biochemistry, neuroimaging, and other objective indicators to judge and identify that the novel model plays a key role in new strategies for developing representative models; novel models should simulate different clinical features of PD-MCI or PDD; inducible α-Syn overexpression and SH-SY5Y-A53T cellular models are good candidate models of PD-MCI or PDD.
Collapse
Affiliation(s)
- Yaohua Fan
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jiajun Han
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijun Zhao
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Chunxiao Wu
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China.,Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peipei Wu
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zifeng Huang
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiaoqian Hao
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - YiChun Ji
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Dongfeng Chen
- Traditional Chinese Medicine Innovation Research Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Meiling Zhu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
10
|
Mitochondrial Metabolism as Target of the Neuroprotective Role of Erythropoietin in Parkinson's Disease. Antioxidants (Basel) 2021; 10:antiox10010121. [PMID: 33467745 PMCID: PMC7830512 DOI: 10.3390/antiox10010121] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/30/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
Existing therapies for Parkinson's disease (PD) are only symptomatic. As erythropoietin (EPO) is emerging for its benefits in neurodegenerative diseases, here, we test the protective effect driven by EPO in in vitro (SH-SY5Y cells challenged by MPP+) and in vivo (C57BL/6J mice administered with MPTP) PD models. EPO restores cell viability in both protective and restorative layouts, enhancing the dopaminergic recovery. Specifically, EPO rescues the PD-induced damage to mitochondria, as shown by transmission electron microscopy, Mitotracker assay and PINK1 expression. Moreover, EPO promotes a rescue of mitochondrial respiration while markedly enhancing the glycolytic rate, as shown by the augmented extracellular acidification rate, contributing to elevated ATP levels in MPP+-challenged cells. In PD mice, EPO intrastriatal infusion markedly improves the outcome of behavioral tests. This is associated with the rescue of dopaminergic markers and decreased neuroinflammation. This study demonstrates cellular and functional recovery following EPO treatment, likely mediated by the 37 Kda isoform of the EPO-receptor. We report for the first time, that EPO-neuroprotection is exerted through restoring ATP levels by accelerating the glycolytic rate. In conclusion, the redox imbalance and neuroinflammation associated with PD may be successfully treated by EPO.
Collapse
|
11
|
Thompson A, Farmer K, Rowe E, Hayley S. Erythropoietin modulates striatal antioxidant signalling to reduce neurodegeneration in a toxicant model of Parkinson's disease. Mol Cell Neurosci 2020; 109:103554. [DOI: 10.1016/j.mcn.2020.103554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
|
12
|
Smaga I, Fierro D, Mesa J, Filip M, Knackstedt LA. Molecular changes evoked by the beta-lactam antibiotic ceftriaxone across rodent models of substance use disorder and neurological disease. Neurosci Biobehav Rev 2020; 115:116-130. [PMID: 32485268 DOI: 10.1016/j.neubiorev.2020.05.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
Ceftriaxone is a beta-lactam antibiotic that increases the expression of the major glutamate transporter, GLT-1. As such, ceftriaxone ameliorates symptoms across multiple rodent models of neurological diseases and substance use disorders. However, the mechanism behind GLT-1 upregulation is unknown. The present review synthesizes this literature in order to identify commonalities in molecular changes. We find that ceftriaxone (200 mg/kg for at least two days) consistently restores GLT-1 expression in multiple rodent models of neurological disease, especially when GLT-1 is decreased in the disease model. The same dose given to healthy/drug-naive rodents does not reliably upregulate GLT-1 in any brain region except the hippocampus. Increased GLT-1 expression does not consistently arise from transcriptional regulation, and is likely to be due to trafficking changes. In addition to altered transporter expression, ceftriaxone ameliorates neuropathologies (e.g. tau, amyloid beta, cell death) and aberrant protein expression associated with a number of neurological disease models. Taken together, these results indicate that ceftriaxone remains a strong candidate for treatment of multiple disorders in the clinic.
Collapse
Affiliation(s)
- Irena Smaga
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL, 31-343, Kraków, Poland
| | - Daniel Fierro
- Department of Psychology, University of Florida, 945 Center Dr., Gainesville, FL, 32611, USA
| | - Javier Mesa
- Department of Psychology, University of Florida, 945 Center Dr., Gainesville, FL, 32611, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32611, USA
| | - Malgorzata Filip
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL, 31-343, Kraków, Poland
| | - Lori A Knackstedt
- Department of Psychology, University of Florida, 945 Center Dr., Gainesville, FL, 32611, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
13
|
Chang CC, Li HH, Tsou SH, Hung HC, Liu GY, Korolenko TA, Lai TJ, Ho YJ, Lin CL. The Pluripotency Factor Nanog Protects against Neuronal Amyloid β-Induced Toxicity and Oxidative Stress through Insulin Sensitivity Restoration. Cells 2020; 9:cells9061339. [PMID: 32471175 PMCID: PMC7348813 DOI: 10.3390/cells9061339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 11/16/2022] Open
Abstract
Amyloid β (Aβ) is a peptide fragment of the amyloid precursor protein that triggers the progression of Alzheimer's Disease (AD). It is believed that Aβ contributes to neurodegeneration in several ways, including mitochondria dysfunction, oxidative stress and brain insulin resistance. Therefore, protecting neurons from Aβ-induced neurotoxicity is an effective strategy for attenuating AD pathogenesis. Recently, applications of stem cell-based therapies have demonstrated the ability to reduce the progression and outcome of neurodegenerative diseases. Particularly, Nanog is recognized as a stem cell-related pluripotency factor that enhances self-renewing capacities and helps reduce the senescent phenotypes of aged neuronal cells. However, whether the upregulation of Nanog can be an effective approach to alleviate Aβ-induced neurotoxicity and senescence is not yet understood. In the present study, we transiently overexpressed Nanog-both in vitro and in vivo-and investigated the protective effects and underlying mechanisms against Aβ. We found that overexpression of Nanog is responsible for attenuating Aβ-triggered neuronal insulin resistance, which restores cell survival through reducing intracellular mitochondrial superoxide accumulation and cellular senescence. In addition, upregulation of Nanog expression appears to increase secretion of neurotrophic factors through activation of the Nrf2 antioxidant defense pathway. Furthermore, improvement of memory and learning were also observed in rat model of Aβ neurotoxicity mediated by upregulation of Nanog in the brain. Taken together, our study suggests a potential role for Nanog in attenuating the neurotoxic effects of Aβ, which in turn, suggests that strategies to enhance Nanog expression may be used as a novel intervention for reducing Aβ neurotoxicity in the AD brain.
Collapse
Affiliation(s)
- Ching-Chi Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
| | - Hsin-Hua Li
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
| | - Sing-Hua Tsou
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
| | - Hui-Chih Hung
- Department of Life Sciences and Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402204, Taiwan;
| | - Guang-Yaw Liu
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
| | - Tatiana A. Korolenko
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk 630117, Russia;
| | - Te-Jen Lai
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University, Taichung 402367, Taiwan
- Correspondence: (Y.-J.H.); (C.-L.L.); Tel.: +886-4-2473-0022-11673 (Y.-J.H.); +886-4-2473-0022-11690 (C.-L.L.)
| | - Chih-Li Lin
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
- Correspondence: (Y.-J.H.); (C.-L.L.); Tel.: +886-4-2473-0022-11673 (Y.-J.H.); +886-4-2473-0022-11690 (C.-L.L.)
| |
Collapse
|
14
|
Korolenko TA, Shintyapina AB, Pupyshev AB, Akopyan AA, Russkikh GS, Dikovskaya MA, Vavilin VA, Zavjalov EL, Tikhonova MA, Amstislavskaya TG. The regulatory role of cystatin C in autophagy and neurodegeneration. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Autophagy is a dynamic cellular process involved in the turnover of proteins, protein complexes, and organelles through lysosomal degradation. It is particularly important in neurons, which do not have a proliferative option for cellular repair. Autophagy has been shown to be suppressed in the striatum of a transgenic mouse model of Parkinson’s disease. Cystatin C is one of the potent regulators of autophagy. Changes in the expression and secretion of cystatin C in the brain have been shown in amyotrophic lateral sclerosis, Alzheimer’s and Parkinson’s diseases, and in some animal models of neurodegeneration, thus proving a protective function of cystatin C. It has been suggested that cystatin C plays the primary role in amyloidogenesis and shows promise as a therapeutic agent for neurodegenerative diseases (Alzheimer’s and Parkinson’s diseases). Cystatin C colocalizes with the amyloid β-protein in the brain during Alzheimer’s disease. Controlled expression of a cystatin C peptide has been proposed as a new approach to therapy for Alzheimer’s disease. In Parkinson’s disease, serum cystatin C levels can predict disease severity and cognitive dysfunction, although the exact involvement of cystatin C remains unclear. The aim: to study the role of cystatin C in neurodegeneration and evaluate the results in relation to the mechanism of autophagy. In our study on humans, a higher concentration of cystatin C was noted in cerebrospinal fluid than in serum; much lower concentrations were observed in other biological fluids (intraocular fluid, bile, and sweat). In elderly persons (61–80 years old compared to practically healthy people at 40–60 years of age), we revealed increased cystatin C levels both in serum and intraocular fluid. In an experiment on C57Bl/6J mice, cystatin C concentration was significantly higher in brain tissue than in the liver and spleen: an indication of an important function of this cysteine protease inhibitor in the brain. Using a transgenic mouse model of Parkinson’s disease (5 months old), we demonstrated a significant increase in osmotic susceptibility of brain lysosomes, depending on autophagy, while in a murine model of Alzheimer’s disease, this parameter did not differ from that in the appropriate control.
Collapse
Affiliation(s)
- T. A. Korolenko
- Scientific Research Institute of Physiology and Basic Medicine
| | - A. B. Shintyapina
- Scientific Research Institute of Molecular Biology and Biophysics, Federal Research Center for Basic and Translational Medicine
| | - A. B. Pupyshev
- Scientific Research Institute of Physiology and Basic Medicine
| | - A. A. Akopyan
- Scientific Research Institute of Physiology and Basic Medicine
| | - G. S. Russkikh
- Scientific Research Institute of Biochemistry, Federal Research Center for Basic and Translational Medicine
| | - M. A. Dikovskaya
- Scientific Research Institute of Physiology and Basic Medicine; S.N. Fedorov NMRC “MNTK “Eye Microsurgery”, Novosibirsk Branch
| | - V. A. Vavilin
- Scientific Research Institute of Molecular Biology and Biophysics, Federal Research Center for Basic and Translational Medicine; Institute of Cytology and Genetics, SB RAS
| | | | - M. A. Tikhonova
- Scientific Research Institute of Physiology and Basic Medicine; Novosibirsk State University
| | - T. G. Amstislavskaya
- Scientific Research Institute of Physiology and Basic Medicine; Novosibirsk State University
| |
Collapse
|
15
|
Ho YJ, Shen MS, Tai CH, Li HH, Chen JH, Liao WC, Chiu PY, Lee IY, Lin CL, Hung CS. Use of Ceftriaxone in Treating Cognitive and Neuronal Deficits Associated With Dementia With Lewy Bodies. Front Neurosci 2019; 13:507. [PMID: 31178684 PMCID: PMC6543807 DOI: 10.3389/fnins.2019.00507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/02/2019] [Indexed: 01/08/2023] Open
Abstract
Dementia with Lewy bodies (DLB) is caused by accumulation of Lewy bodies, destruction of mitochondria, and excess of glutamate in synapses, which eventually leads to excitotoxicity, neurodegeneration, and cognitive impairments. Ceftriaxone (CEF) reduces excitotoxicity by increasing glutamate transporter 1 expression and glutamate reuptake. We investigated whether CEF can prevent cognitive decline and neurological deficits and increase neurogenesis in DLB rats. Male Wistar rats infused with viral vector containing human alpha-synuclein (α-syn) gene, SNCA, in the lateral ventricle were used as a rat model of DLB. CEF (100 mg/kg/day, i.p.) was injected in these rats for 27 days. The active avoidance test and object recognition test was performed. Finally, the brains of all the rats were immunohistochemically stained to measure α-syn, neuronal density, and newborn cells in the hippocampus and substantia nigra. The results revealed that DLB rats had learning and object recognition impairments and exhibited cell loss in the nigrostriatal dopaminergic system, and hippocampal CA1, and dentate gyrus (DG). Additionally, DLB rats had fewer newborn cells in the DG and substantia nigra pars reticulata and more α-syn immune-positive cells in the DG. Treatment with CEF improved cognitive function, reduced cell loss, and increased the number of newborn cells in the brain. To our knowledge, this is the first study showing that CEF prevents loss of neurogenesis in the brain of DLB rats. CEF may therefore has clinical potential for treating DLB.
Collapse
Affiliation(s)
- Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University Hospital - Chung Shan Medical University, Taichung, Taiwan
| | - Mei-Shiuan Shen
- Department of Psychology, Chung Shan Medical University Hospital - Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Hwei Tai
- Department of Neurology, College of Medicine, National Taiwan University Hospital - National Taiwan University, Taipei, Taiwan
| | - Hsin-Hua Li
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jian-Horng Chen
- School of Physical Therapy, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Chieh Liao
- Department of Anatomy - Department of Pediatrics, Faculty of Medicine, Chung Shan Medical University Hospital - Chung Shan Medical University, Taichung, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - I-Yen Lee
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Chih-Li Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ching-Sui Hung
- Occupational Safety and Health Office, Taipei City Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
Tai CH, Bellesi M, Chen AC, Lin CL, Li HH, Lin PJ, Liao WC, Hung CS, Schwarting RK, Ho YJ. A new avenue for treating neuronal diseases: Ceftriaxone, an old antibiotic demonstrating behavioral neuronal effects. Behav Brain Res 2019; 364:149-156. [DOI: 10.1016/j.bbr.2019.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/14/2019] [Accepted: 02/12/2019] [Indexed: 12/27/2022]
|
17
|
Yimer EM, Hishe HZ, Tuem KB. Repurposing of the β-Lactam Antibiotic, Ceftriaxone for Neurological Disorders: A Review. Front Neurosci 2019; 13:236. [PMID: 30971875 PMCID: PMC6444273 DOI: 10.3389/fnins.2019.00236] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
To date, there is no cure or disease-modifying agents available for most well-known neurological disorders. Current therapy is typically focused on relieving symptoms and supportive care in improving the quality of life of affected patients. Furthermore, the traditional de novo drug discovery technique is more challenging, particularly for neurological disorders. Therefore, the repurposing of existing drugs for these conditions is believed to be an efficient and dynamic approach that can substantially reduce the investments spent on drug development. Currently, there is emerging evidence that suggests the potential effect of a beta-lactam antibiotic, ceftriaxone (CEF), to alleviate the symptoms of different experimentally-induced neurological disorders: Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, epileptic-seizure, brain ischemia, traumatic brain injuries, and neuropathic pain. CEF also affects the markers of oxidative status and neuroinflammation, glutamatergic systems as well as various aggregated toxic proteins involved in the pathogenesis of different neurological disorders. Moreover, it was found that CEF administration to drug dependent animal models improved the withdrawal symptoms upon drug discontinuation. Thus, this review aimed to describe the effects of CEF against multiple models of neurological illnesses, drug dependency, and withdrawal. It also emphasizes the possible mechanisms of neuroprotective actions of CEF with respective neurological maladies.
Collapse
Affiliation(s)
- Ebrahim M Yimer
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Hailemichael Zeru Hishe
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Kald Beshir Tuem
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
18
|
Crowley EK, Nolan YM, Sullivan AM. Exercise as a therapeutic intervention for motor and non-motor symptoms in Parkinson's disease: Evidence from rodent models. Prog Neurobiol 2018; 172:2-22. [PMID: 30481560 DOI: 10.1016/j.pneurobio.2018.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/25/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is characterised by degeneration of dopaminergic neurons of the nigrostriatal pathway, which leads to the cardinal motor symptoms of the disease - tremor, rigidity and postural instability. A number of non-motor symptoms are also associated with PD, including cognitive impairment, mood disturbances and dysfunction of gastrointestinal and autonomic systems. Current therapies provide symptomatic relief but do not halt the disease process, so there is an urgent need for preventative strategies. Lifestyle interventions such as aerobic exercise have shown potential to lower the risk of developing PD and to alleviate both motor and non-motor symptoms. However, there is a lack of large-scale randomised clinical trials that have employed exercise in PD patients. This review will focus on the evidence from studies on rodent models of PD, for employing exercise as an intervention for both motor and non-motor symptoms.
Collapse
Affiliation(s)
- E K Crowley
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Y M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - A M Sullivan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
19
|
Tikhonova MA, Amstislavskaya TG, Belichenko VM, Fedoseeva LA, Kovalenko SP, Pisareva EE, Avdeeva AS, Kolosova NG, Belyaev ND, Aftanas LI. Modulation of the expression of genes related to the system of amyloid-beta metabolism in the brain as a novel mechanism of ceftriaxone neuroprotective properties. BMC Neurosci 2018; 19:13. [PMID: 29745864 PMCID: PMC5998892 DOI: 10.1186/s12868-018-0412-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background The dominant hypothesis about the pathogenesis of Alzheimer’s disease (AD) is the “amyloid cascade” concept and modulating the expression of proteins involved in the metabolism of amyloid-beta (Aβ) is proposed as an effective strategy for the prevention and therapy of AD. Recently, we found that an antibiotic ceftriaxone (CEF), which possesses neuroprotective activity, reduced cognitive deficits and neurodegenerative changes in OXYS rats, a model of sporadic AD. The molecular mechanisms of this effect are not completely clear, we suggested that the drug might serve as the regulator of the expression of the genes involved in the metabolism of Aβ and the pathogenesis of AD. The study was aimed to determine the effects of CEF on mRNA levels of Bace1 (encoding β-secretase BACE1 involved in Aβ production), Mme, Ide, Ece1, Ace2 (encoding enzymes involved in Aβ degradation), Epo (encoding erythropoietin related to endothelial function and clearance of Aβ across the blood brain barrier) in the frontal cortex, hippocampus, striatum, hypothalamus, and amygdala of OXYS and Wistar (control strain) male rats. Starting from the age of 14 weeks, animals received CEF (100 mg/kg/day, i.p., 36 days) or saline. mRNA levels were evaluated with RT-qPCR method. Biochemical parameters of plasma were measured for control of system effects of the treatment. Results To better understand strain variations studied here, we compared the gene expression between untreated OXYS and Wistar rats. This comparison showed a significant decrease in mRNA levels of Ace2 in the frontal cortex and hypothalamus, and of Actb in the amygdala of untreated OXYS rats. Analysis of potential effects of CEF revealed its novel targets. In the compound-treated OXYS cohort, CEF diminished mRNA levels of Bace1 and Ace2 in the hypothalamus, and Aktb in the frontal cortex. Furthermore, CEF augmented Mme, Ide, and Epo mRNA levels in the amygdala as well as the levels of Ece1 and Aktb in the striatum. Finally, CEF also attenuated the activity of ALT and AST in plasma of OXYS rats. Conclusion Those findings disclosed novel targets for CEF action that might be involved into neuroprotective mechanisms at early, pre-plaque stages of AD-like pathology development.
Collapse
Affiliation(s)
- Maria A Tikhonova
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia. .,Novosibirsk State University, Novosibirsk, 630090, Russia.
| | - Tamara G Amstislavskaya
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia.,Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Victor M Belichenko
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia
| | - Larisa A Fedoseeva
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia.,Federal Research Center "Institute of Cytology and Genetics", Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Sergey P Kovalenko
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia
| | - Ekaterina E Pisareva
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia
| | - Alla S Avdeeva
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia
| | - Nataliya G Kolosova
- Federal Research Center "Institute of Cytology and Genetics", Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | | | - Lyubomir I Aftanas
- Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine" (SRIPhBM), Timakov St., 4, Novosibirsk, 630117, Russia.,Novosibirsk State University, Novosibirsk, 630090, Russia
| |
Collapse
|
20
|
Neuroprotective effects of ceftriaxone treatment on cognitive and neuronal deficits in a rat model of accelerated senescence. Behav Brain Res 2017; 330:8-16. [DOI: 10.1016/j.bbr.2017.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/26/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022]
|
21
|
Hsieh MH, Meng WY, Liao WC, Weng JC, Li HH, Su HL, Lin CL, Hung CS, Ho YJ. Ceftriaxone reverses deficits of behavior and neurogenesis in an MPTP-induced rat model of Parkinson's disease dementia. Brain Res Bull 2017; 132:129-138. [PMID: 28576659 DOI: 10.1016/j.brainresbull.2017.05.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/22/2017] [Accepted: 05/23/2017] [Indexed: 12/29/2022]
Abstract
Hyperactivity of the glutamatergic system is involved in excitotoxicity and neurodegeneration in Parkinson's disease (PD) so that glutamatergic modulation maybe a potential therapeutic target for PD. Ceftriaxone (CEF) has been reported to increase glutamate uptake by increasing glutamate transporter expression and has been demonstrated neuroprotective effects in animal study. The aim of this study was to determine the effects of CEF on behavior and neurogenesis in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD rat model. MPTP was stereotaxically injected into the substantia nigra pars compacta (SNc) of male Wistar rats. Starting on the same day after MPTP lesioning (day 0), the rats were injected daily with either CEF or saline for 14days and underwent a T-maze test on days 8-10 and an object recognition test on days 12-14, then the brain was taken for histological evaluation on day 15. The results showed that MPTP lesioning resulted in decreased motor function, working memory, and object recognition and reduced neurogenesis in the substantial nigra and dentate gyrus of the hippocampus. These behavioral and neuronal changes were prevented by CEF treatment. To our knowledge, this is the first study showing that CEF prevents loss of neurogenesis in the brain of PD rats. CEF may therefore have clinical potential in the treatment of PD.
Collapse
Affiliation(s)
- Ming-Hong Hsieh
- Department of Psychiatry, Chung Shan Medical University Hospital, Department of Psychiatry, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Wan-Yun Meng
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Wen-Chieh Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan, ROC
| | - Jun-Cheng Weng
- Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Hsin-Hua Li
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Hong-Lin Su
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung-Hsing University, Taichung 402, Taiwan, ROC
| | - Chih-Li Lin
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, ROC.
| | - Ching-Sui Hung
- Occupational Safety and Health Office, Taipei City Hospital, Taipei 10341, Taiwan, ROC.
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan, ROC.
| |
Collapse
|
22
|
Clark IA, Vissel B. Excess cerebral TNF causing glutamate excitotoxicity rationalizes treatment of neurodegenerative diseases and neurogenic pain by anti-TNF agents. J Neuroinflammation 2016; 13:236. [PMID: 27596607 PMCID: PMC5011997 DOI: 10.1186/s12974-016-0708-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/30/2016] [Indexed: 02/06/2023] Open
Abstract
The basic mechanism of the major neurodegenerative diseases, including neurogenic pain, needs to be agreed upon before rational treatments can be determined, but this knowledge is still in a state of flux. Most have agreed for decades that these disease states, both infectious and non-infectious, share arguments incriminating excitotoxicity induced by excessive extracellular cerebral glutamate. Excess cerebral levels of tumor necrosis factor (TNF) are also documented in the same group of disease states. However, no agreement exists on overarching mechanism for the harmful effects of excess TNF, nor, indeed how extracellular cerebral glutamate reaches toxic levels in these conditions. Here, we link the two, collecting and arguing the evidence that, across the range of neurodegenerative diseases, excessive TNF harms the central nervous system largely through causing extracellular glutamate to accumulate to levels high enough to inhibit synaptic activity or kill neurons and therefore their associated synapses as well. TNF can be predicted from the broader literature to cause this glutamate accumulation not only by increasing glutamate production by enhancing glutaminase, but in addition simultaneously reducing glutamate clearance by inhibiting re-uptake proteins. We also discuss the effects of a TNF receptor biological fusion protein (etanercept) and the indirect anti-TNF agents dithio-thalidomides, nilotinab, and cannabinoids on these neurological conditions. The therapeutic effects of 6-diazo-5-oxo-norleucine, ceptriaxone, and riluzole, agents unrelated to TNF but which either inhibit glutaminase or enhance re-uptake proteins, but do not do both, as would anti-TNF agents, are also discussed in this context. By pointing to excess extracellular glutamate as the target, these arguments greatly strengthen the case, put now for many years, to test appropriately delivered ant-TNF agents to treat neurodegenerative diseases in randomly controlled trials.
Collapse
Affiliation(s)
- Ian A Clark
- Biomedical Sciences and Biochemistry, Research School of Biology, Australian National University, Acton, Canberra, Australian Capital Territory, 0200, Australia.
| | - Bryce Vissel
- Neurodegeneration Research Group, Garvan Institute, 384 Victoria Street, Sydney, New South Wales, 2010, Australia
| |
Collapse
|
23
|
Weng JC, Tikhonova MA, Chen JH, Shen MS, Meng WY, Chang YT, Chen KH, Liang KC, Hung CS, Amstislavskaya TG, Ho YJ. Ceftriaxone prevents the neurodegeneration and decreased neurogenesis seen in a Parkinson's disease rat model: An immunohistochemical and MRI study. Behav Brain Res 2016; 305:126-39. [PMID: 26940602 DOI: 10.1016/j.bbr.2016.02.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 02/19/2016] [Accepted: 02/26/2016] [Indexed: 02/06/2023]
Abstract
Manganese-enhanced magnetic resonance imaging (MEMRI) is a widely used technique for detecting neuronal activity in the brain of a living animal. Ceftriaxone (CEF) has been shown to have neuroprotective effects in neurodegenerative diseases. The present study was aimed at clarifying whether, in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease (PD) rat model, the known CEF-induced neuronal protection was accompanied by neurogenesis and decreased loss of neuronal activity. After MPTP lesioning (day 0), the rats were treated with CEF (100mg/kg/day, i.p.) or saline for 15 days. They were then injected with MnCl2 (40mg/kg, i.p.) on day 13 and underwent a brain MRI scan on day 14, then the brain was taken for histological evaluation on day 15. The results showed that MPTP lesioning resulted in decreased neuronal activity and density in the nigrostriatal dopaminergic (DAergic) system and the hippocampal CA1, CA3, and dentate gyrus (DG) areas and reduced neurogenesis in the DG, but in hyperactivity in the subthalamic nucleus (STN). These neuronal changes were prevented by CEF treatment. Positive correlations between MEMRI R1 values and neuronal density in the hippocampus were evidenced. Neuronal densities in the hippocampus and SNc were positively correlated. In addition, the R1 value of the STN showed a positive correlation with its neuronal activity but showed a negative correlation with the density of DAergic neurons in the SNc. Therefore, MEMRI R1 value may serve as a good indicator for PD severity and the effect of treatment. To our knowledge, this is the first study showing that CEF prevents loss of neuronal activity and neurogenesis in the brain of PD rats. CEF may therefore have clinical potential in the treatment of PD.
Collapse
Affiliation(s)
- Jun-Cheng Weng
- Department of Medical Imaging and Radiological Sciences, Department of Medical Imaging, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Maria A Tikhonova
- Laboratory of Experimental Models of Neurodegenerative Processes, Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine", Novosibirsk 630117, Russia
| | - Jian-Horng Chen
- School of Physical Therapy, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Mei-Shiuan Shen
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Wan-Yun Meng
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Yen-Ting Chang
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan, ROC
| | - Ke-Hsin Chen
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan, ROC
| | - Keng-Chen Liang
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan, ROC; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei 10617, Taiwan, ROC
| | - Ching-Sui Hung
- Occupational Safety and Health Office, Taipei City Hospital, Taipei 10341, Taiwan, ROC.
| | - Tamara G Amstislavskaya
- Laboratory of Experimental Models of Emotional Pathology, Federal State Budgetary Scientific Institution "Scientific Research Institute of Physiology and Basic Medicine", Novosibirsk 630117, Russia.
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan, ROC.
| |
Collapse
|