1
|
Chandolia P, Rahi V, Kumar P. Neuroprotective effect of silymarin against 3-Nitropropionic acid-induced neurotoxicity in rats. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100130. [PMID: 36568269 PMCID: PMC9780065 DOI: 10.1016/j.crphar.2022.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 12/27/2022] Open
Abstract
(HD) Huntington's disease is a severe hereditary catastrophic neurological disease with an autosomal dominant heritable changes manifested by cognitive, behavioural, and motor progression deficits, resulting in death. Several mechanisms are involved in the pathogenesis of this complex and rare disease, including excitotoxicity, mitochondrial dysfunction, neurotransmitters imbalance, and oxidative stress. Silymarin was selected as an investigational drug, due to its numerous activities in current research, it possesses substantial antioxidant and neuroprotective functionalities. The present research attempts, i.p. injections of 3-NPA (10 mg/kg) were given for 21 days to trigger Huntington-like symptoms in rats. The percentage fluctuations in body weight, the footfall counts, and the time required to transverse the beam and motor functions were analyzed at multiple time points. Oxidative stress markers like MDA/LPO, GSH, protein, nitrite, catalase, and superoxide dismutase levels were examined in the striatum region. The current study results conclusively demonstrate that chronic 3-NPA administration significantly decreased the body weight and showed marked abnormalities in motor coordination, locomotion, and increased striatal generation of free radicals. Furthermore, treatment with silymarin (100 & 200 mg/kg/p.o.), mitigated 3-NPA triggered behavioural and biochemical alterations. Our study results could conclude that Silymarin may be advantageous and might develop an adjuvant treatment for the management of Huntington's disease.
Collapse
Affiliation(s)
- Priyanka Chandolia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
| | - Vikrant Rahi
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India,Corresponding author. Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
2
|
Techera Antunes FT, Caminski ES, Picada JN, Regner AP, Dallegrave E, Hubner de Souza A. In vivo treatment with a subacute low dose of 3-nitropropionic acid does not induce genotoxicity or mutagenicity in rats. Toxicon 2021; 195:20-23. [PMID: 33689791 DOI: 10.1016/j.toxicon.2021.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/02/2021] [Accepted: 03/03/2021] [Indexed: 10/22/2022]
Abstract
3-nitropropionic acid (3-NP) is a toxin that causes neural damage in the striatum and can lead to the development of Huntington's disease manifestations in animal models. Several studies have shown genotoxicity related to the 3-NP treatment. This study investigated potential genotoxicity and mutagenicity that was induced by a low dose (6.25 mg/kg i. p.) 3-NP subacute treatment (daily, over 6 days) in a rat model. The arterial blood and the frontal cortex were analyzed by the comet assay and the bone marrow by micronucleus. Surprisingly, the 3-NP subacute treatment with the low dose did not show genotoxic or mutagenic effects.
Collapse
Affiliation(s)
- Flavia Tasmin Techera Antunes
- Program of Postgraduation in Cellular and Molecular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil.
| | - Emanuelle Sistherenn Caminski
- Department of Pharmacoscience, Federal University Science of Health of Porto Alegre (UFCSPA), Rio Grande do Sul, Brazil.
| | | | - Andrea Pereira Regner
- Program of Postgraduation in Cellular and Molecular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil.
| | - Eliane Dallegrave
- Department of Pharmacoscience, Federal University Science of Health of Porto Alegre (UFCSPA), Rio Grande do Sul, Brazil.
| | - Alessandra Hubner de Souza
- Program of Postgraduation in Cellular and Molecular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil.
| |
Collapse
|
3
|
Musi CA, Agrò G, Santarella F, Iervasi E, Borsello T. JNK3 as Therapeutic Target and Biomarker in Neurodegenerative and Neurodevelopmental Brain Diseases. Cells 2020; 9:cells9102190. [PMID: 32998477 PMCID: PMC7600688 DOI: 10.3390/cells9102190] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 01/01/2023] Open
Abstract
The c-Jun N-terminal kinase 3 (JNK3) is the JNK isoform mainly expressed in the brain. It is the most responsive to many stress stimuli in the central nervous system from ischemia to Aβ oligomers toxicity. JNK3 activity is spatial and temporal organized by its scaffold protein, in particular JIP-1 and β-arrestin-2, which play a crucial role in regulating different cellular functions in different cellular districts. Extensive evidence has highlighted the possibility of exploiting these adaptors to interfere with JNK3 signaling in order to block its action. JNK plays a key role in the first neurodegenerative event, the perturbation of physiological synapse structure and function, known as synaptic dysfunction. Importantly, this is a common mechanism in many different brain pathologies. Synaptic dysfunction and spine loss have been reported to be pharmacologically reversible, opening new therapeutic directions in brain diseases. Being JNK3-detectable at the peripheral level, it could be used as a disease biomarker with the ultimate aim of allowing an early diagnosis of neurodegenerative and neurodevelopment diseases in a still prodromal phase.
Collapse
Affiliation(s)
- Clara Alice Musi
- Department of Pharmacological and Biomolecular Sciences, Milan University, 20133 Milan, Italy;
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
| | - Graziella Agrò
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
| | - Francesco Santarella
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
| | - Erika Iervasi
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
- Department of Experimental Medicine, University of Genoa, Via De Toni 14, 16132 Genoa, Italy
| | - Tiziana Borsello
- Department of Pharmacological and Biomolecular Sciences, Milan University, 20133 Milan, Italy;
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20156 Milan, Italy; (G.A.); (F.S.); (E.I.)
- Correspondence: or ; Tel.: +39-023-901-4469; Fax: +39-023-900-1916
| |
Collapse
|
4
|
Granat L, Hunt RJ, Bateman JM. Mitochondrial retrograde signalling in neurological disease. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190415. [PMID: 32362256 DOI: 10.1098/rstb.2019.0415] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuronal mitochondrial dysfunction causes primary mitochondrial diseases and likely contributes to neurodegenerative diseases including Parkinson's and Alzheimer's disease. Mitochondrial dysfunction has also been documented in neurodevelopmental disorders such as tuberous sclerosis complex and autism spectrum disorder. Only symptomatic treatments exist for neurodevelopmental disorders, while neurodegenerative diseases are largely untreatable. Altered mitochondrial function activates mitochondrial retrograde signalling pathways, which enable signalling to the nucleus to reprogramme nuclear gene expression. In this review, we discuss the role of mitochondrial retrograde signalling in neurological diseases. We summarize how mitochondrial dysfunction contributes to neurodegenerative disease and neurodevelopmental disorders. Mitochondrial signalling mechanisms that have relevance to neurological disease are discussed. We then describe studies documenting retrograde signalling pathways in neurons and glia, and in animal models of neuronal mitochondrial dysfunction and neurological disease. Finally, we suggest how specific retrograde signalling pathways can be targeted to develop novel treatments for neurological diseases. This article is part of the theme issue 'Retrograde signalling from endosymbiotic organelles'.
Collapse
Affiliation(s)
- Lucy Granat
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Rachel J Hunt
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Joseph M Bateman
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
5
|
Early epigenomic and transcriptional changes reveal Elk-1 transcription factor as a therapeutic target in Huntington's disease. Proc Natl Acad Sci U S A 2019; 116:24840-24851. [PMID: 31744868 DOI: 10.1073/pnas.1908113116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Huntington's disease (HD) is a chronic neurodegenerative disorder characterized by a late clinical onset despite ubiquitous expression of the mutant Huntingtin gene (HTT) from birth. Transcriptional dysregulation is a pivotal feature of HD. Yet, the genes that are altered in the prodromal period and their regulators, which present opportunities for therapeutic intervention, remain to be elucidated. Using transcriptional and chromatin profiling, we found aberrant transcription and changes in histone H3K27acetylation in the striatum of R6/1 mice during the presymptomatic disease stages. Integrating these data, we identified the Elk-1 transcription factor as a candidate regulator of prodromal changes in HD. Exogenous expression of Elk-1 exerted beneficial effects in a primary striatal cell culture model of HD, and adeno-associated virus-mediated Elk-1 overexpression alleviated transcriptional dysregulation in R6/1 mice. Collectively, our work demonstrates that aberrant gene expression precedes overt disease onset in HD, identifies the Elk-1 transcription factor as a key regulator linked to early epigenetic and transcriptional changes in HD, and presents evidence for Elk-1 as a target for alleviating molecular pathology in HD.
Collapse
|
6
|
Colle D, Santos DB, de Souza V, Lopes MW, Leal RB, de Souza Brocardo P, Farina M. Sodium selenite protects from 3-nitropropionic acid-induced oxidative stress in cultured primary cortical neurons. Mol Biol Rep 2018; 46:751-762. [PMID: 30511305 DOI: 10.1007/s11033-018-4531-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
Abstract
Selenium (Se) is an essential trace element for humans; its intake is needed to allow the proper synthesis of 25 different selenoproteins that are necessary to the normal functioning of several organs, including the brain. Accordingly, decreased Se levels have been associated with neurological disorders. In the present study, we investigated the potential beneficial effects of Se, as sodium selenite, against 3-nitropropionic acid (3-NP)-induced oxidative stress in primary cultures of mouse cortical neurons. 3-NP treatment caused a significant decrease in cellular viability, which was accompanied by decreases in mitochondrial complex II activity and reduced glutathione (GSH) content, as well as increases in reactive oxygen species (ROS) generation and oxidized glutathione (GSSG) levels. Sodium selenite pretreatment (6 days) attenuated 3-NP-induced decrease in cell viability. In addition, sodium selenite pretreatment significantly protected against 3-NP-induced increase in ROS generation and decrease in GSH/GSSG ratio. Of note, sodium selenite pretreatment did not change 3-NP-induced decrease of mitochondrial complex II activity, suggesting that Se modulates secondary events resultant from 3-NP-induced mitochondrial dyshomeostasis. In addition, sodium selenite pretreatment significantly increased glutathione peroxidase (GPx) activity. Our data provide insights into the mechanism of protection by sodium selenite, which is related, at least in part, to GPx induction.
Collapse
Affiliation(s)
- Dirleise Colle
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil. .,Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, CEP 88040-900, Brazil.
| | - Danúbia Bonfanti Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Viviane de Souza
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Mark William Lopes
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Rodrigo Bainy Leal
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Patricia de Souza Brocardo
- Departamento de Ciências Morfológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil.
| |
Collapse
|
7
|
Peñalver P, Belmonte-Reche E, Adán N, Caro M, Mateos-Martín ML, Delgado M, González-Rey E, Morales JC. Alkylated resveratrol prodrugs and metabolites as potential therapeutics for neurodegenerative diseases. Eur J Med Chem 2018; 146:123-138. [PMID: 29407944 DOI: 10.1016/j.ejmech.2018.01.037] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 11/17/2022]
Abstract
Resveratrol is a naturally occurring stilbene which has shown promising results as treatment for several neurodegenerative diseases. However, its application is limited due to its low efficacy and bioavailability. Here, we have designed and synthesized alkylated resveratrol prodrugs combining structural modification to improve antioxidant and anti-inflammatory properties and the preparation of prodrugs to extend drug bioavailability. For comparison we also studied resveratrol prodrugs and alkylated resveratrol derivatives. Methylated and butylated resveratrol derivatives showed the best in vitro neuroprotective and anti-inflammatory activity. The glucosyl- and glucosyl-acyl- prodrugs of these derivatives showed lower toxicity on zebra fish embryo. When neuroprotection was examined on pentylenetetrazole challenged zebra fish, they were capable of reverting neuronal damage but to a lower extent than resveratrol. Nevertheless, 3-O-(6'-O-octanoyl)-β-d-glucopyranoside resveratrol (compound 8) recovered AChE activity over 100% whereas resveratrol only up to 92%. In a 3-nitropropionic acid mice model of Huntington's disease, resveratrol derivative 8 delayed the onset and reduced the severity of HD-like symptoms, by improving locomotor activity and protecting against weight loss. Its effects involved an equal antioxidant but better anti-inflammatory profile than resveratrol as shown by SOD2 expression in brain tissue and circulating levels of IL-6 (11 vs 18 pg/mL), respectively. Finally, the octanoyl chain in compound 8 could be playing a role in inflammation and neuronal development indicating it could be acting as a double-drug, instead of as a prodrug.
Collapse
Affiliation(s)
- Pablo Peñalver
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - Efres Belmonte-Reche
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - Norma Adán
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - Marta Caro
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - María Luisa Mateos-Martín
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - Mario Delgado
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain
| | - Elena González-Rey
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain.
| | - Juan Carlos Morales
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain.
| |
Collapse
|
8
|
Dunbar GL, Sandstrom MI, Rossignol J, Lescaudron L. Neurotrophic Enhancers as Therapy for Behavioral Deficits in Rodent Models of Huntington's Disease: Use of Gangliosides, Substituted Pyrimidines, and Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2016; 5:63-79. [PMID: 16801683 DOI: 10.1177/1534582306289367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The interest in using neurotrophic factors as potential treatments for neurodegenerative disorders, such as Huntington's disease, has grown in the past decade. A major impediment for the clinical utility of neurotrophic factors is their inability to cross the blood-brain barrier in therapeutically significant amounts. Although several novel mechanisms for delivering exogenous neurotrophins to the brain have been developed, most of them involve invasive procedures or present significant risks. One approach to circumventing these problems is using therapeutic agents that can be administered systemically and have the ability to enhance the activity of neurotrophic factors. This review highlights the use of gangliosides, substituted pyrimidines, and mesenchymal stem cells as neurotrophic enhancers that have significant therapeutic potential while avoiding the pitfalls of delivering exogenous neurotrophic factors through the blood-brain barrier. The review focuses on the potential of these neurotrophic enhancers for treating the behavioral deficits in rodent models of Huntington's disease.
Collapse
|
9
|
Shinomol GK, Ranganayaki S, Joshi AK, Gayathri N, Gowda H, Muralidhara, Srinivas Bharath MM. Characterization of age-dependent changes in the striatum: Response to the mitochondrial toxin 3-nitropropionic acid. Mech Ageing Dev 2016; 161:66-82. [PMID: 27143313 DOI: 10.1016/j.mad.2016.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/10/2016] [Accepted: 04/23/2016] [Indexed: 11/26/2022]
Abstract
Neurodegenerative phenomena are associated with mitochondrial dysfunction and this could be exacerbated by aging. Age-dependence of mitochondrial response to toxins could help understand these mechanisms and evolve novel therapeutics. 3-Nitropropionic acid (3-NPA) is a mitochondrial toxin that induces neurotoxicity in the striatum via inhibition of complex II. We investigated the age-related events that contribute to 3-NPA toxicity. 3-NPA induced neuronal death, oxidative stress and altered mitochondrial structure in neuronal cells. 3-NPA injection in vivo caused motor impairment, mitochondrial dysfunction and oxidative damage with different trend in young and adult mice. To understand the age-dependent mechanisms, we carried out proteomic analysis of the striatal protein extract from young mice (control: YC vs. 3-NPA treated: YT) and adult mice (control: AC vs. 3-NPA treated: AT). Among the 3752 identified proteins, 33 differentially expressed proteins (mitochondrial, synaptic and microsomal proteins) were unique either to YT or AT. Interestingly, comparison of the proteomic profile in AC and YC indicated that 161 proteins (linked with cytoskeletal structure, neuronal development, axogenesis, protein transport, cell adhesion and synaptic function) were down-regulated in AC compared to YC. We surmise that aging contributes to the cellular and molecular architecture in the mouse striatum with implications for neurodegeneration.
Collapse
Affiliation(s)
- G K Shinomol
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - S Ranganayaki
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - Apurva K Joshi
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - N Gayathri
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India
| | - Harsha Gowda
- Institute of Bioinformatics (IOB), Discoverer, Industrial Technology Park Limited (ITPL), Whitefield, Bangalore 560066, Karnataka, India
| | - Muralidhara
- Department of Biochemistry and Nutrition, Central Food Technological Research Institute, Mysore 570020, Karnataka, India
| | - M M Srinivas Bharath
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India; Neurotoxicology laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 2900, Hosur Road, Bangalore 560029, Karnataka, India.
| |
Collapse
|
10
|
Qi L, Sun X, Li FE, Zhu BS, Braun FK, Liu ZQ, Tang JL, Wu C, Xu F, Wang HH, Velasquez LA, Zhao K, Lei FR, Zhang JG, Shen YT, Zou JX, Meng HM, An GL, Yang L, Zhang XD. HMGB1 Promotes Mitochondrial Dysfunction-Triggered Striatal Neurodegeneration via Autophagy and Apoptosis Activation. PLoS One 2015; 10:e0142901. [PMID: 26565401 PMCID: PMC4643922 DOI: 10.1371/journal.pone.0142901] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 10/28/2015] [Indexed: 01/24/2023] Open
Abstract
Impairments in mitochondrial energy metabolism are thought to be involved in many neurodegenerative diseases. The mitochondrial inhibitor 3-nitropropionic acid (3-NP) induces striatal pathology mimicking neurodegeneration in vivo. Previous studies showed that 3-NP also triggered autophagy activation and apoptosis. In this study, we focused on the high-mobility group box 1 (HMGB1) protein, which is important in oxidative stress signaling as well as in autophagy and apoptosis, to explore whether the mechanisms of autophagy and apoptosis in neurodegenerative diseases are associated with metabolic impairment. To elucidate the role of HMGB1 in striatal degeneration, we investigated the impact of HMGB1 on autophagy activation and cell death induced by 3-NP. We intoxicated rat striata with 3-NP by stereotaxic injection and analyzed changes in expression HMGB1, proapoptotic proteins caspase-3 and phospho-c-Jun amino-terminal kinases (p-JNK). 3-NP–induced elevations in p-JNK, cleaved caspase-3, and autophagic marker LC3-II as well as reduction in SQSTM1 (p62), were significantly reduced by the HMGB1 inhibitor glycyrrhizin. Glycyrrhizin also significantly inhibited 3-NP–induced striatal damage. Neuronal death was replicated by exposing primary striatal neurons in culture to 3-NP. It was clear that HMGB1 was important for basal autophagy which was shown by rescue of cells through HMGB1 targeting shRNA approach.3-NP also induced the expression of HMGB1, p-JNK, and LC3-II in striatal neurons, and p-JNK expression was significantly reduced by shRNA knockdown of HMGB1, an effect that was reversed by exogenously increased expression of HMGB1. These results suggest that HMGB1 plays important roles in signaling for both autophagy and apoptosis in neurodegeneration induced by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Lin Qi
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
- Laboratory of Molecular Neuro-Oncology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States of America
| | - Xue Sun
- Department of Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Feng-E Li
- Department of Interventional Treatment, Tianjin Medical University Cancer Hospital and Institution, Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Bao-Song Zhu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Frank K. Braun
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- Laboratory of Molecular Neuro-Oncology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States of America
| | - Zhi-Qiang Liu
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Jin-Le Tang
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Chao Wu
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Fei Xu
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Hui-Han Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Luis A. Velasquez
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Kui Zhao
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Feng-Rui Lei
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ji-Gang Zhang
- Department of Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yun-Tian Shen
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jian-Xuan Zou
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Hui-Min Meng
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Gang-Li An
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Lin Yang
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
- * E-mail: (XDZ); (LY)
| | - Xing-Ding Zhang
- Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- * E-mail: (XDZ); (LY)
| |
Collapse
|
11
|
Wityak J, McGee KF, Conlon MP, Song RH, Duffy BC, Clayton B, Lynch M, Wang G, Freeman E, Haber J, Kitchen DB, Manning DD, Ismail J, Khmelnitsky Y, Michels P, Webster J, Irigoyen M, Luche M, Hultman M, Bai M, Kuok ID, Newell R, Lamers M, Leonard P, Yates D, Matthews K, Ongeri L, Clifton S, Mead T, Deupree S, Wheelan P, Lyons K, Wilson C, Kiselyov A, Toledo-Sherman L, Beconi M, Muñoz-Sanjuan I, Bard J, Dominguez C. Lead optimization toward proof-of-concept tools for Huntington's disease within a 4-(1H-pyrazol-4-yl)pyrimidine class of pan-JNK inhibitors. J Med Chem 2015; 58:2967-87. [PMID: 25760409 DOI: 10.1021/jm5013598] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Through medicinal chemistry lead optimization studies focused on calculated properties and guided by X-ray crystallography and computational modeling, potent pan-JNK inhibitors were identified that showed submicromolar activity in a cellular assay. Using in vitro ADME profiling data, 9t was identified as possessing favorable permeability and a low potential for efflux, but it was rapidly cleared in liver microsomal incubations. In a mouse pharmacokinetics study, compound 9t was brain-penetrant after oral dosing, but exposure was limited by high plasma clearance. Brain exposure at a level expected to support modulation of a pharmacodynamic marker in mouse was achieved when the compound was coadministered with the pan-cytochrome P450 inhibitor 1-aminobenzotriazole.
Collapse
Affiliation(s)
- John Wityak
- †CHDI Foundation, Inc., 6080 Center Drive, Suite 100, Los Angeles, California 90045, United States
| | - Kevin F McGee
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Michael P Conlon
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Ren Hua Song
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Bryan C Duffy
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Brent Clayton
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Michael Lynch
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Gwen Wang
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Emily Freeman
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - James Haber
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Douglas B Kitchen
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - David D Manning
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Jiffry Ismail
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Yuri Khmelnitsky
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Peter Michels
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Jeff Webster
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Macarena Irigoyen
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Michele Luche
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Monica Hultman
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Mei Bai
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - IokTeng D Kuok
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Ryan Newell
- ‡Albany Molecular Research Inc. (AMRI), 26 Corporate Circle, Albany, New York 12212-5098, United States
| | - Marieke Lamers
- §BioFocus Discovery Services, Charles River Laboratories, Chesterford Research Park, Little Chesterford, CB10 1XL, United Kingdom
| | - Philip Leonard
- §BioFocus Discovery Services, Charles River Laboratories, Chesterford Research Park, Little Chesterford, CB10 1XL, United Kingdom
| | - Dawn Yates
- §BioFocus Discovery Services, Charles River Laboratories, Chesterford Research Park, Little Chesterford, CB10 1XL, United Kingdom
| | - Kim Matthews
- §BioFocus Discovery Services, Charles River Laboratories, Chesterford Research Park, Little Chesterford, CB10 1XL, United Kingdom
| | - Lynette Ongeri
- §BioFocus Discovery Services, Charles River Laboratories, Chesterford Research Park, Little Chesterford, CB10 1XL, United Kingdom
| | - Steve Clifton
- §BioFocus Discovery Services, Charles River Laboratories, Chesterford Research Park, Little Chesterford, CB10 1XL, United Kingdom
| | - Tania Mead
- §BioFocus Discovery Services, Charles River Laboratories, Chesterford Research Park, Little Chesterford, CB10 1XL, United Kingdom
| | - Susan Deupree
- ∥Tandem Laboratories, 2202 Ellis Road, Durham, North Carolina 27703, United States
| | - Pat Wheelan
- ∥Tandem Laboratories, 2202 Ellis Road, Durham, North Carolina 27703, United States
| | - Kathy Lyons
- ⊥Pharmacokinetics Consultant to CHDI, P.O. Box 64, Holland, New York 14080, United States
| | - Claire Wilson
- #Evotec, 114 Milton Park, Abingdon, OX14 4SA, United Kingdom
| | - Alex Kiselyov
- †CHDI Foundation, Inc., 6080 Center Drive, Suite 100, Los Angeles, California 90045, United States
| | - Leticia Toledo-Sherman
- †CHDI Foundation, Inc., 6080 Center Drive, Suite 100, Los Angeles, California 90045, United States
| | - Maria Beconi
- †CHDI Foundation, Inc., 6080 Center Drive, Suite 100, Los Angeles, California 90045, United States
| | - Ignacio Muñoz-Sanjuan
- †CHDI Foundation, Inc., 6080 Center Drive, Suite 100, Los Angeles, California 90045, United States
| | - Jonathan Bard
- †CHDI Foundation, Inc., 6080 Center Drive, Suite 100, Los Angeles, California 90045, United States
| | - Celia Dominguez
- †CHDI Foundation, Inc., 6080 Center Drive, Suite 100, Los Angeles, California 90045, United States
| |
Collapse
|
12
|
GEORGIOU-KARISTIANIS NELLIE, LONG JEFFREYD, LOURENS SPENCERG, STOUT JULIEC, MILLS JAMESA, PAULSEN JANES. Movement sequencing in Huntington disease. World J Biol Psychiatry 2014; 15:459-71. [PMID: 24678867 PMCID: PMC4389285 DOI: 10.3109/15622975.2014.895042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES To examine longitudinal changes in movement sequencing in prodromal Huntington's disease (HD) participants (795 prodromal HD; 225 controls) from the PREDICT-HD study. METHODS Prodromal HD participants were tested over seven annual visits and were stratified into three groups (low, medium, high) based on their CAG-Age Product (CAP) score, which indicates likely increasing proximity to diagnosis. A cued movement sequence task assessed the impact of advance cueing on response initiation and execution via three levels of advance information. RESULTS Compared to controls, all CAP groups showed longer initiation and movement times across all conditions at baseline, demonstrating a disease gradient for the majority of outcomes. Across all conditions, the high CAP group had the highest mean for baseline testing, but also demonstrated an increase in movement time across the study. For initiation time, the high CAP group showed the highest mean baseline time across all conditions, but also faster decreasing rates of change over time. CONCLUSIONS With progress to diagnosis, participants may increasingly use compensatory strategies, as evidenced by faster initiation. However, this occurred in conjunction with slowed execution times, suggesting a decline in effectively accessing control processes required to translate movement into effective execution.
Collapse
Affiliation(s)
| | - JEFFREY D. LONG
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA,Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - SPENCER G. LOURENS
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - JULIE C. STOUT
- School of Psychological Sciences, Monash University, Wellington Road, Clayton, Victoria, Australia,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - JAMES A. MILLS
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - JANE S. PAULSEN
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA,Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA,Department of Psychology, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
13
|
Ethyl pyruvate ameliorates 3-nitropropionic acid-induced striatal toxicity through anti-neuronal cell death and anti-inflammatory mechanisms. Brain Behav Immun 2014; 38:151-65. [PMID: 24576481 DOI: 10.1016/j.bbi.2014.01.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/15/2014] [Accepted: 01/26/2014] [Indexed: 01/17/2023] Open
Abstract
The potential neuroprotective value of ethyl pyruvate (EP) for the treatment of the striatal toxicity is largely unknown. We investigated whether EP promotes the survival of striatal neurons in a 3-nitropropionic acid (3-NP)-induced mouse model of Huntington's disease (HD). EP (5, 10, 20, and 40mg/kg/day, i.p.) was daily injected from 30min before 3-NP intoxication (pretreatment) and from onset/progression/peak point of neurological impairment by 3-NP intoxication. EP produced a neuroprotective effect in dose- and time-dependant manners. EP pretreatment of 40mg/kg/day produced the best neuroprotective effect among other conditions. Pretreatment of EP significantly attenuated neurological impairment and lethality and prevented formation of lesion area and neuronal loss in the striatum after 3-NP intoxication. This neuroprotection afforded by EP was associated with the suppression of succinate dehydrogenase activity, apoptosis, and microglial activation. The suppressive effect of EP corresponded to the down-regulation of mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB) signal pathways, and mRNA expression of inflammatory mediators including tumor necrosis factor-alpha, interleukin (IL)-1β, IL-6, inducible nitric oxide synthase, and cyclooxygenase-2 in the striatum after 3-NP intoxication. Interestingly, the intrathecal introduction of inhibitors MAPKs and NF-κB into control mice decreased the lethality after 3-NP intoxication. Our findings indicate that EP may effectively alleviate 3-NP-induced striatal toxicity by inhibition of the MAPKs and NF-κB pathways in the striatum, and that EP has a wide therapeutic window, suggesting that EP may have therapeutic value in the treatment of aspects of HD's disease related to inflammation.
Collapse
|
14
|
Choi YJ, Kim NH, Lim MS, Lee HJ, Kim SS, Chun W. Geldanamycin attenuates 3‑nitropropionic acid‑induced apoptosis and JNK activation through the expression of HSP 70 in striatal cells. Int J Mol Med 2014; 34:24-34. [PMID: 24756698 PMCID: PMC4072345 DOI: 10.3892/ijmm.2014.1747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/14/2014] [Indexed: 11/05/2022] Open
Abstract
Although selective striatal cell death is a characteristic hallmark in the pathogenesis of Huntington's disease (HD), the underlying mechanism of striatal susceptibility remains to be clarified. Heat shock proteins (HSPs) have been reported to suppress the aggregate formation of mutant huntingtin and concurrent striatal cell death. In a previous study, we observed that heat shock transcription factor 1 (HSF1), a major transcription factor of HSPs, significantly attenuated 3‑nitropropionic acid (3NP)‑induced reactive oxygen species (ROS) production and apoptosis through the expression of HSP 70 in striatal cells. To investigate the differential roles of HSPs in 3NP‑induced striatal cell death, the effect of geldanamycin (GA), an HSP 90 inhibitor, was examined in 3NP‑stimulated striatal cells. GA significantly attenuated 3NP‑induced striatal apoptosis and ROS production with an increased expression of HSP 70. Triptolide (TL), an HSP 70 inhibitor, abolished GA‑mediated protective effects in 3NP‑stimulated striatal cells. To understand the underlying mechanism by which GA‑mediated HSP 70 protects striatal cells against 3NP stimulation, the involvement of various signaling pathways was examined. GA significantly attenuated 3NP‑induced c‑Jun N‑terminal kinase (JNK) phosphorylation and subsequent c‑Jun phosphorylation in striatal cells. Taken together, the present study demonstrated that GA exhibits protective properties against 3NP‑induced apoptosis and JNK activation via the induction of HSP 70 in striatal cells, suggesting that expression of HSP 70 may be a valuable therapeutic target in the treatment of HD.
Collapse
Affiliation(s)
- Yong-Joon Choi
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Nam Ho Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Man Sup Lim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Sung Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| |
Collapse
|
15
|
Pozniak PD, White MK, Khalili K. TNF-α/NF-κB signaling in the CNS: possible connection to EPHB2. J Neuroimmune Pharmacol 2013; 9:133-41. [PMID: 24277482 DOI: 10.1007/s11481-013-9517-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/13/2013] [Indexed: 12/27/2022]
Abstract
Tumor necrosis factor-alpha, TNF-α, is a cytokine that is a well-known factor in multiple disease conditions and is recognized for its major role in central nervous system signaling. TNF-α signaling is most commonly associated with neurotoxicity, but in some conditions it has been found to be neuroprotective. TNF-α has long been known to induce nuclear factor-kappa B, NF-κB, signaling by, in most cases, translocating the p65 (RelA) DNA binding factor to the nucleus. p65 is a key member of NF-κB, which is well established as a family of transcription factors that regulates many signaling events, including growth and process development, in neuronal cell populations. NF-κB has been shown to affect both the receiving aspect of neuronal signaling events in dendritic development as well as the sending of neuronal signals in axonal development. In both cases, NK-κB functions as a promoter and/or inhibitor of growth, depending on the environmental conditions and signaling cascade. In addition, NF-κB is involved in memory formation or neurogenesis, depending on the region of the brain in which the signaling occurs. The ephrin (Eph) receptor family represents a subfamily of receptor tyrosine kinases, RTKs, which received much attention due to its potential involvement in neuronal cell health and function. There are two subsets of ephrin receptors, Eph A and Eph B, each with distinct functions in cardiovascular and skeletal development and axon guidance and synaptic plasticity. The presence of multiple binding sites for NF-κB within the regulatory region of EphB2 gene and its potential regulation by NF-κB pathway suggests that TNF-α may modulate EphB2 via NF-κB and that this may contribute to the neuroprotective activity of TNF-α.
Collapse
Affiliation(s)
- Paul D Pozniak
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 741 MERB, 3500N. Broad Street, Philadelphia, PA, 19140, USA
| | | | | |
Collapse
|
16
|
Park JE, Lee ST, Im WS, Kim M. Growth Hormone Deteriorates the Functional Outcome in an Experimental Model of Huntington's Disease Induced by 3-Nitropionic Acid. J Mov Disord 2013; 6:28-33. [PMID: 24868423 PMCID: PMC4027642 DOI: 10.14802/jmd.13007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 06/14/2013] [Accepted: 08/20/2013] [Indexed: 12/18/2022] Open
Abstract
Background and Purpose: Growth hormone (GH) has been frequently used to control the aging process in healthy individuals, probably due to its slowing effect on senescence-associated degeneration. Mitochondrial dysfunction is related to the aging process, and one of the chemical models of Huntington’s disease is that it can be induced by mitochondrial toxin. To investigate the potential application of GH to modify the progression of Huntington’s disease (HD), we examined whether GH can protect the functional deterioration by striatal damage induced by 3-nitropropionic acid (3NP). Methods: 3NP (63 mg/kg/day) was delivered to Lewis rats by osmotic pumps for five consecutive days, and the rats received intraperitoneal administration of GH or vehicle (saline) throughout the experiment. Neurological deficits and body weight were monitored. A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test was performed to further determine the mitochondrial activity in cultured N18TG2 neuroblastoma cells in vitro. Results: 3NP-treated rats showed progressive neurologic deficits with striatal damage. Application of GH accelerated behavioral deterioration, particularly between day 3 and day 5, resulting in reduced survival outcome. The body weights of rats given 3NP were decreased, but GH did not affect such decrease compared to the non-treated control group. The effect of GH on cultured neuronal cells was a decrease in the MTT absorbance, suggesting a lower number of cells in a dose dependent pattern. Conclusions: Those results suggest that application of GH to a 3NP-induced experimental model of HD deteriorates the progress of functional deficits, possibly disturbing mitochondrial activities.
Collapse
Affiliation(s)
- Jung-Eun Park
- Department of Neurology, Clinical Research Institute, Seoul National University Hospital, Seoul, Korea ; Program in Neuroscience, Seoul National University, Seoul, Korea
| | - Soon-Tae Lee
- Department of Neurology, Clinical Research Institute, Seoul National University Hospital, Seoul, Korea ; Program in Neuroscience, Seoul National University, Seoul, Korea
| | - Woo-Seok Im
- Department of Neurology, Clinical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Manho Kim
- Department of Neurology, Clinical Research Institute, Seoul National University Hospital, Seoul, Korea ; Program in Neuroscience, Seoul National University, Seoul, Korea
| |
Collapse
|
17
|
Binawade Y, Jagtap A. Neuroprotective Effect of Lutein Against 3-Nitropropionic Acid–Induced Huntington's Disease–Like Symptoms: Possible Behavioral, Biochemical, and Cellular Alterations. J Med Food 2013; 16:934-43. [DOI: 10.1089/jmf.2012.2698] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yogita Binawade
- Department of Pharmacology, Bombay College of Pharmacy, Mumbai, India
| | - Aarti Jagtap
- Department of Pharmacology, Bombay College of Pharmacy, Mumbai, India
| |
Collapse
|
18
|
Essential roles of mitochondrial depolarization in neuron loss through microglial activation and attraction toward neurons. Brain Res 2013; 1505:75-85. [PMID: 23415650 DOI: 10.1016/j.brainres.2013.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/04/2013] [Accepted: 02/05/2013] [Indexed: 12/17/2022]
Abstract
As life spans increased, neurodegenerative disorders that affect aging populations have also increased. Progressive neuronal loss in specific brain regions is the most common cause of neurodegenerative disease; however, key determinants mediating neuron loss are not fully understood. Using a model of mitochondrial membrane potential (ΔΨm) loss, we found only 25% cell loss in SH-SY5Y (SH) neuronal mono-cultures, but interestingly, 85% neuronal loss occurred when neurons were co-cultured with BV2 microglia. SH neurons overexpressing uncoupling protein 2 exhibited an increase in neuron-microglia interactions, which represent an early step in microglial phagocytosis of neurons. This result indicates that ΔΨm loss in SH neurons is an important contributor to recruitment of BV2 microglia. Notably, we show that ΔΨm loss in BV2 microglia plays a crucial role in microglial activation and phagocytosis of damaged SH neurons. Thus, our study demonstrates that ΔΨm loss in both neurons and microglia is a critical determinant of neuron loss. These findings also offer new insights into neuroimmunological and bioenergetical aspects of neurodegenerative disease.
Collapse
|
19
|
Korean Red Ginseng Extract Attenuates 3-Nitropropionic Acid-Induced Huntington's-Like Symptoms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:237207. [PMID: 23431333 PMCID: PMC3568869 DOI: 10.1155/2013/237207] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 11/29/2012] [Accepted: 12/02/2012] [Indexed: 11/17/2022]
Abstract
Korean red ginseng (KRG) possesses neuroprotective activity. However, the potential neuroprotective value of KRG for the striatal toxicity is largely unknown. We investigated whether KRG extract (KRGE) could have a neuroprotective effect in a 3-nitropropionic acid- (3-NP) induced (i.p.) Huntington's disease (HD) model. KRGE (50, 100, and 250 mg/kg/day, p.o.) was administrated 10 days before 3-NP injection (pre-administration), from the same time with 3-NP injection (co-administration), or from the peak point of neurological impairment by 3-NP injection (post-administration). Pre-administration of KRGE produced the greatest neuroprotective effect in this model. Pre-administration of KRGE significantly decreased 3-NP-induced neurological impairment, lethality, lesion area, and neuronal loss in the 3-NP-injected striatum. KRGE attenuated microglial activation and phosphorylation of mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB) signal pathway. KRGE also reduced the level of mRNA expression of tumor necrosis factor-alpha, interleukin- (IL-) 1β, IL-6, inducible nitric oxide synthase, and OX-42. Interestingly, the intrathecal administration of SB203580 (a p38 inhibitor) or PD98059 (an inhibitor of MAPK Kinase, MEK) increased the survival rate in the 3-NP-induced HD model. Pre-administration of KRGE may effectively inhibit 3-NP-induced striatal toxicity via the inhibition of the phosphorylation of MAPKs and NF-κB pathways, indicating its therapeutic potential for suppressing Huntington's-like symptoms.
Collapse
|
20
|
Junyent F, de Lemos L, Verdaguer E, Pallàs M, Folch J, Beas-Zárate C, Camins A, Auladell C. Lack of Jun-N-terminal kinase 3 (JNK3) does not protect against neurodegeneration induced by 3-nitropropionic acid. Neuropathol Appl Neurobiol 2012; 38:311-21. [PMID: 21883373 DOI: 10.1111/j.1365-2990.2011.01214.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
AIMS 3-Nitropropionic acid (3-NP) is a toxin that replicates most of the clinical and pathophysiological symptoms of Huntington's disease, inducing neurodegeneration in the striatum due to the inhibition of mitochondrial succinate dehydrogenase. Different pathways have been implicated in the cell death induced by 3-NP in rodents. One of them is the Jun-N-terminal kinase (JNK) pathway, which may play a role in the neurodegenerative process in different diseases. Moreover, the lack of one isoform of JNK (JNK3) has been associated with neuroprotection in different experimental models of neurodegeneration. Therefore, in the present study the role of JNK3 in the experimental Huntington's model induced by 3-NP administration was evaluated. METHODS 3-NP was intraperitoneally administered once a day for 3 days to wild-type and Jnk3-null mice. Coronal brain sections were used to determine cell death and astrogliosis in striatum. Western blots were performed to determine the involvement of different pathways in both wild-type and Jnk3-null mice. RESULTS Although JNK activation was observed following 3-NP administration, the results indicate that the lack of JNK3 does not confer neuroprotection against 3-NP toxicity. Thus, other pathways must be involved in the neurodegeneration induced in this model. One of the possible pathways towards 3-NP-induced apoptosis could involve the calpains, as their activity was increased in wild-type and Jnk3-null mice. CONCLUSION Although JNK3 is a key protein involved in cell death in different neurodegenerative diseases, the present study demonstrates that the lack of JNK3 does not confer neuroprotection against 3-NP-induced neuronal death.
Collapse
Affiliation(s)
- F Junyent
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina, Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Universitat de Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Akopian G, Crawford C, Petzinger G, Jakowec MW, Walsh JP. Brief mitochondrial inhibition causes lasting changes in motor behavior and corticostriatal synaptic physiology in the Fischer 344 rat. Neuroscience 2012; 215:149-59. [PMID: 22554779 PMCID: PMC3371111 DOI: 10.1016/j.neuroscience.2012.04.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 04/19/2012] [Accepted: 04/25/2012] [Indexed: 11/26/2022]
Abstract
The striatum is particularly vulnerable to mitochondrial dysfunction and this problem is linked to pathology created by environmental neurotoxins, stimulants like amphetamine, and metabolic disease and ischemia. We studied the course of recovery following a single systemic injection of the mitochondrial complex II inhibitor 3-nitropropionic acid (3-NP) and found 3-NP caused lasting changes in motor behavior that were associated with altered activity-dependent plasticity at corticostriatal synapses in Fischer 344 rats. The changes in synapse behavior varied with the time after exposure to the 3-NP injection. The earliest time point studied, 24h after 3-NP, revealed 3-NP-induced an exaggeration of D1 Dopamine (DA) receptor dependent long-term potentiation (LTP) that reversed to normal by 48 h post-3-NP exposure. Thereafter, the likelihood and degree of inducing D2 DA receptor dependent long-term depression (LTD) gradually increased, relative to saline controls, peaking at 1 month after the 3-NP exposure. NMDA receptor binding did not change over the same post 3-NP time points. These data indicate even brief exposure to 3-NP can have lasting behavioral effects mediated by changes in the way DA and glutamate synapses interact.
Collapse
Affiliation(s)
- G Akopian
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089-0191, USA
| | | | | | | | | |
Collapse
|
22
|
Lam PY, Ko KM. Beneficial effect of (-)schisandrin B against 3-nitropropionic acid-induced cell death in PC12 cells. Biofactors 2012; 38:219-25. [PMID: 22488872 DOI: 10.1002/biof.1009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 01/07/2012] [Indexed: 01/15/2023]
Abstract
Huntington's disease (HD) is characterized by the dysfunction of mitochondrial energy metabolism, which is associated with the functional impairment of succinate dehydrogenase (mitochondrial complex II), and pyruvate dehydrogenase (PDH). Treatment with 3-nitropropionic acid (3-NP), a potent irreversible inhibitor of succinate dehydrogenase, replicates most of the pathophysiological features of HD. In the present study, we investigated the effect of (-)schisandrin B [(-)Sch B, a potent enantiomer of schisandrin B] on 3-NP-induced cell injury in rat differentiated neuronal PC12 cells. The 3-NP caused cell necrosis, as assessed by lactate dehydrogenase (LDH) leakage, and mitochondrion-dependent cell apoptosis, as assessed by caspase-3 and caspase-9 activation, in differentiated PC12 cells. The cytotoxicity induced by 3-NP was associated with a depletion of cellular reduced glutathione (GSH) as well as the activation of redox-sensitive c-Jun N-terminal kinase (JNK) pathway and the inhibition of PDH. (-)Sch B pretreatment (5 and 15 μM) significantly reduced the extent of necrotic and apoptotic cell death in 3-NP-challenged cells. The cytoprotection afforded by (-)Sch B pretreatment was associated with the attenuation of 3-NP-induced GSH depletion as well as JNK activation and PDH inhibition. (-)Sch B pretreatment enhanced cellular glutathione redox status and ameliorated the 3-NP-induced cellular energy crisis, presumably by suppressing the activated JNK-mediated PDH inhibition, thereby protecting against necrotic and apoptotic cell death in differentiated PC12 cells.
Collapse
Affiliation(s)
- Philip Y Lam
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China
| | | |
Collapse
|
23
|
Chaumeil MM, Valette J, Baligand C, Brouillet E, Hantraye P, Bloch G, Gaura V, Rialland A, Krystkowiak P, Verny C, Damier P, Remy P, Bachoud-Levi AC, Carlier P, Lebon V. pH as a biomarker of neurodegeneration in Huntington's disease: a translational rodent-human MRS study. J Cereb Blood Flow Metab 2012; 32:771-9. [PMID: 22373643 PMCID: PMC3345921 DOI: 10.1038/jcbfm.2012.15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Early diagnosis and follow-up of neurodegenerative diseases are often hampered by the lack of reliable biomarkers. Neuroimaging techniques like magnetic resonance spectroscopy (MRS) offer promising tools to detect biochemical alterations at early stages of degeneration. Intracellular pH, which can be measured noninvasively by (31)P-MRS, has shown variations in several brain diseases. Our purpose has been to evaluate the potential of MRS-measured pH as a relevant biomarker of early degeneration in Huntington's disease (HD). We used a translational approach starting with a preclinical validation of our hypothesis before adapting the method to HD patients. (31)P-MRS-derived cerebral pH was first measured in rodents during chronic intoxication with 3-nitropropionic acid (3NP). A significant pH increase was observed early into the intoxication protocol (pH=7.17±0.02 after 3 days) as compared with preintoxication (pH=7.08±0.03). Furthermore, pH changes correlated with the 3NP-induced inhibition of succinate dehydrogenase and preceded striatum lesions. Using a similar MRS approach implemented on a clinical MRI, we then showed that cerebral pH was significantly higher in HD patients (n=7) than in healthy controls (n=6) (7.05±0.03 versus 7.02±0.01, respectively, P=0.026). Altogether, both preclinical and human data strongly argue in favor of MRS-measured pH being a promising biomarker for diagnosis and follow-up of HD.
Collapse
Affiliation(s)
- Myriam M Chaumeil
- 1] Commissariat à l'Energie Atomique, Institut d'Imagerie Biomédicale, Molecular Imaging Research Center, Centre National de la Recherche Scientifique, Unité de Recherche Associée, Fontenay-aux-Roses Cedex, France [2] Institut de Myologie, Laboratoire de RMN, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wu CL, Yin JH, Hwang CS, Chen SD, Yang DY, Yang DI. c-Jun-dependent sulfiredoxin induction mediates BDNF protection against mitochondrial inhibition in rat cortical neurons. Neurobiol Dis 2012; 46:450-62. [PMID: 22402332 DOI: 10.1016/j.nbd.2012.02.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 01/20/2012] [Accepted: 02/20/2012] [Indexed: 01/19/2023] Open
Abstract
In current study, we tested the hypothesis that c-Jun-dependent sulfiredoxin expression mediates protective effects of brain-derived neurotrophic factor (BDNF) against neurotoxicity induced by 3-nitropropionic acid (3-NP), a mitochondrial complex II inhibitor, in primary rat cortical cultures. We found that BDNF-dependent c-Jun expression and nuclear translocation required prior phosphorylation of extracellular signal-regulated kinase (ERK)1/2, but not Akt. BDNF also transiently activated the expression of sulfiredoxin, an ATP-dependent antioxidant enzyme, at both mRNA and protein levels. Furthermore, both c-Jun siRNA and ERK1/2 inhibitor PD98059 suppressed BDNF-induced sulfiredoxin expression. Finally, PD98059, c-Jun siRNA, and sulfiredoxin siRNA all abrogated BDNF-mediated 3-NP resistance. Together, these results established a signaling cascade of "BDNF → ERK1/2-Pi → c-Jun → sulfiredoxin → 3-NP resistance". We therefore conclude that c-Jun-induced sulfiredoxin mediates the BDNF-dependent neuroprotective effects against 3-NP toxicity in primary rat cortical neurons, at least in part.
Collapse
Affiliation(s)
- Chia-Lin Wu
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
25
|
Bowman AB, Kwakye GF, Herrero Hernández E, Aschner M. Role of manganese in neurodegenerative diseases. J Trace Elem Med Biol 2011; 25:191-203. [PMID: 21963226 PMCID: PMC3230726 DOI: 10.1016/j.jtemb.2011.08.144] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 08/16/2011] [Indexed: 12/17/2022]
Abstract
Manganese (Mn) is an essential ubiquitous trace element that is required for normal growth, development and cellular homeostasis. Exposure to high Mn levels causes a clinical disease characterized by extrapyramidal symptom resembling idiopathic Parkinson's disease (IPD). The present review focuses on the role of various transporters in maintaining brain Mn homeostasis along with recent methodological advances in real-time measurements of intracellular Mn levels. We also provide an overview on the role for Mn in IPD, discussing the similarities (and differences) between manganism and IPD, and the relationship between α-synuclein and Mn-related protein aggregation, as well as mitochondrial dysfunction, Mn and PD. Additional sections of the review discuss the link between Mn and Huntington's disease (HD), with emphasis on huntingtin function and the potential role for altered Mn homeostasis and toxicity in HD. We conclude with a brief survey on the potential role of Mn in the etiologies of Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS) and prion disease. Where possible, we discuss the mechanistic commonalities inherent to Mn-induced neurotoxicity and neurodegenerative disorders.
Collapse
Affiliation(s)
- Aaron B Bowman
- Department of Neurology, Vanderbilt Kennedy Center, Center for Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN 37232-8552, United States
| | | | | | | |
Collapse
|
26
|
Junyent F, de Lemos L, Verdaguer E, Folch J, Ferrer I, Ortuño-Sahagún D, Beas-Zárate C, Romero R, Pallàs M, Auladell C, Camins A. Gene expression profile in JNK3 null mice: a novel specific activation of the PI3K/AKT pathway. J Neurochem 2011; 117:244-52. [DOI: 10.1111/j.1471-4159.2011.07195.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Ji L, Shen K, Jiang P, Morahan G, Wang Z. Critical roles of cellular glutathione homeostasis and jnk activation in andrographolide-mediated apoptotic cell death in human hepatoma cells. Mol Carcinog 2011; 50:580-91. [DOI: 10.1002/mc.20741] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 12/08/2010] [Accepted: 12/24/2010] [Indexed: 01/11/2023]
|
28
|
Dowie MJ, Scotter EL, Molinari E, Glass M. The therapeutic potential of G-protein coupled receptors in Huntington's disease. Pharmacol Ther 2010; 128:305-23. [PMID: 20708032 DOI: 10.1016/j.pharmthera.2010.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 07/14/2010] [Indexed: 01/29/2023]
Abstract
Huntington's disease is a late-onset autosomal dominant inherited neurodegenerative disease characterised by increased symptom severity over time and ultimately premature death. An expanded CAG repeat sequence in the huntingtin gene leads to a polyglutamine expansion in the expressed protein, resulting in complex dysfunctions including cellular excitotoxicity and transcriptional dysregulation. Symptoms include cognitive deficits, psychiatric changes and a movement disorder often referred to as Huntington's chorea, which involves characteristic involuntary dance-like writhing movements. Neuropathologically Huntington's disease is characterised by neuronal dysfunction and death in the striatum and cortex with an overall decrease in cerebral volume (Ho et al., 2001). Neuronal dysfunction begins prior to symptom presentation, and cells of particular vulnerability include the striatal medium spiny neurons. Huntington's is a devastating disease for patients and their families and there is currently no cure, or even an effective therapy for disease symptoms. G-protein coupled receptors are the most abundant receptor type in the central nervous system and are linked to complex downstream pathways, manipulation of which may have therapeutic application in many neurological diseases. This review will highlight the potential of G-protein coupled receptor drug targets as emerging therapies for Huntington's disease.
Collapse
Affiliation(s)
- Megan J Dowie
- Centre for Brain Research, University of Auckland, Private Bag 92019 Auckland, New Zealand
| | | | | | | |
Collapse
|
29
|
de Lemos L, Junyent F, Verdaguer E, Folch J, Romero R, Pallàs M, Ferrer I, Auladell C, Camins A. Differences in activation of ERK1/2 and p38 kinase in Jnk3 null mice following KA treatment. J Neurochem 2010; 114:1315-22. [PMID: 20534003 DOI: 10.1111/j.1471-4159.2010.06853.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The MAPK family is formed by extracellular signal-regulated kinases p38 kinase and stress-activated protein kinases (SAPK/JNK). There are three genes that encode for three JNK proteins. JNK3 is mainly expressed in the central nervous system and has been related to various processes in that tissue. Specifically, JNK3 plays a crucial role in neuronal death in several neurodegenerative diseases. The activation of this kinase has been described in epilepsy, Alzheimer's disease, Parkinson's disease and Huntington's disease. Different studies have shown that the lack of the Jnk3 gene confers neuroprotection. However, the specific mechanism involved in such neuroprotection has not yet been elucidated. Therefore, in the present study, we analyzed the neuroprotection in mice lacking Jnk3 against neuronal death induced by kainic acid. Moreover, we analyzed the activation of different MAPKs. The results revealed that neuronal death was attenuated and different activation/inactivation of p38 and extracellular signal-regulated kinases 1/2 was reported with respect to control. Therefore, the data indicate that the lack of the JNK3 protein modulates other MAPKs and these changes could also have a pivotal role in neuroprotection.
Collapse
Affiliation(s)
- Luisa de Lemos
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mirandola SR, Melo DR, Saito A, Castilho RF. 3-nitropropionic acid-induced mitochondrial permeability transition: comparative study of mitochondria from different tissues and brain regions. J Neurosci Res 2010; 88:630-9. [PMID: 19795369 DOI: 10.1002/jnr.22239] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The adult rat striatum is particularly vulnerable to systemic administration of the succinate dehydrogenase inhibitor 3-nitropropionic acid (3NP), which is known to induce degeneration of the caudate-putamen, as occurs in Huntington's disease. The aim of the present study was to compare the susceptibility of isolated mitochondria from different rat brain regions (striatum, cortex, and cerebellum) as well as from the liver, kidney, and heart to mitochondrial permeability transition (MPT) induced by 3NP and Ca(2+). In the presence of micromolar Ca(2+) concentrations, 3NP induces MPT in a dose-dependent manner, as estimated by mitochondrial swelling and a decrease in the transmembrane electrical potential. A 3NP concentration capable of promoting a 10% inhibition of ADP-stimulated, succinate-supported respiration was sufficient to stimulate Ca(2+)-induced MPT. Brain and heart mitochondria were generally more sensitive to 3NP and Ca(2+)-induced MPT than mitochondria from liver and kidney. In addition, a partial inhibition of mitochondrial respiration by 3NP resulted in more pronounced MPT in striatal mitochondria than in cortical or cerebellar organelles. A similar inhibition of succinate dehydrogenase activity was observed in rat tissue homogenates obtained from various brain regions as well as from liver, kidney, and heart 24 hr after a single i.p. 3NP dose. Mitochondria isolated from forebrains of 3NP-treated rats were also more susceptible to Ca(2+)-induced MPT than those of control rats. We propose that the increased susceptibility of the striatum to 3NP-induced neurodegeneration may be partially explained by its susceptibility to MPT, together with the greater vulnerability of this brain region to glutamate receptor-mediated Ca(2+) influx.
Collapse
Affiliation(s)
- Sandra R Mirandola
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas , Campinas, Brazil
| | | | | | | |
Collapse
|
31
|
Han I, You Y, Kordower JH, Brady ST, Morfini GA. Differential vulnerability of neurons in Huntington's disease: the role of cell type-specific features. J Neurochem 2010; 113:1073-91. [PMID: 20236390 DOI: 10.1111/j.1471-4159.2010.06672.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Abnormal expansion of a polyglutamine tract in huntingtin (Htt) protein results in Huntington's disease (HD), an autosomal dominant neurodegenerative disorder involving progressive loss of motor and cognitive function. Contrasting with the ubiquitous tissue expression of polyglutamine-expanded Htt, HD pathology is characterized by the increased vulnerability of specific neuronal populations within the striatum and the cerebral cortex. Morphological, biochemical, and functional characteristics of neurons affected in HD that might render these cells more vulnerable to the toxic effects of polyglutamine-Htt are covered in this review. The differential vulnerability of neurons observed in HD is discussed in the context of various major pathogenic mechanisms proposed to date, and in line with evidence showing a 'dying-back' pattern of degeneration in affected neuronal populations.
Collapse
Affiliation(s)
- Ina Han
- Department of Anatomy and Cell Biology. University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
32
|
Túnez I, Tasset I, Pérez-De La Cruz V, Santamaría A. 3-Nitropropionic acid as a tool to study the mechanisms involved in Huntington's disease: past, present and future. Molecules 2010; 15:878-916. [PMID: 20335954 PMCID: PMC6263191 DOI: 10.3390/molecules15020878] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 01/12/2010] [Accepted: 02/01/2010] [Indexed: 11/17/2022] Open
Abstract
Huntington's disease (HD) is an inheritable autosomal-dominant disorder whose causal mechanisms remain unknown. Experimental models have begun to uncover these pathways, thus helping to understand the mechanisms implicated and allowing for the characterization of potential targets for new therapeutic strategies. 3-Nitropropionic acid is known to produce in animals behavioural, biochemical and morphologic changes similar to those occurring in HD. For this reason, this phenotypic model is gaining attention as a valuable tool to mimick this disorder and further developing new therapies. In this review, we will focus on the past and present research of this molecule, to finally bring a perspective on what will be next in this promising field of study.
Collapse
Affiliation(s)
- Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Maimónides de Investigaciones Biomédicas de Córdoba, Universidad de Córdoba, Av. Menéndez Pidal s/n, 14004 Córdoba, Spain.
| | | | | | | |
Collapse
|
33
|
Quaegebeur A, Carmeliet P. Oxygen sensing: a common crossroad in cancer and neurodegeneration. Curr Top Microbiol Immunol 2010; 345:71-103. [PMID: 20582529 DOI: 10.1007/82_2010_83] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prolyl hydroxylase domain (PHD) proteins are cellular oxygen sensors that orchestrate an adaptive response to hypoxia and oxidative stress, executed by hypoxia-inducible factors (HIFs). By increasing oxygen supply, reducing oxygen consumption, and reprogramming metabolism, the PHD/HIF pathway confers tolerance towards hypoxic and oxidative stress. This review discusses the involvement of the PHD/HIF response in two, at first sight, entirely distinct pathologies with opposite outcome, i.e. cancer leading to cellular growth and neurodegeneration resulting in cell death. However, these disorders share common mechanisms of sensing oxygen and oxidative stress. We will focus on how PHD/HIF signaling is pathogenetically implicated in metabolic and vessel alterations in these diseases and how manipulation of this pathway might offer novel treatment opportunities.
Collapse
Affiliation(s)
- Annelies Quaegebeur
- Vesalius Research Center (VRC), VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, 3000, Leuven, Belgium
| | | |
Collapse
|
34
|
Discriminative behavioral assessment unveils remarkable reactive astrocytosis and early molecular correlates in basal ganglia of 3-nitropropionic acid subchronic treated rats. Neurochem Int 2010; 56:152-60. [DOI: 10.1016/j.neuint.2009.09.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 08/21/2009] [Accepted: 09/18/2009] [Indexed: 01/28/2023]
|
35
|
Lukács A, Szabó A, Papp A, Vezér T. Altered open field behavior in rats induced by acute administration of 3-nitropropionic acid: possible glutamatergic and dopaminergic involvement. ACTA BIOLOGICA HUNGARICA 2009; 60:359-67. [PMID: 20015828 DOI: 10.1556/abiol.60.2009.4.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
3-nitropropionic acid (3-NP), a substance used for modelling Huntington's disease, was given to male Wistar rats in a single 20 mg/kg b.w. dose, and the resulting behavioral alterations in spontaneous locomotor activity were measured after 30 minutes. To detect the involvement of neurotransmitter systems in this immediate effect, the NMDA antagonist MK-801 (0.8 mg/kg); as well as an agonist, quinpirole (QP, 5 mg/kg) and an antagonist, sulpiride (SP, 80 mg/kg) of the dopamine D2 receptors, were given before 3-NP to separate groups of rats. Controls were given saline. All substances were injected ip. 3-NP decreased the rats' locomotor, especially vertical, activity, whereas local activity was increased. Based on the further changes of 3-NP effects in the combination groups it could be concluded that dopaminergic rather than glutamatergic mechanisms were possibly involved in the acute behavioral effect of 3-NP.
Collapse
Affiliation(s)
- Anita Lukács
- University of Szeged Department of Public Health Szeged Hungary
| | | | | | | |
Collapse
|
36
|
Gubellini P, Picconi B, Di Filippo M, Calabresi P. Downstream mechanisms triggered by mitochondrial dysfunction in the basal ganglia: from experimental models to neurodegenerative diseases. Biochim Biophys Acta Mol Basis Dis 2009; 1802:151-61. [PMID: 19683569 DOI: 10.1016/j.bbadis.2009.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2009] [Revised: 07/22/2009] [Accepted: 08/06/2009] [Indexed: 12/21/2022]
Abstract
Mitochondrial dysfunctions have been implicated in the cellular processes underlying several neurodegenerative disorders affecting the basal ganglia. These include Huntington's chorea and Parkinson's disease, two highly debilitating motor disorders for which recent research has also involved gene mutation linked to mitochondrial deficits. Experimental models of basal ganglia diseases have been developed by using toxins able to disrupt mitochondrial function: these molecules act by selectively inhibiting mitochondrial respiratory complexes, uncoupling cellular respiration. This in turn leads to oxidative stress and energy deficit that trigger critical downstream mechanisms, ultimately resulting in neuronal vulnerability and loss. Here we review the molecular and cellular downstream effects triggered by mitochondrial dysfunction, and the different experimental models that are obtained by the administration of selective mitochondrial toxins or by the expression of mutant genes.
Collapse
Affiliation(s)
- Paolo Gubellini
- Institut de Biologie du Développement de Marseille-Luminy (IBDML), UMR6216 (CNRS/Université de la Méditerranée), Marseille, France.
| | | | | | | |
Collapse
|
37
|
Trifilieff P, Lavaur J, Pascoli V, Kappès V, Brami-Cherrier K, Pagès C, Micheau J, Caboche J, Vanhoutte P. Endocytosis controls glutamate-induced nuclear accumulation of ERK. Mol Cell Neurosci 2009; 41:325-36. [DOI: 10.1016/j.mcn.2009.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 04/17/2009] [Accepted: 04/20/2009] [Indexed: 10/20/2022] Open
|
38
|
Arumugam TV, Woodruff TM, Lathia JD, Selvaraj PK, Mattson MP, Taylor SM. Neuroprotection in stroke by complement inhibition and immunoglobulin therapy. Neuroscience 2009; 158:1074-89. [PMID: 18691639 PMCID: PMC2639633 DOI: 10.1016/j.neuroscience.2008.07.015] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 07/08/2008] [Accepted: 07/08/2008] [Indexed: 12/18/2022]
Abstract
Activation of the complement system occurs in a variety of neuroinflammatory diseases and neurodegenerative processes of the CNS. Studies in the last decade have demonstrated that essentially all of the activation components and receptors of the complement system are produced by astrocytes, microglia, and neurons. There is also rapidly growing evidence to indicate an active role of the complement system in cerebral ischemic injury. In addition to direct cell damage, regional cerebral ischemia and reperfusion (I/R) induces an inflammatory response involving complement activation and generation of active fragments, such as C3a and C5a anaphylatoxins, C3b, C4b, and iC3b. The use of specific inhibitors to block complement activation or their mediators such as C5a, can reduce local tissue injury after I/R. Consistent with therapeutic approaches that have been successful in models of autoimmune disorders, many of the same complement inhibition strategies are proving effective in animal models of cerebral I/R injury. One new form of therapy, which is less specific in its targeting of complement than monodrug administration, is the use of immunoglobulins. Intravenous immunoglobulin (IVIG) has the potential to inhibit multiple components of inflammation, including complement fragments, pro-inflammatory cytokine production and leukocyte cell adhesion. Thus, IVIG may directly protect neurons, reduce activation of intrinsic inflammatory cells (microglia) and inhibit transendothelial infiltration of leukocytes into the brain parenchyma following an ischemic stroke. The striking neuroprotective actions of IVIG in animal models of ischemic stroke suggest a potential therapeutic potential that merits consideration for clinical trials in stroke patients.
Collapse
Affiliation(s)
- T V Arumugam
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 Coulter Drive, Amarillo, TX 79106, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Levin-Salomon V, Kogan K, Ahn NG, Livnah O, Engelberg D. Isolation of intrinsically active (MEK-independent) variants of the ERK family of mitogen-activated protein (MAP) kinases. J Biol Chem 2008; 283:34500-10. [PMID: 18829462 PMCID: PMC3259889 DOI: 10.1074/jbc.m806443200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 09/29/2008] [Indexed: 11/06/2022] Open
Abstract
MAPKs are key components of cell signaling pathways with a unique activation mechanism: i.e. dual phosphorylation of neighboring threonine and tyrosine residues. The ERK enzymes form a subfamily of MAPKs involved in proliferation, differentiation, development, learning, and memory. The exact role of each Erk molecule in these processes is not clear. An efficient strategy for addressing this question is to activate individually each molecule, for example, by expressing intrinsically active variants of them. However, such molecules were not produced so far. Here, we report on the isolation, via a specifically designed genetic screen, of six variants (each carries a point mutation) of the yeast MAPK Mpk1/Erk that are active, independent of upstream phosphorylation. One of the activating mutations, R68S, occurred in a residue conserved in the mammalian Erk1 (Arg-84) and Erk2 (Arg-65) and in the Drosophila ERK Rolled (Arg-80). Replacing this conserved Arg with Ser rendered these MAPKs intrinsically active to very high levels when tested in vitro as recombinant proteins. Combination of the Arg to Ser mutation with the sevenmaker mutation (producing Erk2(R65S+D319N) and Rolled(R80S+D334N)) resulted in even higher activity (45 and 70%, respectively, in reference to fully active dually phosphorylated Erk2 or Rolled). Erk2(R65S) and Erk2(R65S+D319N) were found to be spontaneously active also when expressed in human HEK293 cells. We further revealed the mechanism of action of the mutants and show that it involves acquisition of autophosphorylation activity. Thus, a first generation of Erk molecules that are spontaneously active in vitro and in vivo has been obtained.
Collapse
Affiliation(s)
- Vered Levin-Salomon
- The Department of Biological Chemistry,
The Alexander Silberman Institute of Life Sciences, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel, The Wolfson
Centre for Applied Structural Biology, The Hebrew University of Jerusalem,
Jerusalem 91904, Israel, and the Department of
Chemistry and Biochemistry, Howard Hughes Medical Institute, University of
Colorado, Boulder, Colorado 80309
| | - Konstantin Kogan
- The Department of Biological Chemistry,
The Alexander Silberman Institute of Life Sciences, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel, The Wolfson
Centre for Applied Structural Biology, The Hebrew University of Jerusalem,
Jerusalem 91904, Israel, and the Department of
Chemistry and Biochemistry, Howard Hughes Medical Institute, University of
Colorado, Boulder, Colorado 80309
| | - Natalie G. Ahn
- The Department of Biological Chemistry,
The Alexander Silberman Institute of Life Sciences, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel, The Wolfson
Centre for Applied Structural Biology, The Hebrew University of Jerusalem,
Jerusalem 91904, Israel, and the Department of
Chemistry and Biochemistry, Howard Hughes Medical Institute, University of
Colorado, Boulder, Colorado 80309
| | - Oded Livnah
- The Department of Biological Chemistry,
The Alexander Silberman Institute of Life Sciences, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel, The Wolfson
Centre for Applied Structural Biology, The Hebrew University of Jerusalem,
Jerusalem 91904, Israel, and the Department of
Chemistry and Biochemistry, Howard Hughes Medical Institute, University of
Colorado, Boulder, Colorado 80309
| | - David Engelberg
- The Department of Biological Chemistry,
The Alexander Silberman Institute of Life Sciences, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel, The Wolfson
Centre for Applied Structural Biology, The Hebrew University of Jerusalem,
Jerusalem 91904, Israel, and the Department of
Chemistry and Biochemistry, Howard Hughes Medical Institute, University of
Colorado, Boulder, Colorado 80309
| |
Collapse
|
40
|
Roze E, Betuing S, Deyts C, Vidailhet M, Caboche J. Physiopathologie de la maladie de Huntington : état des connaissances. Rev Neurol (Paris) 2008; 164:977-94. [DOI: 10.1016/j.neurol.2008.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 01/28/2008] [Accepted: 03/26/2008] [Indexed: 12/16/2022]
|
41
|
Perrin V, Dufour N, Raoul C, Hassig R, Brouillet E, Aebischer P, Luthi-Carter R, Déglon N. Implication of the JNK pathway in a rat model of Huntington's disease. Exp Neurol 2008; 215:191-200. [PMID: 19022249 DOI: 10.1016/j.expneurol.2008.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 09/18/2008] [Accepted: 10/16/2008] [Indexed: 11/28/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder resulting from the expansion of a glutamine repeat (polyQ) in the N-terminus of the huntingtin (htt) protein. Expression of polyQ-containing proteins has been previously shown to induce various cellular stress responses. Among these, activation of the c-Jun N-terminal kinase (JNK) cascade has been observed in cellular models of HD. However, the implication of the JNK pathway has not previously been evaluated in the striatum of HD animal models. Here we report that the JNK pathway participates in HD pathology in a rat model of the disease. Increased phosphorylation of the JNK target c-Jun was observed as early as 4 weeks and persisted for 13 weeks after lentiviral-mediated expression of htt171-82Q. In order to assess the importance of this pathway in HD pathology, JNK inhibitors including dominant-negative mutants of upstream kinases (ASK1(K709R), MEKK1(D1369A)), a c-Jun mutant (Delta169c-Jun) and the active domain of the scaffold protein JIP-1/IBI (IBI-JBD) were tested for their ability to mitigate the effect of htt171-82Q. The overexpression of MEKK1(D1369A) and JIP-1/IBI reduced the polyQ-related loss of DARPP-32 expression, while the other inhibitors had no effect. In all cases, the formation of EM48-positive htt inclusions and P-c-Jun immunoreactivity were unaltered. These results suggest that JNK activation is involved in HD and that blockade of this pathway may be of benefit in counteracting HD-related neurotoxicity.
Collapse
Affiliation(s)
- V Perrin
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Neuropsychological and neuroimaging studies in humans have shown that the prefrontal cortex (PFC) is involved in long-term memory functioning. In general, the participation of the PFC in long-term memory has been attributed to its role in executive control rather than information storage. Accumulating data from recent animal studies, however, suggest the possible role of the PFC in the storage of long-term memory. In support of this view, there is evidence that various projection systems in the PFC support long-term synaptic plasticity. Recording studies have further demonstrated neural correlates of learning in various animal species. Lastly, behavioral and physiological studies indicate that the PFC is critically involved in memory consolidation, retrieval and extinction processes. These studies then suggest that the PFC is an integral part of the neural network where long-term memory trace is stored and retrieved. Though decisive evidence is still lacking at present, we propose here to assign a term 'control memory' (i.e., memory for top-down control processes) as a new type of memory function for the PFC. This new principle of PFC-long-term memory can help organize existing data and provide novel insights into future empirical studies.
Collapse
Affiliation(s)
- Min Whan Jung
- Neuroscience Laboratory, Institute for Medical Sciences, Ajou University School of Medicine, Suwon, Korea.
| | | | | | | | | |
Collapse
|
43
|
Lee ST, Chu K, Park JE, Hong NH, Im WS, Kang L, Han Z, Jung KH, Kim MW, Kim M. Atorvastatin attenuates mitochondrial toxin-induced striatal degeneration, with decreasing iNOS/c-Jun levels and activating ERK/Akt pathways. J Neurochem 2008; 104:1190-200. [DOI: 10.1111/j.1471-4159.2007.05044.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
44
|
Akashiba H, Ikegaya Y, Nishiyama N, Matsuki N. Differential Involvement of Cell Cycle Reactivation between Striatal and Cortical Neurons in Cell Death Induced by 3-Nitropropionic Acid. J Biol Chem 2008; 283:6594-606. [DOI: 10.1074/jbc.m707730200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
45
|
Granulocyte-colony stimulating factor attenuates striatal degeneration with activating survival pathways in 3-nitropropionic acid model of Huntington's disease. Brain Res 2008; 1194:130-7. [DOI: 10.1016/j.brainres.2007.11.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 11/24/2007] [Accepted: 11/27/2007] [Indexed: 12/11/2022]
|
46
|
Induction of apoptosis signal-regulating kinase 1 and oxidative stress mediate age-dependent vulnerability to 3-nitropropionic acid in the mouse striatum. Neurosci Lett 2008; 430:142-6. [DOI: 10.1016/j.neulet.2007.10.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 10/04/2007] [Accepted: 10/25/2007] [Indexed: 01/26/2023]
|
47
|
Neural Stem/Progenitor Cells Initiate the Formation of Cellular Networks That Provide Neuroprotection by Growth Factor-Modulated Antioxidant Expression. Stem Cells 2008; 26:254-65. [DOI: 10.1634/stemcells.2007-0221] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
Saudou F, Humbert S. The biology of Huntington's disease. HANDBOOK OF CLINICAL NEUROLOGY 2008; 89:619-29. [DOI: 10.1016/s0072-9752(07)01257-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
49
|
Roze E, Bettuing S, Deyts C, Marcon E, Brami‐Cherrier K, Pagès C, Humbert S, Mérienne K, Caboche J. Mitogen‐ and stress‐activated protein kinase‐1 deficiency is involved in expanded‐huntingtin‐induced transcriptional dysregulation and striatal death. FASEB J 2007; 22:1083-93. [DOI: 10.1096/fj.07-9814] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Emmanuel Roze
- Université Pierre et Marie Curie‐Paris 6, CNRS, UMR 7102ParisFrance
- Service de NeurologieHôpital Saint‐AntoineAssitance Publique‐Hôpitaux de ParisParisFrance
| | | | - Carole Deyts
- Université Pierre et Marie Curie‐Paris 6, CNRS, UMR 7102ParisFrance
| | - Estelle Marcon
- Université Pierre et Marie Curie‐Paris 6, CNRS, UMR 7102ParisFrance
| | | | - Christiane Pagès
- Université Pierre et Marie Curie‐Paris 6, CNRS, UMR 7102ParisFrance
| | | | - Karine Mérienne
- Institut de Génétique et de Biologie Moléculaire et CellulaireDépartement de pathologie moléculaire; INSERM, U596; CNRS, UMR 7104IllkirchFrance
| | - Jocelyne Caboche
- Université Pierre et Marie Curie‐Paris 6, CNRS, UMR 7102ParisFrance
| |
Collapse
|
50
|
Abstract
The pathomechanisms involved in the neuronal dysfunction in Huntington disease (HD) are still unresolved and may be heterogeneous. One potential mechanism might be related to the induction of mitochondrial dysfunction in the CNS. This might lead firstly to neuronal dysfunction and finally to the activation of apoptotic pathways. Several compounds, which should alleviate mitochondrial dysfunction, have been tested in preclinical models as well as in clinical trials of different scale. Recently we reported the efficacy of Ethyl-eicosapentaenoic acid (Ethyl-EPA) in patients with HD. Ethyl-EPA is a polyunsaturated fatty acid from the n-3 group, which is in clinical development for HD and melancholic depression. In our trial with Ethyl-EPA in HD responding patients could be characterized by either a lower CAG repeat number or a chorea-predominant clinical expression of the disease. Here we would like to describe some evidence on the potential mechanism of action of Ethyl-EPA in HD. We specifically focus on pathways, which are known to be influenced in HD and are modified by Ethyl-EPA and which points to an involvement of mitochondrial function as a common target. Some attention is given to the NF-kappa B pathway and the c-Jun amino-terminal kinases (JNK) pathway, which both may lead to an activation of the antiproliferative factor p53 and consequently mitochondrial dysfunction. Further the effects of EPA or Ethyl-EPA in preclinical models of HD are described. The evidence from these studies led to the design of phase III clinical trials, which are ongoing.
Collapse
Affiliation(s)
- Harald Murck
- Amarin Neuroscience Ltd, Laurelhill Business Park, Stirling, UK.
| | | |
Collapse
|