1
|
Fan Y, Yang W, Wu W, Wang X, Lin Y, Wu L, Wang J, Huan F, Ding H, Gao R. Serum neurotransmitter analysis of motor and non-motor symptoms in Parkinson's patients. Front Aging Neurosci 2024; 16:1423120. [PMID: 39654806 PMCID: PMC11625801 DOI: 10.3389/fnagi.2024.1423120] [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: 04/25/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024] Open
Abstract
Clinical symptoms of Parkinson's disease (PD) are classified into motor and non-motor symptoms. Mental disorders, especially depression, are one of the major non-motor manifestations of PD. However, the underlying mechanisms remain poorly understood. In the present study, 21 neurotransmitters associated with mental disorders were measured in serum samples from patients and controls using the ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) assay. Additionally, five clinical scales-the MDS Unified Parkinson's Disease Rating Scale (UPDRS), the Non-Motor Symptoms Scale (NMSS), the Mini-Mental State Examination (MMSE), the Hamilton Anxiety Scale (HAMA), and the Hamilton Depression Scale (HAMD)-were used to evaluate the severity of both motor and non-motor symptoms in PD patients. Analysis of neurotransmitter metabolism revealed significant changes in the tryptophan (Trp) metabolic pathway in PD patients. Specifically, levels of Trp, kynurenine (KYN), kynurenic acid (KA), nicotinamide (NAM), and 5-methoxyltryptamine (MeOTA) were substantially decreased. Additionally, three other excitation/inhibiting amino acids-glutamic acid (Glu), 4-aminobutyric acid (GABA), and aspartic acid (Asp)-also declined. Moreover, neurotransmitter conversion ratios, such as KA/KYN, nicotinamide/niacin (NAM/NA), 5-hydroxytryptophan/tryptophan (5-HTP/Trp), and quinolinic acid/kynurenic acid (QA/KA), provided more dynamic insights into disrupted neurotransmitter metabolism. Correlation analyses between scale scores and neurotransmitter levels showed that concentrations of xanthurenic acid (XA) and the turnover rate of 3-hydroxykynurenine (3-HK) were negatively correlated with UPDRS scores, while 5-hydroxytryptamine (5-HT) and GABA levels were negatively correlated with non-motor symptoms in PD patients. In summary, this study elucidates, for the first time, the potential association and dynamics between altered neurotransmitter metabolism and the etiology of PD in terms of motor and non-motor functions. These findings offer novel biomarkers and therapeutic targets for the treatment of PD.
Collapse
Affiliation(s)
- Yichun Fan
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Hygienic Analysis and Detection, the Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenping Yang
- The First Affiliated Hospital of Nanjing Medical University, Jiangsu Provincial People’s Hospital, Nanjing, China
| | - Weilan Wu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Hygienic Analysis and Detection, the Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinjing Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yuxin Lin
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Hygienic Analysis and Detection, the Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Linlin Wu
- Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Jun Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Fei Huan
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Haixia Ding
- The First Affiliated Hospital of Nanjing Medical University, Jiangsu Provincial People’s Hospital, Nanjing, China
| | - Rong Gao
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Hygienic Analysis and Detection, the Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Luo H, de Velasco EMF, Gansemer B, Frederick M, Aguado C, Luján R, Thayer SA, Wickman K. Amyloid-β oligomers trigger sex-dependent inhibition of GIRK channel activity in hippocampal neurons in mice. Sci Signal 2024; 17:eado4132. [PMID: 39353038 PMCID: PMC11600338 DOI: 10.1126/scisignal.ado4132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by amyloid plaques and cognitive decline, the latter of which is thought to be driven by soluble oligomeric amyloid-β (oAβ). The dysregulation of G protein-gated inwardly rectifying K+ (GIRK; also known as Kir3) channels has been implicated in rodent models of AD. Here, seeking mechanistic insights, we uncovered a sex-dependent facet of GIRK-dependent signaling in AD-related amyloid pathophysiology. Synthetic oAβ1-42 suppressed GIRK-dependent signaling in hippocampal neurons from male mice, but not from female mice. This effect required cellular prion protein, the receptor mGluR5, and production of arachidonic acid by the phospholipase PLA2. Although oAβ suppressed GIRK channel activity only in male hippocampal neurons, intrahippocampal infusion of oAβ or genetic suppression of GIRK channel activity in hippocampal pyramidal neurons impaired performance on a memory test in both male and female mice. Moreover, genetic enhancement of GIRK channel activity in hippocampal pyramidal neurons blocked oAβ-induced cognitive impairment in both male and female mice. In APP/PS1 AD model mice, GIRK-dependent signaling was diminished in hippocampal CA1 pyramidal neurons from only male mice before cognitive deficit was detected. However, enhancing GIRK channel activity rescued cognitive deficits in older APP/PS1 mice of both sexes. Thus, whereas diminished GIRK channel activity contributes to cognitive deficits in male mice with increased oAβ burden, enhancing its activity may have therapeutic potential for both sexes.
Collapse
Affiliation(s)
- Haichang Luo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Benjamin Gansemer
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - McKinzie Frederick
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carolina Aguado
- Synaptic Structure Laboratory, Departmento de Ciencias Médicas, Instituto de Biomedicina, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete 02006, SPAIN
| | - Rafael Luján
- Synaptic Structure Laboratory, Departmento de Ciencias Médicas, Instituto de Biomedicina, Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, Albacete 02006, SPAIN
| | - Stanley A. Thayer
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
3
|
Han M, Jung S, Lee D. Drug repurposing for Parkinson's disease by biological pathway based edge-weighted network proximity analysis. Sci Rep 2024; 14:21258. [PMID: 39261542 PMCID: PMC11390918 DOI: 10.1038/s41598-024-71922-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
Parkinson's disease is the second most frequent neurodegenerative disease, and its severity is increasing with extended life expectancy. Most of current treatments provide symptomatic relief; however, disease progression is not inhibited. There are multiple trials for treatments that target the causes of the disease but they were flawed. The mechanisms underlying neurodegenerative diseases are intricate, and understanding the interplay among the biological elements involved is crucial. These relationships can be effectively analyzed through biological networks, and the application of network-based analyses in the context of neurodegenerative disease treatment has gained considerable attention. Moreover, considering the significance differences in interactions between biological elements within the network is important. Therefore, we introduce a novel biological pathway based edge-weighted network construction method for drug repurposing in Parkinson's disease. The interaction found in multiple Parkinson's disease-related pathways is more significant than other interactions, and this significance is reflected in the network edge weights. Using the edge-weighted network construction method, we found a significant difference in the efficacy between known and unknown Parkinson's disease drugs. The method predicts drug-disease interactions more accurately than approaches that do not consider the significance differences among interactions, and the paths between the drug and disease within the network correspond to the drug's mechanism of action. In summary, we propose a network-based drug repurposing method using the biological pathway based edge-weighted network. Using this methodology, researchers can find novel drug candidates for the parkinson's disease and their mechanism of actions.
Collapse
Affiliation(s)
- Manyoung Han
- Korea Advanced Institute of Science and Technology, Daejeon, KS015, Korea
| | - Seunghwan Jung
- Korea Advanced Institute of Science and Technology, Daejeon, KS015, Korea
| | - Doheon Lee
- Korea Advanced Institute of Science and Technology, Daejeon, KS015, Korea.
| |
Collapse
|
4
|
Martirosyan A, Ansari R, Pestana F, Hebestreit K, Gasparyan H, Aleksanyan R, Hnatova S, Poovathingal S, Marneffe C, Thal DR, Kottick A, Hanson-Smith VJ, Guelfi S, Plumbly W, Belgard TG, Metzakopian E, Holt MG. Unravelling cell type-specific responses to Parkinson's Disease at single cell resolution. Mol Neurodegener 2024; 19:7. [PMID: 38245794 PMCID: PMC10799528 DOI: 10.1186/s13024-023-00699-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 12/14/2023] [Indexed: 01/22/2024] Open
Abstract
Parkinson's Disease (PD) is the second most common neurodegenerative disorder. The pathological hallmark of PD is loss of dopaminergic neurons and the presence of aggregated α-synuclein, primarily in the substantia nigra pars compacta (SNpc) of the midbrain. However, the molecular mechanisms that underlie the pathology in different cell types is not currently understood. Here, we present a single nucleus transcriptome analysis of human post-mortem SNpc obtained from 15 sporadic Parkinson's Disease (PD) cases and 14 Controls. Our dataset comprises ∼84K nuclei, representing all major cell types of the brain, allowing us to obtain a transcriptome-level characterization of these cell types. Importantly, we identify multiple subpopulations for each cell type and describe specific gene sets that provide insights into the differing roles of these subpopulations. Our findings reveal a significant decrease in neuronal cells in PD samples, accompanied by an increase in glial cells and T cells. Subpopulation analyses demonstrate a significant depletion of tyrosine hydroxylase (TH) enriched astrocyte, microglia and oligodendrocyte populations in PD samples, as well as TH enriched neurons, which are also depleted. Moreover, marker gene analysis of the depleted subpopulations identified 28 overlapping genes, including those associated with dopamine metabolism (e.g., ALDH1A1, SLC6A3 & SLC18A2). Overall, our study provides a valuable resource for understanding the molecular mechanisms involved in dopaminergic neuron degeneration and glial responses in PD, highlighting the existence of novel subpopulations and cell type-specific gene sets.
Collapse
Affiliation(s)
| | - Rizwan Ansari
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0AH, UK
| | | | | | - Hayk Gasparyan
- Armenian Bioinformatics Institute, Yerevan, Armenia
- Department of Mathematics and Mechanics, Yerevan State University, Yerevan, Armenia
| | - Razmik Aleksanyan
- Armenian Bioinformatics Institute, Yerevan, Armenia
- Department of Mathematics and Mechanics, Yerevan State University, Yerevan, Armenia
| | - Silvia Hnatova
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0AH, UK
| | | | | | - Dietmar R Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute, KU Leuven, and Department of Pathology, UZ Leuven, Leuven, Belgium
| | | | | | | | - William Plumbly
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0AH, UK
| | | | - Emmanouil Metzakopian
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0AH, UK.
- bit.bio, The Dorothy Hodgkin Building, Babraham Research Institute, Cambridge, CB22 3FH, UK.
| | - Matthew G Holt
- VIB Center for Brain & Disease Research, KU Leuven, Leuven, Belgium.
- Laboratory of Synapse Biology, i3S, Porto, Portugal.
| |
Collapse
|
5
|
Zhou L, Zhan W, Wei X. Clinical pharmacology and pharmacogenetics of prostaglandin analogues in glaucoma. Front Pharmacol 2022; 13:1015338. [PMID: 36313286 PMCID: PMC9596770 DOI: 10.3389/fphar.2022.1015338] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/29/2022] [Indexed: 07/30/2023] Open
Abstract
Glaucoma is the main cause of irreversible visual loss worldwide, and comprises a group of progressive, age-related, and chronic optic neuropathies. Prostaglandin analogs are considered a first-line treatment in the management of glaucoma and have the best efficacy in reducing intraocular pressure. When comparing these therapeutic agents between them, long-term therapy with 0.03% bimatoprost is the most effective followed by treatment with 0.005% latanoprost and 0.004% travoprost. The prevalence of adverse events is lower for latanoprost than for other prostaglandin analogs. However, some patients do not respond to the treatment with prostaglandin analogs (non-responders). Intraocular pressure-lowering efficacy differs significantly between individuals partly owing to genetic factors. Rs1045642 in ABCB1, rs4241366 in SLCO2A1, rs9503012 in GMDS, rs10306114 in PTGS1, rs11568658 in MRP4, rs10786455 and rs6686438 in PTGFR were reported to be positive with the response to prostaglandin analogs in patients with glaucoma. A negative association was found between single nucleotide polymorphisms of PTGFR (rs11578155 and rs6672484) and the response to prostaglandin analogs in patients with glaucoma. The current review is an analysis of the information relevant to prostaglandin analog treatments based on previous literatures. It describes in detail the clinical pharmacology and pharmacogenetics of drugs belonging to this therapeutical class to provide a sound pharmacological basis for their proper use in ophthalmological clinical practice.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyi Zhan
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Hoch L, Bourg N, Degrugillier F, Bruge C, Benabides M, Pellier E, Tournois J, Mahé G, Maignan N, Dawe J, Georges M, Papazian D, Subramanian N, Simon S, Fanen P, Delevoye C, Richard I, Nissan X. Dual Blockade of Misfolded Alpha-Sarcoglycan Degradation by Bortezomib and Givinostat Combination. Front Pharmacol 2022; 13:856804. [PMID: 35571097 PMCID: PMC9093689 DOI: 10.3389/fphar.2022.856804] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Limb-girdle muscular dystrophy type R3 (LGMD R3) is a rare genetic disorder characterized by a progressive proximal muscle weakness and caused by mutations in the SGCA gene encoding alpha-sarcoglycan (α-SG). Here, we report the results of a mechanistic screening ascertaining the molecular mechanisms involved in the degradation of the most prevalent misfolded R77C-α-SG protein. We performed a combinatorial study to identify drugs potentializing the effect of a low dose of the proteasome inhibitor bortezomib on the R77C-α-SG degradation inhibition. Analysis of the screening associated to artificial intelligence-based predictive ADMET characterization of the hits led to identification of the HDAC inhibitor givinostat as potential therapeutical candidate. Functional characterization revealed that givinostat effect was related to autophagic pathway inhibition, unveiling new theories concerning degradation pathways of misfolded SG proteins. Beyond the identification of a new therapeutic option for LGMD R3 patients, our results shed light on the potential repurposing of givinostat for the treatment of other genetic diseases sharing similar protein degradation defects such as LGMD R5 and cystic fibrosis.
Collapse
Affiliation(s)
- Lucile Hoch
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Nathalie Bourg
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, Evry, France
| | | | - Céline Bruge
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Manon Benabides
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Emilie Pellier
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Johana Tournois
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Gurvan Mahé
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | | | | | | | | | | | | | - Pascale Fanen
- Université Paris Est Creteil, INSERM, IMRB, Créteil, France.,Département de Genetique, DMU Biologie-Pathologie, GH Mondor-A. Chenevier, AP-HP, Creteil, France
| | - Cédric Delevoye
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Isabelle Richard
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, Evry, France
| | - Xavier Nissan
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| |
Collapse
|
7
|
Desvaux E, Aussy A, Hubert S, Keime-Guibert F, Blesius A, Soret P, Guedj M, Pers JO, Laigle L, Moingeon P. Model-based computational precision medicine to develop combination therapies for autoimmune diseases. Expert Rev Clin Immunol 2021; 18:47-56. [PMID: 34842494 DOI: 10.1080/1744666x.2022.2012452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION The complex pathophysiology of autoimmune diseases (AIDs) is being progressively deciphered, providing evidence for a multiplicity of pro-inflammatory pathways underlying heterogeneous clinical phenotypes and disease evolution. AREAS COVERED Treatment strategies involving drug combinations are emerging as a preferred option to achieve remission in a vast majority of patients affected by systemic AIDs. The design of appropriate drug combinations can benefit from AID modeling following a comprehensive multi-omics molecular profiling of patients combined with Artificial Intelligence (AI)-powered computational analyses. Such disease models support patient stratification in homogeneous subgroups, shed light on dysregulated pro-inflammatory pathways and yield hypotheses regarding potential therapeutic targets and candidate biomarkers to stratify and monitor patients during treatment. AID models inform the rational design of combination therapies interfering with independent pro-inflammatory pathways related to either one of five prominent immune compartments contributing to the pathophysiology of AIDs, i.e. pro-inflammatory signals originating from tissues, innate immune mechanisms, T lymphocyte activation, autoantibodies and B cell activation, as well as soluble mediators involved in immune cross-talk. EXPERT OPINION The optimal management of AIDs in the future will rely upon rationally designed combination therapies, as a modality of a model-based Computational Precision Medicine taking into account the patients' biological and clinical specificities.
Collapse
Affiliation(s)
- Emiko Desvaux
- Servier, Research and Development, Suresnes Cedex, France.,U1227 -Laboratoire d'Immunologie, Univ Brest, CHRU Morvan, Brest Cedex, France
| | - Audrey Aussy
- Servier, Research and Development, Suresnes Cedex, France
| | - Sandra Hubert
- Servier, Research and Development, Suresnes Cedex, France
| | | | - Alexia Blesius
- Servier, Research and Development, Suresnes Cedex, France
| | - Perrine Soret
- Servier, Research and Development, Suresnes Cedex, France
| | - Mickaël Guedj
- Servier, Research and Development, Suresnes Cedex, France
| | | | | | | |
Collapse
|
8
|
Hippocampal and Reticulo-Thalamic Parvalbumin Interneurons and Synaptic Re-Organization during Sleep Disorders in the Rat Models of Parkinson's Disease Neuropathology. Int J Mol Sci 2021; 22:ijms22168922. [PMID: 34445628 PMCID: PMC8396216 DOI: 10.3390/ijms22168922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022] Open
Abstract
We investigated the alterations of hippocampal and reticulo-thalamic (RT) GABAergic parvalbumin (PV) interneurons and their synaptic re-organizations underlying the prodromal local sleep disorders in the distinct rat models of Parkinson’s disease (PD). We demonstrated for the first time that REM sleep is a predisposing state for the high-voltage sleep spindles (HVS) induction in all experimental models of PD, particularly during hippocampal REM sleep in the hemiparkinsonian models. There were the opposite underlying alterations of the hippocampal and RT GABAergic PV+ interneurons along with the distinct MAP2 and PSD-95 expressions. Whereas the PD cholinopathy enhanced the number of PV+ interneurons and suppressed the MAP2/PSD-95 expression, the hemiparkinsonism with PD cholinopathy reduced the number of PV+ interneurons and enhanced the MAP2/PSD-95 expression in the hippocampus. Whereas the PD cholinopathy did not alter PV+ interneurons but partially enhanced MAP2 and suppressed PSD-95 expression remotely in the RT, the hemiparkinsonism with PD cholinopathy reduced the PV+ interneurons, enhanced MAP2, and did not change PSD-95 expression remotely in the RT. Our study demonstrates for the first time an important regulatory role of the hippocampal and RT GABAergic PV+ interneurons and the synaptic protein dynamic alterations in the distinct rat models of PD neuropathology.
Collapse
|
9
|
Vetel S, Foucault-Fruchard L, Tronel C, Buron F, Vergote J, Bodard S, Routier S, Sérrière S, Chalon S. Neuroprotective and anti-inflammatory effects of a therapy combining agonists of nicotinic α7 and σ1 receptors in a rat model of Parkinson's disease. Neural Regen Res 2021; 16:1099-1104. [PMID: 33269756 PMCID: PMC8224116 DOI: 10.4103/1673-5374.300451] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
To date there is no treatment able to stop or slow down the loss of dopaminergic neurons that characterizes Parkinson’s disease. It was recently observed in a rodent model of Alzheimer’s disease that the interaction between the α7 subtype of nicotinic acetylcholine receptor (α7-nAChR) and sigma-1 receptor (σ1-R) could exert neuroprotective effects through the modulation of neuroinflammation which is one of the key components of the pathophysiology of Parkinson’s disease. In this context, the aim of the present study was to assess the effects of the concomitant administration of N-(3R)-1-azabicyclo[2.2.2]oct-3-yl-furo[2,3-c]pyridine-5-carboxamide (PHA) 543613 as an α7-nAChR agonist and 2-(4-morpholinethyl) 1-phenylcyclohexanecarboxylate (PRE)-084 as a σ1-R agonist in a well-characterized 6-hydroxydopamine rat model of Parkinson’s disease. The animals received either vehicle separately or the dual therapy PHA/PRE once a day until day 14 post-lesion. Although no effect was noticed in the amphetamine-induced rotation test, our data has shown that the PHA/PRE treatment induced partial protection of the dopaminergic neurons (15–20%), assessed by the dopamine transporter density in the striatum and immunoreactive tyrosine hydroxylase in the substantia nigra. Furthermore, this dual therapy reduced the degree of glial activation consecutive to the 6-hydroxydopamine lesion, i.e, the 18 kDa translocation protein density and glial fibrillary acidic protein staining in the striatum, and the CD11b and glial fibrillary acidic protein staining in the substantia nigra. Hence, this study reports for the first time that concomitant activation of α7-nAChR and σ1-R can provide a partial recovery of the nigro-striatal dopaminergic neurons through the modulation of microglial activation. The study was approved by the Regional Ethics Committee (CEEA Val de Loire n°19) validated this protocol (Authorization N°00434.02) on May 15, 2014.
Collapse
Affiliation(s)
- Steven Vetel
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | - Claire Tronel
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Frédéric Buron
- Institut de Chimie Organique et Analytique, ICOA, UMR CNRS 7311, Université d'Orléans, Orléans, France
| | - Jackie Vergote
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvie Bodard
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvain Routier
- Institut de Chimie Organique et Analytique, ICOA, UMR CNRS 7311, Université d'Orléans, Orléans, France
| | - Sophie Sérrière
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| |
Collapse
|
10
|
Boussicault L, Laffaire J, Schmitt P, Rinaudo P, Callizot N, Nabirotchkin S, Hajj R, Cohen D. Combination of acamprosate and baclofen (PXT864) as a potential new therapy for amyotrophic lateral sclerosis. J Neurosci Res 2020; 98:2435-2450. [PMID: 32815196 PMCID: PMC7693228 DOI: 10.1002/jnr.24714] [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: 02/12/2020] [Revised: 07/16/2020] [Accepted: 07/25/2020] [Indexed: 12/11/2022]
Abstract
There is currently no therapy impacting the course of amyotrophic lateral sclerosis (ALS). The only approved treatments are riluzole and edaravone, but their efficacy is modest and short‐lasting, highlighting the need for innovative therapies. We previously demonstrated the ability of PXT864, a combination of low doses of acamprosate and baclofen, to synergistically restore cellular and behavioral activity in Alzheimer's and Parkinson's disease models. The overlapping genetic, molecular, and cellular characteristics of these neurodegenerative diseases supported investigating the effectiveness of PXT864 in ALS. As neuromuscular junction (NMJ) alterations is a key feature of ALS, the effects of PXT864 in primary neuron‐muscle cocultures injured by glutamate were studied. PXT864 significantly and synergistically preserved NMJ and motoneuron integrity following glutamate excitotoxicity. PXT864 added to riluzole significantly improved such protection. PXT864 activity was then assessed in primary cultures of motoneurons derived from SOD1G93A rat embryos. These motoneurons presented severe maturation defects that were significantly improved by PXT864. In this model, glutamate application induced an accumulation of TDP‐43 protein in the cytoplasm, a hallmark that was completely prevented by PXT864. The anti‐TDP‐43 aggregation effect was also confirmed in a cell line expressing TDP‐43 fused to GFP. These results demonstrate the value of PXT864 as a promising therapeutic strategy for the treatment of ALS.
Collapse
|
11
|
Prukop T, Wernick S, Boussicault L, Ewers D, Jäger K, Adam J, Winter L, Quintes S, Linhoff L, Barrantes-Freer A, Bartl M, Czesnik D, Zschüntzsch J, Schmidt J, Primas G, Laffaire J, Rinaudo P, Brureau A, Nabirotchkin S, Schwab MH, Nave KA, Hajj R, Cohen D, Sereda MW. Synergistic PXT3003 therapy uncouples neuromuscular function from dysmyelination in male Charcot-Marie-Tooth disease type 1A (CMT1A) rats. J Neurosci Res 2020; 98:1933-1952. [PMID: 32588471 DOI: 10.1002/jnr.24679] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/13/2020] [Accepted: 05/31/2020] [Indexed: 12/11/2022]
Abstract
Charcot-Marie-Tooth disease 1 A (CMT1A) is caused by an intrachromosomal duplication of the gene encoding for PMP22 leading to peripheral nerve dysmyelination, axonal loss, and progressive muscle weakness. No therapy is available. PXT3003 is a low-dose combination of baclofen, naltrexone, and sorbitol which has been shown to improve disease symptoms in Pmp22 transgenic rats, a bona fide model of CMT1A disease. However, the superiority of PXT3003 over its single components or dual combinations have not been tested. Here, we show that in a dorsal root ganglion (DRG) co-culture system derived from transgenic rats, PXT3003 induced myelination when compared to its single and dual components. Applying a clinically relevant ("translational") study design in adult male CMT1A rats for 3 months, PXT3003, but not its dual components, resulted in improved performance in behavioral motor and sensory endpoints when compared to placebo. Unexpectedly, we observed only a marginally increased number of myelinated axons in nerves from PXT3003-treated CMT1A rats. However, in electrophysiology, motor latencies correlated with increased grip strength indicating a possible effect of PXT3003 on neuromuscular junctions (NMJs) and muscle fiber pathology. Indeed, PXT3003-treated CMT1A rats displayed an increased perimeter of individual NMJs and a larger number of functional NMJs. Moreover, muscles of PXT3003 CMT1A rats displayed less neurogenic atrophy and a shift toward fast contracting muscle fibers. We suggest that ameliorated motor function in PXT3003-treated CMT1A rats result from restored NMJ function and muscle innervation, independent from myelination.
Collapse
Affiliation(s)
- Thomas Prukop
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Stephanie Wernick
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - David Ewers
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Karoline Jäger
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Julia Adam
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Lorenz Winter
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Susanne Quintes
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Lisa Linhoff
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Michael Bartl
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Dirk Czesnik
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Schmidt
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | | - Markus H Schwab
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | | | | | - Michael W Sereda
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Saxena A, Dhyani V, Suman G, Giri L. Effect of topology and time window on probability distribution underlying baclofen induced Ca 2+ response in hippocampal neurons .. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:2997-3000. [PMID: 31946519 DOI: 10.1109/embc.2019.8857601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cytosolic Ca2+ oscillation in neurons regulate a wide range of cellular functions starting from cell division to apoptosis. The major challenge in analyzing the large-scale heterogeneous calcium data obtained from hippocampal neurons is that there is no specific tool available for probability density function (pdf) fitting and model ranking. First, we focus on the ranking of various pdf and selection of a particular pdf using maximum log-likelihood. Five pdfs were analyzed in this study, exponential, gamma, log-normal, Rayleigh, and Weibull. Next, we used the statistical models to find the effect of two factors, the network topology and time window for the calcium response. The robustness of the best pdf was validated using multiple datasets obtained through random sampling of neurons from a neuron pool. GPCR targeting drug, baclofen was chosen as the model drug to inhibit Ca2+ response. Strongly-connected neurons show a significant change in Ca2+ oscillations after the addition of drug in comparison to weakly-connected neurons. The proposed technique can be used to study the dose-response from a large number of calcium imaging videos having heterogeneous responses.
Collapse
|
13
|
Nabirotchkin S, Peluffo AE, Rinaudo P, Yu J, Hajj R, Cohen D. Next-generation drug repurposing using human genetics and network biology. Curr Opin Pharmacol 2020; 51:78-92. [PMID: 31982325 DOI: 10.1016/j.coph.2019.12.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022]
Abstract
Drug repurposing has attracted increased attention, especially in the context of drug discovery rates that remain too low despite a recent wave of approvals for biological therapeutics (e.g. gene therapy). These new biological entities-based treatments have high costs that are difficult to justify for small markets that include rare diseases. Drug repurposing, involving the identification of single or combinations of existing drugs based on human genetics data and network biology approaches represents a next-generation approach that has the potential to increase the speed of drug discovery at a lower cost. This Pharmacological Perspective reviews progress and perspectives in combining human genetics, especially genome-wide association studies, with network biology to drive drug repurposing for rare and common diseases with monogenic or polygenic etiologies. Also, highlighted here are important features of this next generation approach to drug repurposing, which can be combined with machine learning methods to meet the challenges of personalized medicine.
Collapse
Affiliation(s)
- Serguei Nabirotchkin
- Network Biology & Drug Discovery Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Alex E Peluffo
- Data Science Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France.
| | - Philippe Rinaudo
- Data Science Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Jinchao Yu
- Data Science Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Rodolphe Hajj
- Preclinical Research and Pharmacology Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Daniel Cohen
- Chief Executive Officer, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| |
Collapse
|
14
|
Tkatchenko TV, Tkatchenko AV. Pharmacogenomic Approach to Antimyopia Drug Development: Pathways Lead the Way. Trends Pharmacol Sci 2019; 40:833-852. [PMID: 31676152 DOI: 10.1016/j.tips.2019.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 12/29/2022]
Abstract
Myopia is the most common eye disorder in the world which is caused by a mismatch between the optical power of the eye and its excessively long axial length. Recent studies revealed that the regulation of the axial length of the eye occurs via a complex signaling cascade, which originates in the retina and propagates across all ocular tissues to the sclera. The complexity of this regulatory cascade has made it particularly difficult to develop effective antimyopia drugs. The current pharmacological treatment options for myopia are limited to atropine and 7-methylxanthine, which have either significant adverse effects or low efficacy. In this review, we focus on the recent advances in genome-wide studies of the signaling pathways underlying myopia development and discuss the potential of systems genetics and pharmacogenomic approaches for the development of antimyopia drugs.
Collapse
Affiliation(s)
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
15
|
Murueta-Goyena A, Andikoetxea A, Gómez-Esteban JC, Gabilondo I. Contribution of the GABAergic System to Non-Motor Manifestations in Premotor and Early Stages of Parkinson's Disease. Front Pharmacol 2019; 10:1294. [PMID: 31736763 PMCID: PMC6831739 DOI: 10.3389/fphar.2019.01294] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Non-motor symptoms are common in Parkinson’s disease (PD) and they represent a major source of disease burden. Several non-motor manifestations, such as rapid eye movement sleep behavior disorder, olfactory loss, gastrointestinal abnormalities, visual alterations, cognitive and mood disorders, are known to precede the onset of motor signs. Nonetheless, the mechanisms mediating these alterations are poorly understood and probably involve several neurotransmitter systems. The dysregulation of GABAergic system has received little attention in PD, although the spectrum of non-motor symptoms might be linked to this pathway. This Mini Review aims to provide up-to-date information about the involvement of the GABAergic system for explaining non-motor manifestations in early stages of PD. Therefore, special attention is paid to the clinical data derived from patients with isolated REM sleep behavior disorder or drug-naïve patients with PD, as they represent prodromal and early stages of the disease, respectively. This, in combination with animal studies, might help us to understand how the disturbance of the GABAergic system is related to non-motor manifestations of PD.
Collapse
Affiliation(s)
- Ane Murueta-Goyena
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Ane Andikoetxea
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Juan Carlos Gómez-Esteban
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Iñigo Gabilondo
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,IKERBASQUE Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
16
|
Campolo M, Paterniti I, Siracusa R, Filippone A, Esposito E, Cuzzocrea S. TLR4 absence reduces neuroinflammation and inflammasome activation in Parkinson's diseases in vivo model. Brain Behav Immun 2019; 76:236-247. [PMID: 30550933 DOI: 10.1016/j.bbi.2018.12.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a progressive, disabling neurodegenerative disorder. It has been shown Toll like receptor (TLR) 4-deficient mice protect against MPTP toxicity, suggesting that dopaminergic cell death is TLR4-dependent. The aim of this study was to demonstrate, in an in vivo model of PD, how TLR4 plays its important role in the pathogenesis of PD by using MPTP neurotoxin model (4 × 20 mg/kg, 2 h apart, i.p). Our experiments have demonstrated that the absence of TLR4 prevented dopamine depletion, increased tyrosine hydroxylase and dopamine transporter activities and reduced the number of α-synuclein-positive neurons. The absence of TLR4 also had an impact on inflammatory processes, modulating the transcription factors NF-κB p65 and AP-1, and reducing astrogliosis. Importantly, we demonstrated that the absence of TLR4 modulated inflammosome pathway. Moreover, it has been shown that TLR4 modulated motor and non-motor symptoms typical of PD. Our results clearly demonstrated that absence of TLR4 reduces the development of neuroinflammation associated with PD through NF-κB, AP-1 and inflammasome pathways modulation; therefore, TLR4 could be considered as an encouraging therapeutic target in neurodegenerative disorders.
Collapse
Affiliation(s)
- Michela Campolo
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy; Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, USA.
| |
Collapse
|
17
|
Swain S, Gupta RK, Ratnayake K, Priyanka PD, Singh R, Jana S, Mitra K, Karunarathne A, Giri L. Confocal Imaging and k-Means Clustering of GABA B and mGluR Mediated Modulation of Ca 2+ Spiking in Hippocampal Neurons. ACS Chem Neurosci 2018; 9:3094-3107. [PMID: 30044088 DOI: 10.1021/acschemneuro.8b00297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Imaging cytosolic calcium in neurons is emerging as a new tool in neurological disease diagnosis, drug screening, and toxicity testing. Ca2+ oscillation signatures show a significant variation depending on GPCR targeting agonists. Quantification of Ca2+ spike trains in ligand induced Ca2+ oscillations remains challenging due to their inherent heterogeneity in primary culture. Moreover, there is no framework available for identification of optimal number of clusters and distance metric to cluster Ca2+ spike trains. Using quantitative confocal imaging and clustering analysis, we show the characterization of Ca2+ spiking in GPCR targeting drug-treated primary culture of hippocampal neurons. A systematic framework for selection of the clustering method instead of an intuition-based method was used to optimize the cluster number and distance metric. The results discern neurons with diverse Ca2+ response patterns, including higher amplitude fast spiking and lower spiking responses, and their relative percentage in a neuron population in absence and presence of GPCR-targeted drugs. The proposed framework was employed to show that the clustering pattern of Ca2+ spiking can be controlled using GABAB and mGluR targeting drugs. This approach can be used for unbiased measurement of neural activity and identification of spiking population with varying amplitude and frequencies, providing a platform for high-content drug screening.
Collapse
Affiliation(s)
- Sarpras Swain
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Rishikesh Kumar Gupta
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Kasun Ratnayake
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Pantula Devi Priyanka
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Ranjana Singh
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Soumya Jana
- Department of Electrical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Kishalay Mitra
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Lopamudra Giri
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| |
Collapse
|
18
|
Marshall LJ, Willett C. Parkinson's disease research: adopting a more human perspective to accelerate advances. Drug Discov Today 2018; 23:1950-1961. [PMID: 30240875 DOI: 10.1016/j.drudis.2018.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) affects 1% of the population over 60 years old and, with global increases in the aging population, presents huge economic and societal burdens. The etiology of PD remains unknown; most cases are idiopathic, presumed to result from genetic and environmental risk factors. Despite 200 years since the first description of PD, the mechanisms behind initiation and progression of the characteristic neurodegenerative processes are not known. Here, we review progress and limitations of the multiple PD animal models available and identify advances that could be implemented to better understand pathological processes, improve disease outcome, and reduce dependence on animal models. Lessons learned from reducing animal use in PD research could serve as guideposts for wider biomedical research.
Collapse
Affiliation(s)
- Lindsay J Marshall
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA
| | - Catherine Willett
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA.
| |
Collapse
|
19
|
Morley KC, Lagopoulos J, Logge W, Chitty K, Baillie A, Haber PS. Neurometabolite Levels in Alcohol Use Disorder Patients During Baclofen Treatment and Prediction of Relapse to Heavy Drinking. Front Psychiatry 2018; 9:412. [PMID: 30233431 PMCID: PMC6131632 DOI: 10.3389/fpsyt.2018.00412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/13/2018] [Indexed: 12/20/2022] Open
Abstract
Background and Aims: Baclofen, a GABAB agonist, is used as a treatment for alcohol dependence. We aimed to examine brain metabolites following administration of baclofen or placebo in alcohol dependent individuals enrolled in a randomized placebo-controlled trial. Methods: Participants included 31 alcohol dependent individuals (recent drinking: N = 16; and abstinent: N = 15) who had received daily baclofen (BAC 30-75 mg = 20) or placebo (PL = 11) for at least 2 weeks (average 17 days). Using in vivo proton magnetic resonance spectroscopy (1H-MRS), spectra from the right parietal lobe were analyzed to obtain measures of GABA, Glutamate (Glu), Glutathione (GSH) and N-Acetyl Apartate (NAA) 120 min following administration of PL or BAC. Results: When weighting alcohol dependent participants according to recent alcohol consumption (within 24 h), there were significant differences between BAC and PL on parietal concentrations of GSH (p < 0.01) and NAA (p < 0.05). Multiple linear regression revealed a significant predictive effect of GSH on heavy drinking days at 12 weeks follow-up (Model: F = 14.28, R2 = 0.85; GSH: B = -1.22, p = 0.01) and also percentage days abstinent at 12 weeks follow-up (Model: F = 6.50, R2 = 0.72; GSH: B = 0.99, p = 0.06). Conclusion: Our data provide preliminary evidence that the effect of baclofen may be mediated by increased parietal concentrations of the antioxidant GSH and NAA in recently drinking alcohol dependent patients. GSH/Cr levels were also predictive of improved drinking outcomes in the trial and suggests a role for neural oxidative stress in alcohol use disorder.
Collapse
Affiliation(s)
- Kirsten C Morley
- NHMRC Centre of Research Excellence in Mental Health and Substance Use, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Jim Lagopoulos
- Sunshine Coast Mind and Neuroscience, University of Sunshine Coast, Birtinya, QLD, Australia
| | - Warren Logge
- NHMRC Centre of Research Excellence in Mental Health and Substance Use, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Kate Chitty
- School of Pharmacology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Andrew Baillie
- NHMRC Centre of Research Excellence in Mental Health and Substance Use, Health Sciences, University of Sydney, Sydney, NSW, Australia
| | - Paul S Haber
- NHMRC Centre of Research Excellence in Mental Health and Substance Use, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Drug Health Services, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
20
|
de Beaurepaire R, Sinclair JMA, Heydtmann M, Addolorato G, Aubin HJ, Beraha EM, Caputo F, Chick JD, de La Selle P, Franchitto N, Garbutt JC, Haber PS, Jaury P, Lingford-Hughes AR, Morley KC, Müller CA, Owens L, Pastor A, Paterson LM, Pélissier F, Rolland B, Stafford A, Thompson A, van den Brink W, Leggio L, Agabio R. The Use of Baclofen as a Treatment for Alcohol Use Disorder: A Clinical Practice Perspective. Front Psychiatry 2018; 9:708. [PMID: 30662411 PMCID: PMC6328471 DOI: 10.3389/fpsyt.2018.00708] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder (AUD) is a brain disorder associated with high rates of mortality and morbidity worldwide. Baclofen, a selective gamma-aminobutyric acid-B (GABA-B) receptor agonist, has emerged as a promising drug for AUD. The use of this drug remains controversial, in part due to uncertainty regarding dosing and efficacy, alongside concerns about safety. To date there have been 15 randomized controlled trials (RCTs) investigating the use of baclofen in AUD; three using doses over 100 mg/day. Two additional RCTs have been completed but have not yet been published. Most trials used fixed dosing of 30-80 mg/day. The other approach involved titration until the desired clinical effect was achieved, or unwanted effects emerged. The maintenance dose varies widely from 30 to more than 300 mg/day. Baclofen may be particularly advantageous in those with liver disease, due to its limited hepatic metabolism and safe profile in this population. Patients should be informed that the use of baclofen for AUD is as an "off-label" prescription, that no optimal fixed dose has been established, and that existing clinical evidence on efficacy is inconsistent. Baclofen therapy requires careful medical monitoring due to safety considerations, particularly at higher doses and in those with comorbid physical and/or psychiatric conditions. Baclofen is mostly used in some European countries and Australia, and in particular, for patients who have not benefitted from the currently used and approved medications for AUD.
Collapse
Affiliation(s)
| | - Julia M A Sinclair
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mathis Heydtmann
- Department of Gastroenterology, Royal Alexandra Hospital Paisley, Paisley, United Kingdom
| | - Giovanni Addolorato
- AUD and Alcohol Related Diseases Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Department of Internal Medicine and Gastroenterology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Henri-Jean Aubin
- Faculté de Médecine, Centre de Recherche en Epidémiologie et Santé des Populations, Université Paris-Sud, Paris, France.,Faculté de Médecine, Université de Versailles Saint-Quentin-en-Yvelines, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Paris, France.,Hôpitaux Universitaires Paris-Sud, Paris, France
| | - Esther M Beraha
- Department of Psychology, University of Amsterdam, Amsterdam, Netherlands
| | - Fabio Caputo
- Department of Internal Medicine, SS. Annunziata Hospital, Cento, Italy
| | - Jonathan D Chick
- Castle Craig Hospital, Blyth Bridge, United Kingdom.,School of Health and Social Care, Edinburgh Napier University, Edinburgh, United Kingdom
| | | | - Nicolas Franchitto
- Department of Addiction Medicine, Poisons and Substance Abuse Treatment Centre, Toulouse-Purpan University Hospital, Toulouse, France
| | - James C Garbutt
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Paul S Haber
- National Health Medical Research Council, Centre of Research Excellence in Mental Health and Substance Use, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Drug Health Services, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Philippe Jaury
- Département de Médecine Générale, Faculté de Médecine, Université Paris Descartes, Paris, France
| | - Anne R Lingford-Hughes
- Neuropsychopharmacology Unit, Division of Brain Sciences, Centre for Psychiatry, Imperial College London, London, United Kingdom
| | - Kirsten C Morley
- Discipline of Addiction Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Christian A Müller
- Department of Psychiatry, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lynn Owens
- Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Adam Pastor
- Department Addiction Medicine, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Louise M Paterson
- Neuropsychopharmacology Unit, Division of Brain Sciences, Centre for Psychiatry, Imperial College London, London, United Kingdom
| | - Fanny Pélissier
- Poison Control Center, Toulouse-Purpan University Hospital, Toulouse, France
| | - Benjamin Rolland
- Service Universitaire d'Addictologie de Lyon, Lyon, France.,University of Lyon, Lyon, France
| | | | - Andrew Thompson
- Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Wim van den Brink
- Department of Psychiatry, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam University, Amsterdam, Netherlands
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, Division of Intramural Clinical and Basic Research, National Institute on Alcohol Abuse and Alcoholism, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Bethesda, MD, United States.,Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States.,Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, United States
| | - Roberta Agabio
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
21
|
Crabbé M, Van der Perren A, Weerasekera A, Himmelreich U, Baekelandt V, Van Laere K, Casteels C. Altered mGluR5 binding potential and glutamine concentration in the 6-OHDA rat model of acute Parkinson's disease and levodopa-induced dyskinesia. Neurobiol Aging 2018; 61:82-92. [DOI: 10.1016/j.neurobiolaging.2017.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 05/24/2017] [Accepted: 09/08/2017] [Indexed: 01/28/2023]
|
22
|
Flavonoids from Pterogyne nitens Inhibit Hepatitis C Virus Entry. Sci Rep 2017; 7:16127. [PMID: 29170411 PMCID: PMC5701011 DOI: 10.1038/s41598-017-16336-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C virus (HCV) is one of the leading causes of liver diseases and transplantation worldwide. The current available therapy for HCV infection is based on interferon-α, ribavirin and the new direct-acting antivirals (DAAs), such as NS3 protease and NS5B polymerase inhibitors. However, the high costs of drug design, severe side effects and HCV resistance presented by the existing treatments demonstrate the need for developing more efficient anti-HCV agents. This study aimed to evaluate the antiviral effects of sorbifolin (1) and pedalitin (2), two flavonoids from Pterogyne nitens on the HCV replication cycle. These compounds were investigated for their anti-HCV activities using genotype 2a JFH-1 subgenomic replicons and infectious virus systems. Flavonoids 1 and 2 inhibited virus entry up to 45.0% and 78.7% respectively at non-cytotoxic concentrations. The mechanism of the flavonoid 2 block to virus entry was demonstrated to be by both the direct action on virus particles and the interference on the host cells. Alternatively, the flavonoid 1 activity was restricted to its virucidal effect. Additionally, no inhibitory effects on HCV replication and release were observed by treating cells with these flavonoids. These data are the first description of 1 and 2 possessing in vitro anti-HCV activity.
Collapse
|
23
|
Tseng CK, Hsu SP, Lin CK, Wu YH, Lee JC, Young KC. Celastrol inhibits hepatitis C virus replication by upregulating heme oxygenase-1 via the JNK MAPK/Nrf2 pathway in human hepatoma cells. Antiviral Res 2017; 146:191-200. [PMID: 28935193 PMCID: PMC7113881 DOI: 10.1016/j.antiviral.2017.09.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/13/2017] [Accepted: 09/18/2017] [Indexed: 12/17/2022]
Abstract
background and purpose Celastrol, a quinone methide triterpene isolated from the root extracts of Tripterygium wilfordii, can greatly induce the gene expression activity of heme oxygenase-1 (HO-1) to achieve disease prevention and control. HO-1 induction was recently shown to result in anti-HCV activity by inducing type I interferon and inhibiting hepatitis C virus (HCV) NS3/4A protease activity. The aim of the present study is to evaluate the anti-HCV activity of celastrol and characterize its mechanism of inhibition. Methods The anti-HCV activity of celastrol was evaluated using the HCV subgenomic replicon and HCVcc infection systems. The anti-HCV mechanism of celastrol targeting HO-1 expression was clarified using specific inhibitors against several signaling pathways. The transcriptional regulation of celastrol on target gene expression was determined using promoter-based reporter activity assay. The synergistic effect of celastrol and a numbers of clinically used anti-HCV drugs was determined via a drug combination assay. Results Celastrol inhibited HCV replication in both the HCV subgenomic and HCVcc infection systems with EC50 values of 0.37 ± 0.022 and 0.43 ± 0.019 μM, respectively. Celastrol-induced heme oxygenase 1 (HO-1) expression promoted antiviral interferon responses and inhibition of NS3/4A protease activity, thereby blocking HCV replication. These antiviral effects were abrogated by treatment with the HO-1-specific inhibitor SnMP or silencing of HO-1 expression by transfection of shRNA, which indicates that HO-1 induction contributes to the anti-HCV activity of celastrol. JNK mitogen-activated protein kinase and nuclear factor erythroid 2-related factor 2 (Nrf2) were confirmed to be involved in the inductive effect of celastrol on HO-1 expression. Celastrol exhibited synergistic effects in combination with interferon-alpha, the NS5A inhibitor daclatasvir, and the NS5B inhibitor sofosbuvir. Conclusion Celastrol can serve as a potential supplement for blocking HCV replication. Targeting the JNK/Nrf2/HO-1 axis presents a promising strategy against HCV infection. Celastrol inhibits HCV replication. Celastrol induces HO-1 production. Celastrol induces interferon-α production and inhibits HCV NS3/4A protease. Celastrol synergistically inhibits HCV replication in combination with IFN-α, sofosbuvir or daclatasvir.
Collapse
Affiliation(s)
- Chin-Kai Tseng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sung-Po Hsu
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Kuang Lin
- Doctoral Degree Program in Marine Biotechnology, College of Marine Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yu-Hsuan Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jin-Ching Lee
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Natural Products and Drug Development, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Kung-Chia Young
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
24
|
Singh S, Jamwal S, Kumar P. Neuroprotective potential of Quercetin in combination with piperine against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity. Neural Regen Res 2017; 12:1137-1144. [PMID: 28852397 PMCID: PMC5558494 DOI: 10.4103/1673-5374.211194] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
1-Methy-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxin that selectively damages dopaminergic neurons in the substantia nigra pars compacta and induces Parkinson's like symptoms in rodents. Quercetin (QC) is a natural polyphenolic bioflavonoid with potent antioxidant and anti-inflammatory properties but lacks of clinical attraction due to low oral bioavailability. Piperine is a well established bioavailability enhancer used pre-clinically to improve the bioavailability of antioxidants (e.g., Quercetin). Therefore, the present study was designed to evaluate the neuroprotective potential of QC together with piperine against MPTP-induced neurotoxicity in rats. MPTP (100 μg/μL/rat, bilaterally) was injected intranigrally on days 1, 4 and 7 using a digital stereotaxic apparatus. QC (25 and 50 mg/kg, intragastrically) and QC (25 mg/kg, intragastrically) in combination with piperine (2.5 mg/kg, intragastrically) were administered daily for 14 days starting from day 8 after the 3rd injection of MPTP. On day 22, animals were sacrificed and the striatum was isolated for oxidative stress parameter (thiobarbituric acid reactive substances, nitrite and glutathione), neuroinflammatory cytokine (interleukin-1β, interleukin-6, and tumor necrosis factor-α) and neurotransmitter (dopamine, norepinephrine, serotonin, gamma-aminobutyric acid, glutamate, 3,4-dihydroxyphenylacetic acid, homovanillic acid, and 5-hydroxyindoleacetic acid) evaluations. Bilateral infusion of MPTP into substantia nigra pars compacta led to significant motor deficits as evidenced by impairments in locomotor activity and rotarod performance in open field test and grip strength and narrow beam walk performance. Both QC (25 and 50 mg/kg) and QC (25 mg/kg) in combination with piperine (2.5 mg/kg), in particular the combination therapy, significantly improved MPTP-induced behavioral abnormalities in rats, reversed the abnormal alterations of neurotransmitters in the striatum, and alleviated oxidative stress and inflammatory response in the striatum. These findings indicate that piperine can enhance the antioxidant and anti-inflammatory properties of QC, and QC in combination with piperine exhibits strong neuroprotective effects against MPTP-induced neurotoxicity.
Collapse
Affiliation(s)
- Shamsher Singh
- Department of Pharmacology, I.S.F College of Pharmacy, Ferozepur Road, Moga, Punjab, India.,I.K. Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Sumit Jamwal
- Department of Pharmacology, I.S.F College of Pharmacy, Ferozepur Road, Moga, Punjab, India.,I.K. Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Puneet Kumar
- Department of Pharmacology, I.S.F College of Pharmacy, Ferozepur Road, Moga, Punjab, India
| |
Collapse
|