1
|
Wang Y, Li H, He Q, Zou R, Cai J, Zhang L. Ferroptosis: underlying mechanisms and involvement in neurodegenerative diseases. Apoptosis 2024; 29:3-21. [PMID: 37848673 DOI: 10.1007/s10495-023-01902-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2023] [Indexed: 10/19/2023]
Abstract
Ferroptosis, a mode of cell death that was recently identified in 2012, is driven by iron-dependent lipid peroxidation and distinct from other mechanisms of cell death such as autophagy and apoptosis. Ferroptosis has the unique features of disruptions in iron equilibrium, iron-induced lipid peroxidation, and the accumulation of glutamate-induced cellular toxicity. The regulation of ferroptosis mainly involves the iron, lipid, and amino acid metabolic pathways, which are controlled by system Xc-, voltage-dependent anion channels, p53 and other pathways. Neurodegenerative diseases involve gradual neuronal loss predominantly within the central nervous system and are categorized into both sporadic and rare hereditary disorders. These diseases result in the progressive decline of specific neuron populations and their interconnections. Recent investigations have revealed a strong correlation between the manifestation and progression of neurodegenerative diseases and ferroptosis. The pharmacological modulation of ferroptosis, whether by induction or inhibition, exhibits promising prospects for therapeutic interventions for these diseases. This review aims to examine the literature on ferroptosis and its implications in various neurodegenerative diseases. We hope to offer novel insights into the potential therapies targeting ferroptosis in central nervous system neurodegenerative diseases. However, there are still limitations of this review. First, despite our efforts to maintain objectivity during our analysis, this review does not cover all the studies on ferroptosis and neurodegenerative diseases. Second, cell death in neurodegenerative diseases is not solely caused by ferroptosis. Future research should focus on the interplay of different cell death mechanisms to better elucidate the specific disease pathogenesis.
Collapse
Affiliation(s)
- Yi Wang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - HongJing Li
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - QianXiong He
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rong Zou
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - JinRui Cai
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Lin Zhang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.
- Key Laboratory of Tibetan Medicine Research, Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Northwest Institute of Plateau Biology, Xining, 810008, Qinghai, China.
| |
Collapse
|
2
|
Nguyen TM, Ngoc DTM, Choi JH, Lee CH. Unveiling the Neural Environment in Cancer: Exploring the Role of Neural Circuit Players and Potential Therapeutic Strategies. Cells 2023; 12:1996. [PMID: 37566075 PMCID: PMC10417274 DOI: 10.3390/cells12151996] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
The regulation of the immune environment within the tumor microenvironment has provided new opportunities for cancer treatment. However, an important microenvironment surrounding cancer that is often overlooked despite its significance in cancer progression is the neural environment surrounding the tumor. The release of neurotrophic factors from cancer cells is implicated in cancer growth and metastasis by facilitating the infiltration of nerve cells into the tumor microenvironment. This nerve-tumor interplay can elicit cancer cell proliferation, migration, and invasion in response to neurotransmitters. Moreover, it is possible that cancer cells could establish a network resembling that of neurons, allowing them to communicate with one another through neurotransmitters. The expression levels of players in the neural circuits of cancers could serve as potential biomarkers for cancer aggressiveness. Notably, the upregulation of certain players in the neural circuit has been linked to poor prognosis in specific cancer types such as breast cancer, pancreatic cancer, basal cell carcinoma, and stomach cancer. Targeting these players with inhibitors holds great potential for reducing the morbidity and mortality of these carcinomas. However, the efficacy of anti-neurogenic agents in cancer therapy remains underexplored, and further research is necessary to evaluate their effectiveness as a novel approach for cancer treatment. This review summarizes the current knowledge on the role of players in the neural circuits of cancers and the potential of anti-neurogenic agents for cancer therapy.
Collapse
Affiliation(s)
- Tuan Minh Nguyen
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Dinh Thi Minh Ngoc
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Jung-Hye Choi
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Chang-Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| |
Collapse
|
3
|
Tucci P, Lattanzi R, Severini C, Saso L. Nrf2 Pathway in Huntington's Disease (HD): What Is Its Role? Int J Mol Sci 2022; 23:ijms232315272. [PMID: 36499596 PMCID: PMC9739588 DOI: 10.3390/ijms232315272] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease that occurs worldwide. Despite some progress in understanding the onset of HD, drugs that block or delay symptoms are still not available. In recent years, many treatments have been proposed; among them, nuclear transcriptional factor-2 (Nrf2) enhancer compounds have been proposed as potential therapeutic agents to treat HD. Nrf2 triggers an endogenous antioxidant pathway activated in different neurodegenerative disorders. Probably, the stimulation of Nrf2 during either the early phase or before HD symptoms' onset, could slow or prevent striatum degeneration. In this review, we present the scientific literature supporting the role of Nrf2 in HD and the potential prophylactic and therapeutic role of this compound.
Collapse
Affiliation(s)
- Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Cinzia Severini
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Viale del Policlinico 155, 00161 Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
4
|
Kacher R, Mounier C, Caboche J, Betuing S. Altered Cholesterol Homeostasis in Huntington’s Disease. Front Aging Neurosci 2022; 14:797220. [PMID: 35517051 PMCID: PMC9063567 DOI: 10.3389/fnagi.2022.797220] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
Huntington’s disease (HD) is an autosomal dominant genetic disorder caused by an expansion of the CAG repeat in the first exon of Huntingtin’s gene. The associated neurodegeneration mainly affects the striatum and the cortex at early stages and progressively spreads to other brain structures. Targeting HD at its earlier stages is under intense investigation. Numerous drugs were tested, with a rate of success of only 3.5% approved molecules used as symptomatic treatment. The restoration of cholesterol metabolism, which is central to the brain homeostasis and strongly altered in HD, could be an interesting disease-modifying strategy. Cholesterol is an essential membrane component in the central nervous system (CNS); alterations of its homeostasis have deleterious consequences on neuronal functions. The levels of several sterols, upstream of cholesterol, are markedly decreased within the striatum of HD mouse model. Transcription of cholesterol biosynthetic genes is reduced in HD cell and mouse models as well as post-mortem striatal and cortical tissues from HD patients. Since the dynamic of brain cholesterol metabolism is complex, it is essential to establish the best method to target it in HD. Cholesterol, which does not cross the blood-brain-barrier, is locally synthesized and renewed within the brain. All cell types in the CNS synthesize cholesterol during development but as they progress through adulthood, neurons down-regulate their cholesterol synthesis and turn to astrocytes for their full supply. Cellular levels of cholesterol reflect the dynamic balance between synthesis, uptake and export, all integrated into the context of the cross talk between neurons and glial cells. In this review, we describe the latest advances regarding the role of cholesterol deregulation in neuronal functions and how this could be a determinant factor in neuronal degeneration and HD progression. The pathways and major mechanisms by which cholesterol and sterols are regulated in the CNS will be described. From this overview, we discuss the main clinical strategies for manipulating cholesterol metabolism in the CNS, and how to reinstate a proper balance in HD.
Collapse
Affiliation(s)
- Radhia Kacher
- Institut du Cerveau - Paris Brain Institute (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- INSERM, U1216, Grenoble Institut Neurosciences, Université Grenoble Alpes, Grenoble, France
| | - Coline Mounier
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
- Centre National de la Recherche Scientifique, UMR 8246, Paris, France
- U1130, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
- Centre National de la Recherche Scientifique, UMR 8246, Paris, France
- U1130, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Sandrine Betuing
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
- Centre National de la Recherche Scientifique, UMR 8246, Paris, France
- U1130, Institut National de la Santé et de la Recherche Médicale, Paris, France
- *Correspondence: Sandrine Betuing,
| |
Collapse
|
5
|
Jannath KA, Akhtar MH, Gurudatt NG, Park DS, Kim KB, Shim YB. Catalytic SrMoO 4 nanoparticles and conducting polymer composite sensor for monitoring of K +-induced dopamine release from neuronal cells. J Mater Chem B 2022; 10:728-736. [PMID: 35019925 DOI: 10.1039/d1tb02295d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Octahedral SrMoO4 nanoparticles (NPs) with a high degree of crystallinity and controlled size (250-350 nm) were synthesized for the first time by employing a facile hydrothermal method. The prepared NPs were composited with a carboxyl group bearing conducting polymer (2,2:5,2-terthiophene-3-(p-benzoic acid, TBA)) to attain a stable sensor probe (pTBA/SrMoO4) which was analyzed using various surface analysis methods. The catalytic performance of the composite electrode was explored as an oxidation catalyst for biological molecules through anchoring on the conducting polymer layer, which functioned as a matrix to enhance the stability and selectivity of the sensor probe. The pTBA/SrMoO4 coated on glassy carbon displayed excellent electrocatalytic performance for the oxidation of some biologically important molecules, including dopamine (DA) in neuronal cells. The sensor immobilized with the catalyst showed an excellent response for DA with a dynamic range between 0.2 and 500 μM and a detection limit of 5 nM. The proposed sensor demonstrates the detection of trace DA released from PC12 cells under K+ stimulation, followed by inhibition of the release of exogenic DA by a Ca2+ channel blocker (nifedipine). The developed method provides an interesting way to monitor the effect of extracellular substances on living cells and the drug potency test.
Collapse
Affiliation(s)
- Khatun A Jannath
- Institute of Biophysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea
| | - Mahmood Hassan Akhtar
- Institute of Biophysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea.,Department of Chemistry, Pusan National University, Busan, 46241, Republic of Korea.
| | - N G Gurudatt
- Institute of Biophysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea.,Department of Chemistry, Pusan National University, Busan, 46241, Republic of Korea.
| | - Deog-Su Park
- Institute of Biophysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea
| | - Kwang Bok Kim
- Digital Health Care R&D Department, Korea Institute of Industrial Technology (KITECH), Cheonan, 31056, Republic of Korea.
| | - Yoon-Bo Shim
- Institute of Biophysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea.,Department of Chemistry, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
6
|
Harris KL, Mason SL, Vallin B, Barker RA. Reduced expression of dopamine D2 receptors on astrocytes in R6/1 HD mice and HD post-mortem tissue. Neurosci Lett 2022; 767:136289. [PMID: 34637857 PMCID: PMC9188264 DOI: 10.1016/j.neulet.2021.136289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022]
Abstract
Dysfunction of the central dopaminergic system is thought to contribute to some of the clinical features of Huntington's disease (HD), and dopamine (DA) receptor antagonists are commonly used to good effect in its treatment. It is well established that there is an early significant reduction in neuronal D2 receptors in HD, considered to be a compensatory response to increased dopaminergic activity. However, no studies have examined the expression of D2 receptors on astrocytes which is important given that these cells have been shown to play a role in the pathogenesis of HD, as well as express dopamine receptors and modulate DA homeostasis in the normal brain. We therefore sought to investigate the expression of D2 receptors on astrocytes in HD, and found them to be reduced in both the R6/1 HD mouse model, and in human post-mortem brain in comparison to controls, suggesting that astrocytes may be important in DA-dependent aspects of HD. Further studies are needed to determine the functional significance of this finding.
Collapse
Affiliation(s)
- Kate L Harris
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK.
| | - Sarah L Mason
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Benjamin Vallin
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK; MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Kim A, Lalonde K, Truesdell A, Gomes Welter P, Brocardo PS, Rosenstock TR, Gil-Mohapel J. New Avenues for the Treatment of Huntington's Disease. Int J Mol Sci 2021; 22:ijms22168363. [PMID: 34445070 PMCID: PMC8394361 DOI: 10.3390/ijms22168363] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HD gene. The disease is characterized by neurodegeneration, particularly in the striatum and cortex. The first symptoms usually appear in mid-life and include cognitive deficits and motor disturbances that progress over time. Despite being a genetic disorder with a known cause, several mechanisms are thought to contribute to neurodegeneration in HD, and numerous pre-clinical and clinical studies have been conducted and are currently underway to test the efficacy of therapeutic approaches targeting some of these mechanisms with varying degrees of success. Although current clinical trials may lead to the identification or refinement of treatments that are likely to improve the quality of life of those living with HD, major efforts continue to be invested at the pre-clinical level, with numerous studies testing novel approaches that show promise as disease-modifying strategies. This review offers a detailed overview of the currently approved treatment options for HD and the clinical trials for this neurodegenerative disorder that are underway and concludes by discussing potential disease-modifying treatments that have shown promise in pre-clinical studies, including increasing neurotropic support, modulating autophagy, epigenetic and genetic manipulations, and the use of nanocarriers and stem cells.
Collapse
Affiliation(s)
- Amy Kim
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Kathryn Lalonde
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Aaron Truesdell
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Priscilla Gomes Welter
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Patricia S. Brocardo
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Tatiana R. Rosenstock
- Institute of Cancer and Genomic Science, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- Department of Pharmacology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Joana Gil-Mohapel
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Correspondence: ; Tel.: +1-250-472-4597; Fax: +1-250-472-5505
| |
Collapse
|
8
|
van der Weijden MC, Rodriguez‐Contreras D, Delnooz CC, Robinson BG, Condon AF, Kielhold ML, Stormezand GN, Ma KY, Dufke C, Williams JT, Neve KA, Tijssen MA, Verbeek DS. A Gain-of-Function Variant in Dopamine D2 Receptor and Progressive Chorea and Dystonia Phenotype. Mov Disord 2021; 36:729-739. [PMID: 33200438 PMCID: PMC8049080 DOI: 10.1002/mds.28385] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/03/2020] [Accepted: 10/26/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND We describe a 4-generation Dutch pedigree with a unique dominantly inherited clinical phenotype of a combined progressive chorea and cervical dystonia carrying a novel heterozygous dopamine D2 receptor (DRD2) variant. OBJECTIVES The objective of this study was to identify the genetic cause of the disease and to further investigate the functional consequences of the genetic variant. METHODS After detailed clinical and neurological examination, whole-exome sequencing was performed. Because a novel variant in the DRD2 gene was found as the likely causative gene defect in our pedigree, we sequenced the DRD2 gene in a cohort of 121 Huntington-like cases with unknown genetic cause (Germany). Moreover, functional characterization of the DRD2 variant included arrestin recruitment, G protein activation, and G protein-mediated inhibition of adenylyl cyclase determined in a cell model, and G protein-regulated inward-rectifying potassium channels measured in midbrain slices of mice. RESULT We identified a novel heterozygous variant c.634A > T, p.Ile212Phe in exon 5 of DRD2 that cosegregated with the clinical phenotype. Screening of the German cohort did not reveal additional putative disease-causing variants. We demonstrated that the D2S/L -I212 F receptor exhibited increased agonist potency and constitutive activation of G proteins in human embryonic kidney 239 cells as well as significantly reduced arrestin3 recruitment. We further showed that the D2S -I212 F receptor exhibited aberrant receptor function in mouse midbrain slices. CONCLUSIONS Our results support an association between the novel p.Ile212Phe variant in DRD2, its modified D2 receptor activity, and the hyperkinetic movement disorder reported in the 4-generation pedigree. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marlous C.M. van der Weijden
- Department of GeneticsUniversity Medical Center GroningenGroningenthe Netherlands
- Expertise Center Movement Disorders GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | | | | | | | - Alec F. Condon
- Vollum InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Michelle L. Kielhold
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandOregonUSA
| | - Gilles N. Stormezand
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center GroningenGroningenthe Netherlands
| | - Kai Yu Ma
- Department of GeneticsUniversity Medical Center GroningenGroningenthe Netherlands
| | - Claudia Dufke
- Institute of Medical Genetics and Applied GenomicsUniversity Hospital TuebingenTuebingenGermany
| | - John T. Williams
- Vollum InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Kim A. Neve
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandOregonUSA
- Research ServiceVirginia Portland Health Care SystemPortlandOregonUSA
| | - Marina A.J. Tijssen
- Expertise Center Movement Disorders GroningenUniversity Medical Center GroningenGroningenthe Netherlands
- Department of NeurologyUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Dineke S. Verbeek
- Department of GeneticsUniversity Medical Center GroningenGroningenthe Netherlands
- Expertise Center Movement Disorders GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| |
Collapse
|
9
|
Bergonzoni G, Döring J, Biagioli M. D1R- and D2R-Medium-Sized Spiny Neurons Diversity: Insights Into Striatal Vulnerability to Huntington's Disease Mutation. Front Cell Neurosci 2021; 15:628010. [PMID: 33642998 PMCID: PMC7902492 DOI: 10.3389/fncel.2021.628010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder caused by an aberrant expansion of the CAG tract within the exon 1 of the HD gene, HTT. HD progressively impairs motor and cognitive capabilities, leading to a total loss of autonomy and ultimate death. Currently, no cure or effective treatment is available to halt the disease. Although the HTT gene is ubiquitously expressed, the striatum appears to be the most susceptible district to the HD mutation with Medium-sized Spiny Neurons (MSNs) (D1R and D2R) representing 95% of the striatal neuronal population. Why are striatal MSNs so vulnerable to the HD mutation? Particularly, why do D1R- and D2R-MSNs display different susceptibility to HD? Here, we highlight significant differences between D1R- and D2R-MSNs subpopulations, such as morphology, electrophysiology, transcriptomic, functionality, and localization in the striatum. We discuss possible reasons for their selective degeneration in the context of HD. Our review suggests that a better understanding of cell type-specific gene expression dysregulation within the striatum might reveal new paths to therapeutic intervention or prevention to ameliorate HD patients' life expectancy.
Collapse
Affiliation(s)
| | | | - Marta Biagioli
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| |
Collapse
|
10
|
The Role of the Renal Dopaminergic System and Oxidative Stress in the Pathogenesis of Hypertension. Biomedicines 2021; 9:biomedicines9020139. [PMID: 33535566 PMCID: PMC7912729 DOI: 10.3390/biomedicines9020139] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 01/11/2023] Open
Abstract
The kidney is critical in the long-term regulation of blood pressure. Oxidative stress is one of the many factors that is accountable for the development of hypertension. The five dopamine receptor subtypes (D1R–D5R) have important roles in the regulation of blood pressure through several mechanisms, such as inhibition of oxidative stress. Dopamine receptors, including those expressed in the kidney, reduce oxidative stress by inhibiting the expression or action of receptors that increase oxidative stress. In addition, dopamine receptors stimulate the expression or action of receptors that decrease oxidative stress. This article examines the importance and relationship between the renal dopaminergic system and oxidative stress in the regulation of renal sodium handling and blood pressure. It discusses the current information on renal dopamine receptor-mediated antioxidative network, which includes the production of reactive oxygen species and abnormalities of renal dopamine receptors. Recognizing the mechanisms by which renal dopamine receptors regulate oxidative stress and their degree of influence on the pathogenesis of hypertension would further advance the understanding of the pathophysiology of hypertension.
Collapse
|
11
|
Azam S, Haque ME, Jakaria M, Jo SH, Kim IS, Choi DK. G-Protein-Coupled Receptors in CNS: A Potential Therapeutic Target for Intervention in Neurodegenerative Disorders and Associated Cognitive Deficits. Cells 2020; 9:cells9020506. [PMID: 32102186 PMCID: PMC7072884 DOI: 10.3390/cells9020506] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
Neurodegenerative diseases are a large group of neurological disorders with diverse etiological and pathological phenomena. However, current therapeutics rely mostly on symptomatic relief while failing to target the underlying disease pathobiology. G-protein-coupled receptors (GPCRs) are one of the most frequently targeted receptors for developing novel therapeutics for central nervous system (CNS) disorders. Many currently available antipsychotic therapeutics also act as either antagonists or agonists of different GPCRs. Therefore, GPCR-based drug development is spreading widely to regulate neurodegeneration and associated cognitive deficits through the modulation of canonical and noncanonical signals. Here, GPCRs’ role in the pathophysiology of different neurodegenerative disease progressions and cognitive deficits has been highlighted, and an emphasis has been placed on the current pharmacological developments with GPCRs to provide an insight into a potential therapeutic target in the treatment of neurodegeneration.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
| | - Md. Ezazul Haque
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
| | - Md. Jakaria
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Song-Hee Jo
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
| | - In-Su Kim
- Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-010-3876-4773 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| | - Dong-Kug Choi
- Department of Applied Life Science & Integrated Bioscience, Graduate School, Konkuk University, Chungju 27478, Korea; (S.A.); (M.E.H.); (M.J.); (S.-H.J.)
- Department of Integrated Bioscience & Biotechnology, College of Biomedical and Health Science, and Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-010-3876-4773 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| |
Collapse
|
12
|
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
|
13
|
Tobore TO. Towards a comprehensive understanding of the contributions of mitochondrial dysfunction and oxidative stress in the pathogenesis and pathophysiology of Huntington's disease. J Neurosci Res 2019; 97:1455-1468. [DOI: 10.1002/jnr.24492] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 12/21/2022]
|
14
|
Koch ET, Raymond LA. Dysfunctional striatal dopamine signaling in Huntington's disease. J Neurosci Res 2019; 97:1636-1654. [PMID: 31304622 DOI: 10.1002/jnr.24495] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/06/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022]
Abstract
Dopamine signaling in the striatum is critical for a variety of behaviors including movement, behavioral flexibility, response to reward and many forms of learning. Alterations to dopamine transmission contribute to pathological features of many neurological diseases, including Huntington's disease (HD). HD is an autosomal dominant genetic disorder caused by a CAG repeat expansion in the Huntingtin gene. The striatum is preferentially degenerated in HD, and this region receives dopaminergic input from the substantia nigra. Studies of HD patients and genetic rodent models have shown changes to levels of dopamine and its receptors in the striatum, and alterations in dopamine receptor signaling and modulation of other neurotransmitters, notably glutamate. Throughout his career, Dr. Michael Levine's research has furthered our understanding of dopamine signaling in the striatum of healthy rodents and HD mouse models. This review will focus on the work of his group and others in elucidating alterations to striatal dopamine signaling that contribute to pathophysiology in HD mouse models, and how these findings relate to human HD studies. We will also discuss current and potential therapeutic interventions for HD that target the dopamine system, and future research directions for this field.
Collapse
Affiliation(s)
- Ellen T Koch
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Galvan L, Francelle L, Gaillard MC, de Longprez L, Carrillo-de Sauvage MA, Liot G, Cambon K, Stimmer L, Luccantoni S, Flament J, Valette J, de Chaldée M, Auregan G, Guillermier M, Joséphine C, Petit F, Jan C, Jarrige M, Dufour N, Bonvento G, Humbert S, Saudou F, Hantraye P, Merienne K, Bemelmans AP, Perrier AL, Déglon N, Brouillet E. The striatal kinase DCLK3 produces neuroprotection against mutant huntingtin. Brain 2019. [PMID: 29534157 PMCID: PMC5917821 DOI: 10.1093/brain/awy057] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The neurobiological functions of a number of kinases expressed in the brain are unknown. Here, we report new findings on DCLK3 (doublecortin like kinase 3), which is preferentially expressed in neurons in the striatum and dentate gyrus. Its function has never been investigated. DCLK3 expression is markedly reduced in Huntington's disease. Recent data obtained in studies related to cancer suggest DCLK3 could have an anti-apoptotic effect. Thus, we hypothesized that early loss of DCLK3 in Huntington's disease may render striatal neurons more susceptible to mutant huntingtin (mHtt). We discovered that DCLK3 silencing in the striatum of mice exacerbated the toxicity of an N-terminal fragment of mHtt. Conversely, overexpression of DCLK3 reduced neurodegeneration produced by mHtt. DCLK3 also produced beneficial effects on motor symptoms in a knock-in mouse model of Huntington's disease. Using different mutants of DCLK3, we found that the kinase activity of the protein plays a key role in neuroprotection. To investigate the potential mechanisms underlying DCLK3 effects, we studied the transcriptional changes produced by the kinase domain in human striatal neurons in culture. Results show that DCLK3 regulates in a kinase-dependent manner the expression of many genes involved in transcription regulation and nucleosome/chromatin remodelling. Consistent with this, histological evaluation showed DCLK3 is present in the nucleus of striatal neurons and, protein-protein interaction experiments suggested that the kinase domain interacts with zinc finger proteins, including the transcriptional activator adaptor TADA3, a core component of the Spt-ada-Gcn5 acetyltransferase (SAGA) complex which links histone acetylation to the transcription machinery. Our novel findings suggest that the presence of DCLK3 in striatal neurons may play a key role in transcription regulation and chromatin remodelling in these brain cells, and show that reduced expression of the kinase in Huntington's disease could render the striatum highly vulnerable to neurodegeneration.
Collapse
Affiliation(s)
- Laurie Galvan
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Laetitia Francelle
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Lucie de Longprez
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Maria-Angeles Carrillo-de Sauvage
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Géraldine Liot
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France.,Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F38000, Grenoble, France.,INSERM U1216, F38000, Grenoble, France.,CHU de Grenoble, F38000, Grenoble, France
| | - Karine Cambon
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Lev Stimmer
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,Inserm UMS27, F-92265 Fontenay-aux-Roses, France
| | - Sophie Luccantoni
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,Inserm UMS27, F-92265 Fontenay-aux-Roses, France
| | - Julien Flament
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,Inserm UMS27, F-92265 Fontenay-aux-Roses, France
| | - Julien Valette
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Michel de Chaldée
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Univ. Paris-Saclay, 91191 Gif-sur-Yvette Cedex, France
| | - Gwenaelle Auregan
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Martine Guillermier
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Charlène Joséphine
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Fanny Petit
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Caroline Jan
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Margot Jarrige
- Inserm U861, I-STEM, AFM, Evry 91030 Cedex France.,UEVE U861, I-STEM, AFM Evry 91030, France
| | - Noëlle Dufour
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Gilles Bonvento
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Sandrine Humbert
- Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F38000, Grenoble, France.,INSERM U1216, F38000, Grenoble, France.,CHU de Grenoble, F38000, Grenoble, France
| | - Frédéric Saudou
- Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F38000, Grenoble, France.,INSERM U1216, F38000, Grenoble, France.,CHU de Grenoble, F38000, Grenoble, France
| | - Philippe Hantraye
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Karine Merienne
- CNRS/Strasbourg University UMR 7364, Laboratory of Adaptive and Cognitive Neuroscience (LNCA), Strasbourg F-67000, France
| | - Alexis-Pierre Bemelmans
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| | - Anselme L Perrier
- Inserm U861, I-STEM, AFM, Evry 91030 Cedex France.,UEVE U861, I-STEM, AFM Evry 91030, France
| | - Nicole Déglon
- Lausanne University Medical School (CHUV), Department of Clinical Neurosciences (DNC), Laboratory of Cellular and Molecular Neurotherapies (LNCM), Lausanne, Switzerland.,Lausanne University Medical School (CHUV), Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies (LNCM), Lausanne, Switzerland
| | - Emmanuel Brouillet
- CEA, DRF, Institut François Jacob, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France.,CNRS, CEA, Paris-Sud Univ., Univ. Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), F-92265, Fontenay-aux-Roses, France
| |
Collapse
|
16
|
Ballaz SJ, Rebec GV. Neurobiology of vitamin C: Expanding the focus from antioxidant to endogenous neuromodulator. Pharmacol Res 2019; 146:104321. [PMID: 31229562 DOI: 10.1016/j.phrs.2019.104321] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 01/06/2023]
Abstract
Ascorbic acid (AA) is a water-soluble vitamin (C) found in all bodily organs. Most mammals synthesize it, humans are required to eat it, but all mammals need it for healthy functioning. AA reaches its highest concentration in the brain where both neurons and glia rely on tightly regulated uptake from blood via the glucose transport system and sodium-coupled active transport to accumulate and maintain AA at millimolar levels. As a prototype antioxidant, AA is not only neuroprotective, but also functions as a cofactor in redox-coupled reactions essential for the synthesis of neurotransmitters (e.g., dopamine and norepinephrine) and paracrine lipid mediators (e.g., epoxiecoisatrienoic acids) as well as the epigenetic regulation of DNA. Although redox capacity led to the promotion of AA in high doses as potential treatment for various neuropathological and psychiatric conditions, ample evidence has not supported this therapeutic strategy. Here, we focus on some long-neglected aspects of AA neurobiology, including its modulatory role in synaptic transmission as demonstrated by the long-established link between release of endogenous AA in brain extracellular fluid and the clearance of glutamate, an excitatory amino acid. Evidence that this link can be disrupted in animal models of Huntington´s disease is revealing opportunities for new research pathways and therapeutic applications (e.g., epilepsy and pain management). In fact, we suggest that improved understanding of the regulation of endogenous AA and its interaction with key brain neurotransmitter systems, rather than administration of AA in excess, should be the target of future brain-based therapies.
Collapse
Affiliation(s)
- Santiago J Ballaz
- School of Biological Sciences and Engineering, Yachay Tech University, Urcuqui, Ecuador.
| | - George V Rebec
- Program in Neuroscience, Department Psychological & Brain Sciences, Indiana University, Bloomington, USA.
| |
Collapse
|
17
|
Boussicault L, Kacher R, Lamazière A, Vanhoutte P, Caboche J, Betuing S, Potier MC. CYP46A1 protects against NMDA-mediated excitotoxicity in Huntington's disease: Analysis of lipid raft content. Biochimie 2018; 153:70-79. [DOI: 10.1016/j.biochi.2018.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
|
18
|
Chen S, Hu M, Shen M, Xu Y, Wang C, Wang X, Li F, Zeng D, Chen F, Zhao G, Chen M, Wang F, Cheng T, Su Y, Zhao J, Wang S, Wang J. Dopamine induces platelet production from megakaryocytes via oxidative stress-mediated signaling pathways. Platelets 2017; 29:702-708. [PMID: 29119850 DOI: 10.1080/09537104.2017.1356451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dopamine (DA), a catecholamine neurotransmitter, is known to for its diverse roles on hematopoiesis, yet its function in thrombopoiesis remains poorly understood. This study shows that DA stimulation can directly induce platelet production from megakaryocytes (MKs) in the final stages of thrombopoiesis via a reactive oxygen species (ROS)-dependent pathway. The mechanism was suggested by the results that DA treatment could significantly elevate the ROS levels in MKs, and time-dependently activate oxidative stress-mediated signaling, including p38 mitogen-activated protein kinase, c-Jun NH2-terminal kinase, and caspase-3 signaling pathways, while the antioxidants N-acetylcysteine and L-glutathione could effectively inhibit the activation of these signaling pathways, as well as the ROS increase and platelet production triggered by DA. Therefore, our data revealed that the direct role and mechanism of DA in thrombopoiesis, which provides new insights into the function recognition of DA in hematopoiesis.
Collapse
Affiliation(s)
- Shilei Chen
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Mengjia Hu
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Mingqiang Shen
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Yang Xu
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Cheng Wang
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Xinmiao Wang
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Fengju Li
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Dongfeng Zeng
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China.,c Department of Hematology, Daping Hospital , Third Military Medical University , Chongqing , China
| | - Fang Chen
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Gaomei Zhao
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Mo Chen
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Fengchao Wang
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Tianmin Cheng
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Yongping Su
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Jinghong Zhao
- b Department of Nephrology, Xinqiao Hospital , Third Military Medical University , Chongqing , China
| | - Song Wang
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| | - Junping Wang
- a State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine , Third Military Medical University , Chongqing , China
| |
Collapse
|
19
|
Nora GJ, Harun R, Fine DF, Hutchison D, Grobart AC, Stezoski JP, Munoz MJ, Kochanek PM, Leak RK, Drabek T, Wagner AK. Ventricular fibrillation cardiac arrest produces a chronic striatal hyperdopaminergic state that is worsened by methylphenidate treatment. J Neurochem 2017; 142:305-322. [PMID: 28445595 DOI: 10.1111/jnc.14058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/21/2023]
Abstract
Cardiac arrest survival rates have improved with modern resuscitation techniques, but many survivors experience impairments associated with hypoxic-ischemic brain injury (HIBI). Currently, little is understood about chronic changes in striatal dopamine (DA) systems after HIBI. Given the common empiric clinical use of DA enhancing agents in neurorehabilitation, investigation evaluating dopaminergic alterations after cardiac arrest (CA) is necessary to optimize rehabilitation approaches. We hypothesized that striatal DA neurotransmission would be altered chronically after ventricular fibrillation cardiac arrest (VF-CA). Fast-scan cyclic voltammetry was used with median forebrain bundle (MFB) maximal electrical stimulations (60Hz, 10s) in rats to characterize presynaptic components of DA neurotransmission in the dorsal striatum (D-Str) and nucleus accumbens 14 days after a 5-min VF-CA when compared to Sham or Naïve. VF-CA increased D-Str-evoked overflow [DA], total [DA] released, and initial DA release rate versus controls, despite also increasing maximal velocity of DA reuptake (Vmax ). Methylphenidate (10 mg/kg), a DA transporter inhibitor, was administered to VF-CA and Shams after establishing a baseline, pre-drug 60 Hz, 5 s stimulation response. Methylphenidate increased initial evoked overflow [DA] more-so in VF-CA versus Sham and reduced D-Str Vmax in VF-CA but not Shams; these findings are consistent with upregulated striatal DA transporter in VF-CA versus Sham. Our work demonstrates that 5-min VF-CA increases electrically stimulated DA release with concomitant upregulation of DA reuptake 2 weeks after brief VF-CA insult. Future work should elucidate how CA insult duration, time after insult, and insult type influence striatal DA neurotransmission and related cognitive and motor functions.
Collapse
Affiliation(s)
- Gerald J Nora
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rashed Harun
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David F Fine
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Daniel Hutchison
- Mylan School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Adam C Grobart
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason P Stezoski
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Miranda J Munoz
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rehana K Leak
- Mylan School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Tomas Drabek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Su P, Liu F. A peptide disrupting the D2R-DAT interaction protects against dopamine neurotoxicity. Exp Neurol 2017; 295:176-183. [PMID: 28579325 DOI: 10.1016/j.expneurol.2017.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/02/2017] [Accepted: 05/24/2017] [Indexed: 01/11/2023]
Abstract
Dopamine reuptake from extracellular space to cytosol leads to accumulation of dopamine, which triggers neurotoxicity in dopaminergic neurons. Previous studies have shown that both dopamine D2 receptor (D2R) and dopamine transporter (DAT) are involved in dopamine neurotoxicity. However, blockade of either D2R or DAT causes side effects due to antagonism of other physiological functions of these two proteins. We previously found that DAT can form a protein complex with D2R and its cell surface expression is facilitated via D2R-DAT interaction, which regulates dopamine reuptake and intracellular dopamine levels. Here we found that an interfering peptide (DAT-S1) disrupting the D2R-DAT interaction protects neurons against dopamine neurotoxicity, and this effect is mediated by inhibiting DAT cell surface expression and inhibiting both caspase-3 and PARP-1 cleavage. This study demonstrates the role of the D2R-DAT complex in dopamine neurotoxicity and investigated the potential mechanisms, which might help better understand the mechanisms of dopamine neurotoxicity. The peptide may provide some insights to improve treatments for dopamine neurotoxicity and related diseases, such as Parkinson's disease, as well as methamphetamine- and 3,4-methsylenedioxy methamphetamine-induced neurotoxicity.
Collapse
Affiliation(s)
- Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada.
| |
Collapse
|
21
|
Tyebji S, Hannan AJ. Synaptopathic mechanisms of neurodegeneration and dementia: Insights from Huntington's disease. Prog Neurobiol 2017; 153:18-45. [PMID: 28377290 DOI: 10.1016/j.pneurobio.2017.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 03/19/2017] [Accepted: 03/30/2017] [Indexed: 12/20/2022]
Abstract
Dementia encapsulates a set of symptoms that include loss of mental abilities such as memory, problem solving or language, and reduces a person's ability to perform daily activities. Alzheimer's disease is the most common form of dementia, however dementia can also occur in other neurological disorders such as Huntington's disease (HD). Many studies have demonstrated that loss of neuronal cell function manifests pre-symptomatically and thus is a relevant therapeutic target to alleviate symptoms. Synaptopathy, the physiological dysfunction of synapses, is now being approached as the target for many neurological and psychiatric disorders, including HD. HD is an autosomal dominant and progressive degenerative disorder, with clinical manifestations that encompass movement, cognition, mood and behaviour. HD is one of the most common tandem repeat disorders and is caused by a trinucleotide (CAG) repeat expansion, encoding an extended polyglutamine tract in the huntingtin protein. Animal models as well as human studies have provided detailed, although not exhaustive, evidence of synaptic dysfunction in HD. In this review, we discuss the neuropathology of HD and how the changes in synaptic signalling in the diseased brain lead to its symptoms, which include dementia. Here, we review and discuss the mechanisms by which the 'molecular orchestras' and their 'synaptic symphonies' are disrupted in neurodegeneration and dementia, focusing on HD as a model disease. We also explore the therapeutic strategies currently in pre-clinical and clinical testing that are targeted towards improving synaptic function in HD.
Collapse
Affiliation(s)
- Shiraz Tyebji
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
22
|
Huang Y, Todd N, Thathiah A. The role of GPCRs in neurodegenerative diseases: avenues for therapeutic intervention. Curr Opin Pharmacol 2017; 32:96-110. [DOI: 10.1016/j.coph.2017.02.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/20/2022]
|
23
|
McColgan P, Gregory S, Razi A, Seunarine KK, Gargouri F, Durr A, Roos RAC, Leavitt BR, Scahill RI, Clark CA, Tabrizi SJ, Rees G, Coleman A, Decolongon J, Fan M, Petkau T, Jauffret C, Justo D, Lehericy S, Nigaud K, Valabrègue R, Choonderbeek A, Hart EPT, Hensman Moss DJ, Crawford H, Johnson E, Papoutsi M, Berna C, Reilmann R, Weber N, Stout J, Labuschagne I, Landwehrmeyer B, Orth M, Johnson H. White matter predicts functional connectivity in premanifest Huntington's disease. Ann Clin Transl Neurol 2017; 4:106-118. [PMID: 28168210 PMCID: PMC5288460 DOI: 10.1002/acn3.384] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 02/01/2023] Open
Abstract
Objectives The distribution of pathology in neurodegenerative disease can be predicted by the organizational characteristics of white matter in healthy brains. However, we have very little evidence for the impact these pathological changes have on brain function. Understanding any such link between structure and function is critical for understanding how underlying brain pathology influences the progressive behavioral changes associated with neurodegeneration. Here, we demonstrate such a link between structure and function in individuals with premanifest Huntington's. Methods Using diffusion tractography and resting state functional magnetic resonance imaging to characterize white matter organization and functional connectivity, we investigate whether characteristic patterns of white matter organization in the healthy human brain shape the changes in functional coupling between brain regions in premanifest Huntington's disease. Results We find changes in functional connectivity in premanifest Huntington's disease that link directly to underlying patterns of white matter organization in healthy brains. Specifically, brain areas with strong structural connectivity show decreases in functional connectivity in premanifest Huntington's disease relative to controls, while regions with weak structural connectivity show increases in functional connectivity. Furthermore, we identify a pattern of dissociation in the strongest functional connections between anterior and posterior brain regions such that anterior functional connectivity increases in strength in premanifest Huntington's disease, while posterior functional connectivity decreases. Interpretation Our findings demonstrate that organizational principles of white matter underlie changes in functional connectivity in premanifest Huntington's disease. Furthermore, we demonstrate functional antero–posterior dissociation that is in keeping with the caudo–rostral gradient of striatal pathology in HD.
Collapse
|
24
|
Kobayashi Y, Matsuda S, Imamura K, Kobayashi H. Hydrogen generation by reaction of Si nanopowder with neutral water. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2017; 19:176. [PMID: 28579914 PMCID: PMC5434163 DOI: 10.1007/s11051-017-3873-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/27/2017] [Indexed: 05/10/2023]
Abstract
Si and its oxide are nonpoisonous materials, and thus, it can be taken for medical effects. We have developed a method of generation of hydrogen by use of reactions of Si nanopowder with water in the neutral pH region. Si nanopowder is fabricated by the simple bead milling method. Si nanopowder reacts with water to generate hydrogen even in cases where pH is set at the neutral region between 7.0 and 8.6. The hydrogen generation rate strongly depends on pH and in the case of pH 8.0, ∼55 ml/g hydrogen which corresponds to that contained in approximately 3 L saturated hydrogen-rich water is generated in 1 h. The reaction rate for hydrogen generation greatly increases with pH, indicating that the reacting species is hydroxide ions. The change of pH after the hydrogen generation reaction is negligibly low compared with that estimated assuming that hydroxide ions are consumed by the reaction. From these results, we conclude the following reaction mechanism: Si nanopowder reacts with hydroxide ions in the rate-determining reaction to form hydrogen molecules, SiO2, and electrons in the conduction band. Then, generated electrons are accepted by water molecules, resulting in production of hydrogen molecules and hydroxide ions. The hydrogen generation rate strongly depends on the crystallite size of Si nanopowder, but not on the size of aggregates of Si nanopowder. The present study shows a possibility to use Si nanopowder for hydrogen generation in the body in order to eliminate hydroxyl radicals which cause various diseases.
Collapse
Affiliation(s)
- Yuki Kobayashi
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Osaka, Ibaraki 567-0047 Japan
| | - Shinsuke Matsuda
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Osaka, Ibaraki 567-0047 Japan
| | - Kentaro Imamura
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Osaka, Ibaraki 567-0047 Japan
| | - Hikaru Kobayashi
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Osaka, Ibaraki 567-0047 Japan
| |
Collapse
|
25
|
Galan-Rodriguez B, Martin E, Brouillet E, Déglon N, Betuing S, Caboche J. Coupling of D2R Short but not D2R Long receptor isoform to the Rho/ROCK signaling pathway renders striatal neurons vulnerable to mutant huntingtin. Eur J Neurosci 2016; 45:198-206. [PMID: 27717053 DOI: 10.1111/ejn.13415] [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: 04/27/2016] [Revised: 09/09/2016] [Accepted: 09/21/2016] [Indexed: 01/27/2023]
Abstract
Huntington's disease, an inherited neurodegenerative disorder, results from abnormal polyglutamine extension in the N-terminal region of the huntingtin protein. This mutation causes preferential degeneration of striatal projection neurons. We previously demonstrated, in vitro, that dopaminergic D2 receptor stimulation acted in synergy with expanded huntingtin to increase aggregates formation and striatal death through activation of the Rho/ROCK signaling pathway. In vivo, in a lentiviral-mediated model of expanded huntingtin expression in the rat striatum, we found that the D2 antagonist haloperidol protects striatal neurons against expanded huntingtin-mediated toxicity. Two variant transcripts are generated by alternative splicing of the of D2 receptor gene, the D2R-Long and the D2R-Short, which are thought to play different functional roles. We show herein that overexpression of D2R-Short, but not D2R-Long in cell lines is associated with activation of the RhoA/ROCK signaling pathway. In striatal neurons in culture, the selective D2 agonist Quinpirole triggers phosphorylation of cofilin, a downstream effector of ROCK, which is abrogated by siRNAs that knockdown both D2R-Long and D2R-Short, but not by siRNAs targeting D2R-Long alone. Aggregate formation and neuronal death induced by expanded huntingtin, were potentiated by Quinpirole. This D2 agonist-mediated effect was selectively inhibited by the siRNA targeting both D2R-Long and D2R-Short but not D2R-Long alone. Our data provide evidence for a specific coupling of D2R-Short to the RhoA/ROCK/cofilin pathway, and its involvement in striatal vulnerability to expanded huntingtin. A new route for targeting Rho-ROCK signaling in Huntington's disease is unraveled with our findings.
Collapse
Affiliation(s)
- Beatriz Galan-Rodriguez
- UMRS-INSERM1130, Neurosciences Paris Seine, Paris, France.,Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Sevilla, Spain
| | - Elodie Martin
- INSERM UMRS_1127/UPMC/CNRS UMR7225, Institut du Cerveau et de la Moelle, Hôpital Pitié-Salpêtrière, Paris, France
| | - Emmanuel Brouillet
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses, France.,Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, Fontenay-aux-Roses, France
| | - Nicole Déglon
- Department of Clinical Neurosciences (DNC), Laboratory of Cellular and Molecular Neurotherapies (LNCM), Lausanne University Medical School (CHUV), Lausanne, Switzerland.,Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies (LNCM), Lausanne University Medical School (CHUV), Lausanne, Switzerland
| | - Sandrine Betuing
- UMRS-INSERM1130, Neurosciences Paris Seine, Paris, France.,UMR CNRS-8246, Paris, France.,Sorbonne Université, UM119, Université Pierre and Marie Curie-Paris 6, 9 quai Saint Bernard, 75005, Paris, France
| | - Jocelyne Caboche
- UMRS-INSERM1130, Neurosciences Paris Seine, Paris, France.,UMR CNRS-8246, Paris, France.,Sorbonne Université, UM119, Université Pierre and Marie Curie-Paris 6, 9 quai Saint Bernard, 75005, Paris, France
| |
Collapse
|
26
|
de Diego-Balaguer R, Schramm C, Rebeix I, Dupoux E, Durr A, Brice A, Charles P, Cleret de Langavant L, Youssov K, Verny C, Damotte V, Azulay JP, Goizet C, Simonin C, Tranchant C, Maison P, Rialland A, Schmitz D, Jacquemot C, Fontaine B, Bachoud-Lévi AC. COMT Val158Met Polymorphism Modulates Huntington's Disease Progression. PLoS One 2016; 11:e0161106. [PMID: 27657697 PMCID: PMC5033325 DOI: 10.1371/journal.pone.0161106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/29/2016] [Indexed: 11/19/2022] Open
Abstract
Little is known about the genetic factors modulating the progression of Huntington's disease (HD). Dopamine levels are affected in HD and modulate executive functions, the main cognitive disorder of HD. We investigated whether the Val158Met polymorphism of the catechol-O-methyltransferase (COMT) gene, which influences dopamine (DA) degradation, affects clinical progression in HD. We carried out a prospective longitudinal multicenter study from 1994 to 2011, on 438 HD gene carriers at different stages of the disease (34 pre-manifest; 172 stage 1; 130 stage 2; 80 stage 3; 17 stage 4; and 5 stage 5), according to Total Functional Capacity (TFC) score. We used the Unified Huntington's Disease Rating Scale to evaluate motor, cognitive, behavioral and functional decline. We genotyped participants for COMT polymorphism (107 Met-homozygous, 114 Val-homozygous and 217 heterozygous). 367 controls of similar ancestry were also genotyped. We compared clinical progression, on each domain, between groups of COMT polymorphisms, using latent-class mixed models accounting for disease duration and number of CAG (cytosine adenine guanine) repeats. We show that HD gene carriers with fewer CAG repeats and with the Val allele in COMT polymorphism displayed slower cognitive decline. The rate of cognitive decline was greater for Met/Met homozygotes, which displayed a better maintenance of cognitive capacity in earlier stages of the disease, but had a worse performance than Val allele carriers later on. COMT polymorphism did not significantly impact functional and behavioral performance. Since COMT polymorphism influences progression in HD, it could be used for stratification in future clinical trials. Moreover, DA treatments based on the specific COMT polymorphism and adapted according to disease duration could potentially slow HD progression.
Collapse
Affiliation(s)
- Ruth de Diego-Balaguer
- INSERM U955, Equipe 01 Neuropsychologie Interventionnelle, 94000, Créteil, France
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Université Paris Est, Faculté de Médecine, 94000, Créteil, France
- ICREA, 08010, Barcelona, Spain
- Universitat de Barcelona, Departament de Cognició, Desenvolupament i Psicologia de L’Educació, 08035, Barcelona, Spain
- IDIBELL, Unitat de Cognició i Plasticitat Cerebral, 08907, L’Hospitalet de Llobregat, Spain
- Institut de Neurociència, Universitat de Barcelona, Barcelona, Spain
| | - Catherine Schramm
- INSERM U955, Equipe 01 Neuropsychologie Interventionnelle, 94000, Créteil, France
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Université Paris Est, Faculté de Médecine, 94000, Créteil, France
| | - Isabelle Rebeix
- INSERM-UPMC-CNRS, UMR 7225–1127, Institut Cerveau Moelle-ICM, Hôpital Pitié-Salpêtrière, 74013, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département des Maladies du Système Nerveux, Hôpital Pitié-Salpêtrière, 74013, Paris, France
| | - Emmanuel Dupoux
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Laboratoire de Sciences Cognitives et Psycholinguistique, ENS-EHESS-CNRS, Paris, 75005, France
| | - Alexandra Durr
- INSERM-UPMC-CNRS, UMR 7225–1127, Institut Cerveau Moelle-ICM, Hôpital Pitié-Salpêtrière, 74013, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département de Génétique, Hôpital Pitié-Salpêtrière, 74013, Paris, France
| | - Alexis Brice
- INSERM-UPMC-CNRS, UMR 7225–1127, Institut Cerveau Moelle-ICM, Hôpital Pitié-Salpêtrière, 74013, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département de Génétique, Hôpital Pitié-Salpêtrière, 74013, Paris, France
| | - Perrine Charles
- Assistance Publique-Hôpitaux de Paris, Département de Génétique, Hôpital Pitié-Salpêtrière, 74013, Paris, France
| | - Laurent Cleret de Langavant
- INSERM U955, Equipe 01 Neuropsychologie Interventionnelle, 94000, Créteil, France
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Université Paris Est, Faculté de Médecine, 94000, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, 94000, Créteil, France
| | - Katia Youssov
- INSERM U955, Equipe 01 Neuropsychologie Interventionnelle, 94000, Créteil, France
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Université Paris Est, Faculté de Médecine, 94000, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, 94000, Créteil, France
| | - Christophe Verny
- CHU d'Angers, Centre de Référence des Maladies Neurogénétiques, Service de Neurologie, 49933, Angers, France
| | - Vincent Damotte
- INSERM-UPMC-CNRS, UMR 7225–1127, Institut Cerveau Moelle-ICM, Hôpital Pitié-Salpêtrière, 74013, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département des Maladies du Système Nerveux, Hôpital Pitié-Salpêtrière, 74013, Paris, France
| | - Jean-Philippe Azulay
- CHU de Marseille—Hôpital de la Timone, Service de Neurologie et Pathologie du Mouvement, 13385, Marseille, France
| | - Cyril Goizet
- CHU de Bordeaux-GH Sud—Hôpital Haut-Lévêque, Service de Neurologie, 33604, Pessac, France
| | - Clémence Simonin
- CHRU de Lille, Service de Neurologie et Pathologie du Mouvement, 59000, Lille, France
- INSERM UMR-S 1172, JPArc, centre de recherche Jean-Pierre-Aubert neurosciences et cancer, Université de Lille, 59000, Lille, France
| | - Christine Tranchant
- CHU de Strasbourg—Hôpital de Hautepierre, Service de Neurologie, 67098, Strasbourg, France
| | - Patrick Maison
- INSERM U955, Equipe 01 Neuropsychologie Interventionnelle, 94000, Créteil, France
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Université Paris Est, Faculté de Médecine, 94000, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Unité de Recherche Clinique, 94000, Créteil, France
| | - Amandine Rialland
- Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Unité de Recherche Clinique, 94000, Créteil, France
| | - David Schmitz
- Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Unité de Recherche Clinique, 94000, Créteil, France
| | - Charlotte Jacquemot
- INSERM U955, Equipe 01 Neuropsychologie Interventionnelle, 94000, Créteil, France
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Université Paris Est, Faculté de Médecine, 94000, Créteil, France
| | - Bertrand Fontaine
- Assistance Publique-Hôpitaux de Paris, Département des Maladies du Système Nerveux, Hôpital Pitié-Salpêtrière, 74013, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département de Génétique, Hôpital Pitié-Salpêtrière, 74013, Paris, France
| | - Anne-Catherine Bachoud-Lévi
- INSERM U955, Equipe 01 Neuropsychologie Interventionnelle, 94000, Créteil, France
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL Research University, 75005, Paris, France
- Université Paris Est, Faculté de Médecine, 94000, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, 94000, Créteil, France
- * E-mail:
| | | |
Collapse
|
27
|
Metabolic signatures of Huntington's disease (HD): 1 H NMR analysis of the polar metabolome in post-mortem human brain. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1675-84. [DOI: 10.1016/j.bbadis.2016.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/27/2016] [Accepted: 06/07/2016] [Indexed: 12/22/2022]
|
28
|
Boussicault L, Alves S, Lamazière A, Planques A, Heck N, Moumné L, Despres G, Bolte S, Hu A, Pagès C, Galvan L, Piguet F, Aubourg P, Cartier N, Caboche J, Betuing S. CYP46A1, the rate-limiting enzyme for cholesterol degradation, is neuroprotective in Huntington's disease. Brain 2016; 139:953-70. [PMID: 26912634 PMCID: PMC4766376 DOI: 10.1093/brain/awv384] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/04/2015] [Indexed: 11/22/2022] Open
Abstract
Huntington’s disease is an autosomal dominant neurodegenerative disease caused by abnormal polyglutamine expansion in huntingtin (Exp-HTT) leading to degeneration of striatal neurons. Altered brain cholesterol homeostasis has been implicated in Huntington’s disease, with increased accumulation of cholesterol in striatal neurons yet reduced levels of cholesterol metabolic precursors. To elucidate these two seemingly opposing dysregulations, we investigated the expression of cholesterol 24-hydroxylase (CYP46A1), the neuronal-specific and rate-limiting enzyme for cholesterol conversion to 24S-hydroxycholesterol (24S-OHC). CYP46A1 protein levels were decreased in the putamen, but not cerebral cortex samples, of post-mortem Huntington’s disease patients when compared to controls.
Cyp46A1
mRNA and CYP46A1 protein levels were also decreased in the striatum of the R6/2 Huntington’s disease mouse model and in ST
hdh
Q111 cell lines.
In vivo
, in a wild-type context, knocking down CYP46A1 expression in the striatum, via an adeno-associated virus-mediated delivery of selective shCYP46A1, reproduced the Huntington’s disease phenotype, with spontaneous striatal neuron degeneration and motor deficits, as assessed by rotarod.
In vitro
, CYP46A1 restoration protected ST
hdh
Q111 and Exp-HTT-expressing striatal neurons in culture from cell death. In the R6/2 Huntington’s disease mouse model, adeno-associated virus-mediated delivery of CYP46A1 into the striatum decreased neuronal atrophy, decreased the number, intensity level and size of Exp-HTT aggregates and improved motor deficits, as assessed by rotarod and clasping behavioural tests. Adeno-associated virus-CYP46A1 infection in R6/2 mice also restored levels of cholesterol and lanosterol and increased levels of desmosterol.
In vitro
, lanosterol and desmosterol were found to protect striatal neurons expressing Exp-HTT from death. We conclude that restoring CYP46A1 activity in the striatum promises a new therapeutic approach in Huntington’s disease.
Collapse
Affiliation(s)
- Lydie Boussicault
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France
| | - Sandro Alves
- 2 INSERM U1169, Le Kremlin-Bicêtre, MIRCEN CEA and Université Paris-Sud, Université Paris Saclay, 91400 Orsay, France
| | - Antonin Lamazière
- 3 Laboratory of Mass Spectrometry, INSERM ERL 1157, CNRS UMR 7203 LBM, Sorbonne Universités- Université Pierre et Marie Curie-Paris 6, CHU Saint-Antoine, 75012 Paris, France
| | - Anabelle Planques
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France 4 Development and Neuropharmacology, Center for Interdisciplinary Research in Biology, INSERM CNRS 7141. Collège de France
| | - Nicolas Heck
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France
| | - Lara Moumné
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France
| | - Gaëtan Despres
- 3 Laboratory of Mass Spectrometry, INSERM ERL 1157, CNRS UMR 7203 LBM, Sorbonne Universités- Université Pierre et Marie Curie-Paris 6, CHU Saint-Antoine, 75012 Paris, France
| | - Susanne Bolte
- 5 Cellular Imaging Facility, Institute of Biology Paris-Seine CNRS FR, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, Paris, France
| | - Amélie Hu
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France 6 Laboratory of Experimental Neurology, Université Libre de Bruxelles, Belgium
| | - Christiane Pagès
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France
| | - Laurie Galvan
- 7 Semel Institute, University California Los Angeles, Los Angeles, USA
| | - Francoise Piguet
- 8 Department of Translational Medicine and Neurogenetics, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), UMR 7104 CNRS/UdS, INSERM U964, BP 10142, 67404 Illkirch Cedex, France
| | - Patrick Aubourg
- 2 INSERM U1169, Le Kremlin-Bicêtre, MIRCEN CEA and Université Paris-Sud, Université Paris Saclay, 91400 Orsay, France
| | - Nathalie Cartier
- 2 INSERM U1169, Le Kremlin-Bicêtre, MIRCEN CEA and Université Paris-Sud, Université Paris Saclay, 91400 Orsay, France
| | - Jocelyne Caboche
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France
| | - Sandrine Betuing
- 1 Neuronal Signaling and Gene Regulation, Neurosciences Paris Seine, Institute of Biology Paris-Seine, Sorbonne Universités, UPMC Université Pierre et Marie Curie-Paris 6, INSERM/UMR-S 1130, CNRS/UMR 8246, 75005 Paris, France
| |
Collapse
|
29
|
Xie W, Wang JQ, Wang QC, Wang Y, Yao S, Tang TS. Adult neural progenitor cells from Huntington's disease mouse brain exhibit increased proliferation and migration due to enhanced calcium and ROS signals. Cell Prolif 2015; 48:517-31. [PMID: 26269226 DOI: 10.1111/cpr.12205] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/10/2015] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Huntington's disease (HD) is an inherited human neurodegenerative disorder characterized by uncontrollable movement, psychiatric disturbance and cognitive decline. Impaired proliferative/differentiational potentials of adult neural progenitor cells (ANPCs) have been thought to be a pathogenic mechanism involved in it. In this study, we aimed to elucidate intrinsic properties of ANPCs subjected to neurodegenerative condition in YAC128 HD mice. MATERIALS AND METHODS ANPCs were isolated from the SVZ regions of 4-month-old WT and YAC128 mice. Cell proliferation, migration and neuronal differentiation in vitro were compared between these two genotypes with/without Ca(2+) inhibitors or ROS scavenger treatments. Differences in ANPC proliferation and differentiation capabilities in vivo between the two genotypes were evaluated using Ki-67 and Doublecortin (DCX) immunofluorescence respectively. RESULTS Compared to WT ANPCs, YAC128 ANPCs had significantly enhanced cell proliferation, migration and neuronal differentiation in vitro, accompanied by increased Ca(2+) and ROS signals. Raised proliferation and migration in YAC128 ANPCs were abolished by Ca(2+) signalling antagonists and ROS scavenging. However, in vivo, HD ANPCs failed to show any elevated proliferation or differentiation. CONCLUSIONS Increased Ca(2+) signalling and higher level of ROS conferred HD ANPC enhancement of proliferation and migration potentials. However, the in vivo micro-environment did not support endogenous ANPCs to respond appropriately to neuronal loss in these YAC128 mouse brains.
Collapse
Affiliation(s)
- Wenjuan Xie
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiu-Qiang Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiao-Chu Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yun Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Sheng Yao
- Department of Neurology, Navy General Hospital, Beijing, 100048, China
| | - Tie-Shan Tang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
30
|
Schwab LC, Garas SN, Garas SN, Drouin-Ouellet J, Mason SL, Stott SR, Barker RA. Dopamine and Huntington's disease. Expert Rev Neurother 2015; 15:445-58. [PMID: 25773746 DOI: 10.1586/14737175.2015.1025383] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Huntington's disease (HD) is an incurable, inherited, progressive neurodegenerative disorder that is defined by a combination of motor, cognitive and psychiatric features. Pre-clinical and clinical studies have demonstrated an important role for the dopamine (DA) system in HD with dopaminergic dysfunction at the level of both DA release and DA receptors. It is, therefore, not surprising that the drug treatments most commonly used in HD are anti-dopaminergic agents. Their use is based primarily on the belief that the characteristic motor impairments are a result of overactivation of the central dopaminergic pathways. While this is a useful starting place, it is clear that the behavior of the central dopaminergic pathways is not fully understood in this condition and may change as a function of disease stage. In addition, how abnormalities in dopaminergic systems may underlie some of the non-motor features of HD has also been poorly investigated and this is especially important given the greater burden these place on the patients' and families' quality of life. In this review, we discuss what is known about central dopaminergic pathways in HD and how this informs us about the mechanisms of action of the dopaminergic therapies used to treat it. By doing so, we will highlight some of the paradoxes that exist and how solving them may reveal new insights for improved treatment of this currently incurable condition, including the possibility that such drugs may even have effects on disease progression and pathogenesis.
Collapse
Affiliation(s)
- Laetitia C Schwab
- John van Geest Centre for Brain Repair, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | | | | | | | | | | | | |
Collapse
|
31
|
Gardoni F, Bellone C. Modulation of the glutamatergic transmission by Dopamine: a focus on Parkinson, Huntington and Addiction diseases. Front Cell Neurosci 2015; 9:25. [PMID: 25784855 PMCID: PMC4345909 DOI: 10.3389/fncel.2015.00025] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/14/2015] [Indexed: 01/11/2023] Open
Abstract
Dopamine (DA) plays a major role in motor and cognitive functions as well as in reward processing by regulating glutamatergic inputs. In particular in the striatum the release of DA rapidly influences synaptic transmission modulating both AMPA and NMDA receptors. Several neurodegenerative and neuropsychiatric disorders, including Parkinson, Huntington and addiction-related diseases, manifest a dysregulation of glutamate and DA signaling. Here, we will focus our attention on the mechanisms underlying the modulation of the glutamatergic transmission by DA in striatal circuits.
Collapse
Affiliation(s)
- Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milano Milano, Italy
| | - Camilla Bellone
- Department of Fundamental Neuroscience, University of Lausanne Lausanne, Switzerland
| |
Collapse
|
32
|
Striatal long noncoding RNA Abhd11os is neuroprotective against an N-terminal fragment of mutant huntingtin in vivo. Neurobiol Aging 2014; 36:1601.e7-16. [PMID: 25619660 DOI: 10.1016/j.neurobiolaging.2014.11.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/28/2014] [Accepted: 11/25/2014] [Indexed: 12/26/2022]
Abstract
A large number of gene products that are enriched in the striatum have ill-defined functions, although they may have key roles in age-dependent neurodegenerative diseases affecting the striatum, especially Huntington disease (HD). In the present study, we focused on Abhd11os, (called ABHD11-AS1 in human) which is a putative long noncoding RNA (lncRNA) whose expression is enriched in the mouse striatum. We confirm that despite the presence of 2 small open reading frames (ORFs) in its sequence, Abhd11os is not translated into a detectable peptide in living cells. We demonstrate that Abhd11os levels are markedly reduced in different mouse models of HD. We performed in vivo experiments in mice using lentiviral vectors encoding either Abhd11os or a small hairpin RNA targeting Abhd11os. Results show that Abhd11os overexpression produces neuroprotection against an N-terminal fragment of mutant huntingtin, whereas Abhd11os knockdown is protoxic. These novel results indicate that the loss lncRNA Abhd11os likely contribute to striatal vulnerability in HD. Our study emphasizes that lncRNA may play crucial roles in neurodegenerative diseases.
Collapse
|
33
|
Lebel M, Robinson P, Cyr M. Canadian Association of Neurosciences Review: The Role of Dopamine Receptor Function in Neurodegenerative Diseases. Can J Neurol Sci 2014; 34:18-29. [PMID: 17352343 DOI: 10.1017/s0317167100005746] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Dopamine (DA) receptors, which are heavily expressed in the caudate/putamen of the brain, represent the molecular target of several drugs used in the treatment of various neurological disorders, such as Parkinson's disease. Although most of the drugs are very effective in alleviating the symptoms associated with these conditions, their long-term utilization could lead to the development of severe side-effects. In addition to uncovering novel mediators of physiological DA receptor functions, recent research advances are suggesting a role of these receptors in toxic effects on neurons. For instance, accumulating evidence indicates that DA receptors, particularly D1 receptors, are central in the neuronal toxicity induced by elevated synaptic levels of DA. In this review, we will discuss recent findings on DA receptors as regulators of long term neuronal dysfunction and neurodegenerative processes.
Collapse
Affiliation(s)
- Manon Lebel
- Neuroscience Research Group, Université du Québec à Trois-Rivières, Canada
| | | | | |
Collapse
|
34
|
Francelle L, Galvan L, Gaillard MC, Guillermier M, Houitte D, Bonvento G, Petit F, Jan C, Dufour N, Hantraye P, Elalouf JM, De Chaldée M, Déglon N, Brouillet E. Loss of the thyroid hormone-binding protein Crym renders striatal neurons more vulnerable to mutant huntingtin in Huntington's disease. Hum Mol Genet 2014; 24:1563-73. [PMID: 25398949 PMCID: PMC4381754 DOI: 10.1093/hmg/ddu571] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The mechanisms underlying preferential atrophy of the striatum in Huntington's disease (HD) are unknown. One hypothesis is that a set of gene products preferentially expressed in the striatum could determine the particular vulnerability of this brain region to mutant huntingtin (mHtt). Here, we studied the striatal protein µ-crystallin (Crym). Crym is the NADPH-dependent p38 cytosolic T3-binding protein (p38CTBP), a key regulator of thyroid hormone (TH) T3 (3,5,3'-triiodo-l-thyronine) transportation. It has been also recently identified as the enzyme that reduces the sulfur-containing cyclic ketimines, which are potential neurotransmitters. Here, we confirm the preferential expression of the Crym protein in the rodent and macaque striatum. Crym expression was found to be higher in the macaque caudate than in the putamen. Expression of Crym was reduced in the BACHD and Knock-in 140CAG mouse models of HD before onset of striatal atrophy. We show that overexpression of Crym in striatal medium-size spiny neurons using a lentiviral-based strategy in mice is neuroprotective against the neurotoxicity of an N-terminal fragment of mHtt in vivo. Thus, reduction of Crym expression in HD could render striatal neurons more susceptible to mHtt suggesting that Crym may be a key determinant of the vulnerability of the striatum. In addition our work points to Crym as a potential molecular link between striatal degeneration and the THs deregulation reported in HD patients.
Collapse
Affiliation(s)
- Laetitia Francelle
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Laurie Galvan
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Martine Guillermier
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Diane Houitte
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Gilles Bonvento
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Fanny Petit
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Caroline Jan
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Noëlle Dufour
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Philippe Hantraye
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France
| | - Jean-Marc Elalouf
- CEA, iBiTecS, F-91191 Gif-sur-Yvette Cedex, France, CNRS, FRE 3377, F-91191 Gif-sur-Yvette Cedex, France, Université Paris-Sud, FRE 3377, F-91191 Gif-sur-Yvette Cedex, France
| | - Michel De Chaldée
- CEA, iBiTecS, F-91191 Gif-sur-Yvette Cedex, France, CNRS, FRE 3377, F-91191 Gif-sur-Yvette Cedex, France, Université Paris-Sud, FRE 3377, F-91191 Gif-sur-Yvette Cedex, France
| | - Nicole Déglon
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France, Laboratory of Cellular and Molecular Neurotherapies, Department of Clinical Neurociences, Lausanne University Hospital, Lausanne, Switzerland
| | - Emmanuel Brouillet
- CEA, DSV, I²BM, Molecular Imaging Research Center (MIRCen), F-92265 Fontenay-aux-Roses, France, Neurodegenerative Diseases Laboratory, CNRS CEA URA 2210, F-92265 Fontenay-aux-Roses, France,
| |
Collapse
|
35
|
Francelle L, Galvan L, Brouillet E. Possible involvement of self-defense mechanisms in the preferential vulnerability of the striatum in Huntington's disease. Front Cell Neurosci 2014; 8:295. [PMID: 25309327 PMCID: PMC4176035 DOI: 10.3389/fncel.2014.00295] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/03/2014] [Indexed: 01/01/2023] Open
Abstract
HD is caused by a mutation in the huntingtin gene that consists in a CAG repeat expansion translated into an abnormal poly-glutamine (polyQ) tract in the huntingtin (Htt) protein. The most striking neuropathological finding in HD is the atrophy of the striatum. The regional expression of mutant Htt (mHtt) is ubiquitous in the brain and cannot explain by itself the preferential vulnerability of the striatum in HD. mHtt has been shown to produce an early defect in transcription, through direct alteration of the function of key regulators of transcription and in addition, more indirectly, as a result of compensatory responses to cellular stress. In this review, we focus on gene products that are preferentially expressed in the striatum and have down- or up-regulated expression in HD and, as such, may play a crucial role in the susceptibility of the striatum to mHtt. Many of these striatal gene products are for a vast majority down-regulated and more rarely increased in HD. Recent research shows that some of these striatal markers have a pro-survival/neuroprotective role in neurons (e.g., MSK1, A2A, and CB1 receptors) whereas others enhance the susceptibility of striatal neurons to mHtt (e.g., Rhes, RGS2, D2 receptors). The down-regulation of these latter proteins may be considered as a potential self-defense mechanism to slow degeneration. For a majority of the striatal gene products that have been identified so far, their function in the striatum is unknown and their modifying effects on mHtt toxicity remain to be experimentally addressed. Focusing on these striatal markers may contribute to a better understanding of HD pathogenesis, and possibly the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Laetitia Francelle
- Neurodegenerative Disease Laboratory, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Direction des Sciences du Vivant, Institut d'Imagerie BioMédicale, Molecular Imaging Research Center Fontenay-aux-Roses, France ; Centre National de la Recherche Scientifique - Commissariat à l'Énergie Atomique et aux Énergies Alternatives Unité de Recherche Associée 2210 Fontenay-aux-Roses, France
| | - Laurie Galvan
- Neurodegenerative Disease Laboratory, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Direction des Sciences du Vivant, Institut d'Imagerie BioMédicale, Molecular Imaging Research Center Fontenay-aux-Roses, France ; Centre National de la Recherche Scientifique - Commissariat à l'Énergie Atomique et aux Énergies Alternatives Unité de Recherche Associée 2210 Fontenay-aux-Roses, France ; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles Los Angeles, CA, USA
| | - Emmanuel Brouillet
- Neurodegenerative Disease Laboratory, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Direction des Sciences du Vivant, Institut d'Imagerie BioMédicale, Molecular Imaging Research Center Fontenay-aux-Roses, France ; Centre National de la Recherche Scientifique - Commissariat à l'Énergie Atomique et aux Énergies Alternatives Unité de Recherche Associée 2210 Fontenay-aux-Roses, France
| |
Collapse
|
36
|
Wang BY, Xu XS, Cui YX, Wang H, Liu G, Zhao ZJ, Ma JF, Fu XQ. Caenorhabditis elegans eyes absent ortholog EYA-1 Is required for stress resistance. BIOCHEMISTRY (MOSCOW) 2014; 79:653-62. [DOI: 10.1134/s0006297914070074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Margulis J, Finkbeiner S. Proteostasis in striatal cells and selective neurodegeneration in Huntington's disease. Front Cell Neurosci 2014; 8:218. [PMID: 25147502 PMCID: PMC4124811 DOI: 10.3389/fncel.2014.00218] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/17/2014] [Indexed: 12/23/2022] Open
Abstract
Selective neuronal loss is a hallmark of neurodegenerative diseases, including Huntington’s disease (HD). Although mutant huntingtin, the protein responsible for HD, is expressed ubiquitously, a subpopulation of neurons in the striatum is the first to succumb. In this review, we examine evidence that protein quality control pathways, including the ubiquitin proteasome system, autophagy, and chaperones, are significantly altered in striatal neurons. These alterations may increase the susceptibility of striatal neurons to mutant huntingtin-mediated toxicity. This novel view of HD pathogenesis has profound therapeutic implications: protein homeostasis pathways in the striatum may be valuable targets for treating HD and other misfolded protein disorders.
Collapse
Affiliation(s)
- Julia Margulis
- Gladstone Institute of Neurological Disease, J. David Gladstone Institutes San Francisco, CA, USA ; Department of Neurology, University of California at San Francisco San Francisco, CA, USA ; Department of Physiology, University of California at San Francisco San Francisco, CA, USA
| | - Steven Finkbeiner
- Gladstone Institute of Neurological Disease, J. David Gladstone Institutes San Francisco, CA, USA ; Department of Neurology, University of California at San Francisco San Francisco, CA, USA ; Department of Physiology, University of California at San Francisco San Francisco, CA, USA ; Taube/Koret Center for Huntington's Disease Research San Francisco, CA, USA
| |
Collapse
|
38
|
Navarro-Yepes J, Zavala-Flores L, Anandhan A, Wang F, Skotak M, Chandra N, Li M, Pappa A, Martinez-Fong D, Del Razo LM, Quintanilla-Vega B, Franco R. Antioxidant gene therapy against neuronal cell death. Pharmacol Ther 2014; 142:206-30. [PMID: 24333264 PMCID: PMC3959583 DOI: 10.1016/j.pharmthera.2013.12.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a common hallmark of neuronal cell death associated with neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, as well as brain stroke/ischemia and traumatic brain injury. Increased accumulation of reactive species of both oxygen (ROS) and nitrogen (RNS) has been implicated in mitochondrial dysfunction, energy impairment, alterations in metal homeostasis and accumulation of aggregated proteins observed in neurodegenerative disorders, which lead to the activation/modulation of cell death mechanisms that include apoptotic, necrotic and autophagic pathways. Thus, the design of novel antioxidant strategies to selectively target oxidative stress and redox imbalance might represent important therapeutic approaches against neurological disorders. This work reviews the evidence demonstrating the ability of genetically encoded antioxidant systems to selectively counteract neuronal cell loss in neurodegenerative diseases and ischemic brain damage. Because gene therapy approaches to treat inherited and acquired disorders offer many unique advantages over conventional therapeutic approaches, we discussed basic research/clinical evidence and the potential of virus-mediated gene delivery techniques for antioxidant gene therapy.
Collapse
Affiliation(s)
- Juliana Navarro-Yepes
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; Department of Toxicology, CINVESTAV-IPN, Mexico City, Mexico
| | - Laura Zavala-Flores
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Fang Wang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Maciej Skotak
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Namas Chandra
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, Alexandroupolis, Greece
| | - Daniel Martinez-Fong
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | | | | | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
39
|
Marion S, Urs NM, Peterson SM, Sotnikova TD, Beaulieu JM, Gainetdinov RR, Caron MG. Dopamine D2 receptor relies upon PPM/PP2C protein phosphatases to dephosphorylate huntingtin protein. J Biol Chem 2014; 289:11715-11724. [PMID: 24619418 PMCID: PMC4002081 DOI: 10.1074/jbc.m113.544312] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 02/26/2014] [Indexed: 12/24/2022] Open
Abstract
Striatal dopamine D2 receptor (D2R) relies upon G protein- and β-arrestin-dependent signaling pathways to convey its action on motor control and behavior. Considering that D2R activation inhibits Akt in the striatum and that huntingtin physiological functions are affected by Akt phosphorylation, we sought to investigate whether D2R-mediated signaling could regulate huntingtin phosphorylation. We demonstrate that D2R activation decreases huntingtin phosphorylation on its Akt site. This dephosphorylation event depends upon the Gαi-dependent engagement of specific members of the protein phosphatase metallo-dependent (PPM/PP2C) family and is independent of β-arrestin 2. These observations identify the PPM/PP2C family as a mediator of G protein-coupled receptor signaling and thereby suggest a novel mechanism of dopaminergic signaling.
Collapse
Affiliation(s)
- Sébastien Marion
- Departments of Cell Biology, Neurobiology Duke University Medical Center, Durham, North Carolina 27710
| | - Nikhil M Urs
- Departments of Cell Biology, Neurobiology Duke University Medical Center, Durham, North Carolina 27710
| | - Sean M Peterson
- Departments of Cell Biology, Neurobiology Duke University Medical Center, Durham, North Carolina 27710
| | - Tatyana D Sotnikova
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval/IUSMQ, Québec G1J 2G3, Canada
| | - Jean-Martin Beaulieu
- Department of Neuroscience, Italian Institute of Technology, 16163 Genova, Italy
| | - Raul R Gainetdinov
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval/IUSMQ, Québec G1J 2G3, Canada
| | - Marc G Caron
- Departments of Cell Biology, Neurobiology Duke University Medical Center, Durham, North Carolina 27710; Departments of Medicine, Neurobiology Duke University Medical Center, Durham, North Carolina 27710; Neurobiology Duke University Medical Center, Durham, North Carolina 27710.
| |
Collapse
|
40
|
Mishra J, Kumar A. Improvement of Mitochondrial Function by Paliperidone Attenuates Quinolinic Acid-Induced Behavioural and Neurochemical Alterations in Rats: Implications in Huntington’s Disease. Neurotox Res 2014; 26:363-81. [DOI: 10.1007/s12640-014-9469-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 11/29/2022]
|
41
|
Choi ML, Begeti F, Oh JH, Lee SY, O'Keeffe GC, Clelland CD, Tyers P, Cho ZH, Kim YB, Barker RA. Dopaminergic manipulations and its effects on neurogenesis and motor function in a transgenic mouse model of Huntington's disease. Neurobiol Dis 2014; 66:19-27. [PMID: 24561069 DOI: 10.1016/j.nbd.2014.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/29/2014] [Accepted: 02/10/2014] [Indexed: 11/28/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder that is classically defined by a triad of movement and cognitive and psychiatric abnormalities with a well-established pathology that affects the dopaminergic systems of the brain. This has classically been described in terms of an early loss of dopamine D2 receptors (D2R), although interestingly the treatments most effectively used to treat patients with HD block these same receptors. We therefore sought to examine the dopaminergic system in HD not only in terms of striatal function but also at extrastriatal sites especially the hippocampus, given that transgenic (Tg) mice also exhibit deficits in hippocampal-dependent cognitive tests and a reduction in adult hippocampal neurogenesis. We showed that there was an early reduction of D2R in both the striatum and dentate gyrus (DG) of the hippocampus in the R6/1 transgenic HD mouse ahead of any overt motor signs and before striatal neuronal loss. Despite downregulation of D2Rs in these sites, further reduction of the dopaminergic input to these sites by either medial forebrain bundle lesions or receptor blockade using sulpiride was able to improve both deficits in motor performance and adult hippocampal neurogenesis. In contrast, a reduction in dopaminergic innervation of the neurogenic niches resulted in impaired neurogenesis in healthy WT mice. This study therefore provides evidence that D2R blockade improves hippocampal and striatal deficits in HD mice although the underlying mechanism for this is unclear, and suggests that agents working within this network may have greater effects than previously thought.
Collapse
Affiliation(s)
- M L Choi
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - F Begeti
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK; School of Clinical Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0SP, UK
| | - J H Oh
- Neuroscience Research Institute, Gachon University, Incheon 405-760, Republic of Korea
| | - S Y Lee
- Neuroscience Research Institute, Gachon University, Incheon 405-760, Republic of Korea
| | - G C O'Keeffe
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - C D Clelland
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - P Tyers
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - Z H Cho
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - Y B Kim
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - R A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK; Department of Neurology, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| |
Collapse
|
42
|
Cepeda C, Murphy KPS, Parent M, Levine MS. The role of dopamine in Huntington's disease. PROGRESS IN BRAIN RESEARCH 2014; 211:235-54. [PMID: 24968783 PMCID: PMC4409123 DOI: 10.1016/b978-0-444-63425-2.00010-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alterations in dopamine (DA) neurotransmission in Parkinson's disease are well known and widely studied. Much less is known about DA changes that accompany and underlie some of the symptoms of Huntington's disease (HD), a dominant inherited neurodegenerative disorder characterized by chorea, cognitive deficits, and psychiatric disturbances. The cause is an expansion in CAG (glutamine) repeats in the HTT gene. The principal histopathology of HD is the loss of medium-sized spiny neurons (MSNs) and, to a lesser degree, neuronal loss in cerebral cortex, thalamus, hippocampus, and hypothalamus. Neurochemical, electrophysiological, and behavioral studies in HD patients and genetic mouse models suggest biphasic changes in DA neurotransmission. In the early stages, DA neurotransmission is increased leading to hyperkinetic movements that can be alleviated by depleting DA stores. In contrast, in the late stages, DA deficits produce hypokinesia that can be treated by increasing DA function. Alterations in DA neurotransmission affect glutamate receptor modulation and could contribute to excitotoxicity. The mechanisms of DA dysfunction, in particular the increased DA tone in the early stages of the disease, are presently unknown but may include initial upregulation of DA neuron activity caused by the genetic mutation, reduced inhibition resulting from striatal MSN loss, increased excitation from cortical inputs, and DA autoreceptor dysfunction. Targeting both DA and glutamate receptor dysfunction could be the best strategy to treat HD symptoms.
Collapse
Affiliation(s)
- Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Kerry P S Murphy
- Huntington's Disease Research Forum, Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, Buckinghamshire, UK
| | - Martin Parent
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec City, QC, Canada
| | - Michael S Levine
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
43
|
Lauterbach EC. Neuroprotective effects of psychotropic drugs in Huntington's disease. Int J Mol Sci 2013; 14:22558-603. [PMID: 24248060 PMCID: PMC3856079 DOI: 10.3390/ijms141122558] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/16/2013] [Accepted: 10/16/2013] [Indexed: 02/06/2023] Open
Abstract
Psychotropics (antipsychotics, mood stabilizers, antidepressants, anxiolytics, etc.) are commonly prescribed to treat Huntington’s disease (HD). In HD preclinical models, while no psychotropic has convincingly affected huntingtin gene, HD modifying gene, or huntingtin protein expression, psychotropic neuroprotective effects include upregulated huntingtin autophagy (lithium), histone acetylation (lithium, valproate, lamotrigine), miR-222 (lithium-plus-valproate), mitochondrial protection (haloperidol, trifluoperazine, imipramine, desipramine, nortriptyline, maprotiline, trazodone, sertraline, venlafaxine, melatonin), neurogenesis (lithium, valproate, fluoxetine, sertraline), and BDNF (lithium, valproate, sertraline) and downregulated AP-1 DNA binding (lithium), p53 (lithium), huntingtin aggregation (antipsychotics, lithium), and apoptosis (trifluoperazine, loxapine, lithium, desipramine, nortriptyline, maprotiline, cyproheptadine, melatonin). In HD live mouse models, delayed disease onset (nortriptyline, melatonin), striatal preservation (haloperidol, tetrabenazine, lithium, sertraline), memory preservation (imipramine, trazodone, fluoxetine, sertraline, venlafaxine), motor improvement (tetrabenazine, lithium, valproate, imipramine, nortriptyline, trazodone, sertraline, venlafaxine), and extended survival (lithium, valproate, sertraline, melatonin) have been documented. Upregulated CREB binding protein (CBP; valproate, dextromethorphan) and downregulated histone deacetylase (HDAC; valproate) await demonstration in HD models. Most preclinical findings await replication and their limitations are reviewed. The most promising findings involve replicated striatal neuroprotection and phenotypic disease modification in transgenic mice for tetrabenazine and for sertraline. Clinical data consist of an uncontrolled lithium case series (n = 3) suggesting non-progression and a primarily negative double-blind, placebo-controlled clinical trial of lamotrigine.
Collapse
Affiliation(s)
- Edward C Lauterbach
- Department of Psychiatry and Behavioral Sciences, Mercer University School of Medicine, 655 First Street, Macon, GA 31201, USA.
| |
Collapse
|
44
|
Renal dopamine receptors, oxidative stress, and hypertension. Int J Mol Sci 2013; 14:17553-72. [PMID: 23985827 PMCID: PMC3794741 DOI: 10.3390/ijms140917553] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 12/22/2022] Open
Abstract
Dopamine, which is synthesized in the kidney, independent of renal nerves, plays an important role in the regulation of fluid and electrolyte balance and systemic blood pressure. Lack of any of the five dopamine receptor subtypes (D1R, D2R, D3R, D4R, and D5R) results in hypertension. D1R, D2R, and D5R have been reported to be important in the maintenance of a normal redox balance. In the kidney, the antioxidant effects of these receptors are caused by direct and indirect inhibition of pro-oxidant enzymes, specifically, nicotinamide adenine dinucleotide phosphate, reduced form (NADPH) oxidase, and stimulation of anti-oxidant enzymes, which can also indirectly inhibit NADPH oxidase activity. Thus, stimulation of the D2R increases the expression of endogenous anti-oxidants, such as Parkinson protein 7 (PARK7 or DJ-1), paraoxonase 2 (PON2), and heme oxygenase 2 (HO-2), all of which can inhibit NADPH oxidase activity. The D5R decreases NADPH oxidase activity, via the inhibition of phospholipase D2, and increases the expression of HO-1, another antioxidant. D1R inhibits NADPH oxidase activity via protein kinase A and protein kinase C cross-talk. In this review, we provide an overview of the protective roles of a specific dopamine receptor subtype on renal oxidative stress, the different mechanisms involved in this effect, and the role of oxidative stress and impairment of dopamine receptor function in the hypertension that arises from the genetic ablation of a specific dopamine receptor gene in mice.
Collapse
|
45
|
Chen JY, Wang EA, Cepeda C, Levine MS. Dopamine imbalance in Huntington's disease: a mechanism for the lack of behavioral flexibility. Front Neurosci 2013; 7:114. [PMID: 23847463 PMCID: PMC3701870 DOI: 10.3389/fnins.2013.00114] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 06/13/2013] [Indexed: 01/10/2023] Open
Abstract
Dopamine (DA) plays an essential role in the control of coordinated movements. Alterations in DA balance in the striatum lead to pathological conditions such as Parkinson's and Huntington's diseases (HD). HD is a progressive, invariably fatal neurodegenerative disease caused by a genetic mutation producing an expansion of glutamine repeats and is characterized by abnormal dance-like movements (chorea). The principal pathology is the loss of striatal and cortical projection neurons. Changes in brain DA content and receptor number contribute to abnormal movements and cognitive deficits in HD. In particular, during the early hyperkinetic stage of HD, DA levels are increased whereas expression of DA receptors is reduced. In contrast, in the late akinetic stage, DA levels are significantly decreased and resemble those of a Parkinsonian state. Time-dependent changes in DA transmission parallel biphasic changes in glutamate synaptic transmission and may enhance alterations in glutamate receptor-mediated synaptic activity. In this review, we focus on neuronal electrophysiological mechanisms that may lead to some of the motor and cognitive symptoms of HD and how they relate to dysfunction in DA neurotransmission. Based on clinical and experimental findings, we propose that some of the behavioral alterations in HD, including reduced behavioral flexibility, may be caused by altered DA modulatory function. Thus, restoring DA balance alone or in conjunction with glutamate receptor antagonists could be a viable therapeutic approach.
Collapse
Affiliation(s)
- Jane Y Chen
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior and the Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles Los Angeles, CA, USA
| | | | | | | |
Collapse
|
46
|
Jimenez-Shahed J, Jankovic J. Tetrabenazine for treatment of chorea associated with Huntington's disease and other potential indications. Expert Opin Orphan Drugs 2013. [DOI: 10.1517/21678707.2013.787358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
47
|
MAP kinase phosphatase 1 (MKP-1/DUSP1) is neuroprotective in Huntington's disease via additive effects of JNK and p38 inhibition. J Neurosci 2013; 33:2313-25. [PMID: 23392662 DOI: 10.1523/jneurosci.4965-11.2013] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We previously demonstrated that sodium butyrate is neuroprotective in Huntington's disease (HD) mice and that this therapeutic effect is associated with increased expression of mitogen-activated protein kinase/dual-specificity phosphatase 1 (MKP-1/DUSP1). Here we show that enhancing MKP-1 expression is sufficient to achieve neuroprotection in lentiviral models of HD. Wild-type MKP-1 overexpression inhibited apoptosis in primary striatal neurons exposed to an N-terminal fragment of polyglutamine-expanded huntingtin (Htt171-82Q), blocking caspase-3 activation and significantly reducing neuronal cell death. This neuroprotective effect of MKP-1 was demonstrated to be dependent on its enzymatic activity, being ablated by mutation of its phosphatase domain and being attributed to inhibition of specific MAP kinases (MAPKs). Overexpression of MKP-1 prevented the polyglutamine-expanded huntingtin-induced activation of c-Jun N-terminal kinases (JNKs) and p38 MAPKs, whereas extracellular signal-regulated kinase (ERK) 1/2 activation was not altered by either polyglutamine-expanded Htt or MKP-1. Moreover, mutants of MKP-1 that selectively prevented p38 or JNK binding confirmed the important dual contributions of p38 and JNK regulation to MKP-1-mediated neuroprotection. These results demonstrate additive effects of p38 and JNK MAPK inhibition by MKP-1 without consequence to ERK activation in this striatal neuron-based paradigm. MKP-1 also provided neuroprotection in vivo in a lentiviral model of HD neuropathology in rat striatum. Together, these data extend previous evidence that JNK- and p38-mediated pathways contribute to HD pathogenesis and, importantly, show that therapies simultaneously inhibiting both JNK and p38 signaling pathways may lead to improved neuroprotective outcomes.
Collapse
|
48
|
Ghiglieri V, Bagetta V, Calabresi P, Picconi B. Functional interactions within striatal microcircuit in animal models of Huntington's disease. Neuroscience 2012; 211:165-84. [DOI: 10.1016/j.neuroscience.2011.06.075] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 06/25/2011] [Accepted: 06/28/2011] [Indexed: 11/17/2022]
|
49
|
Gil-Mohapel JM. Screening of therapeutic strategies for Huntington's disease in YAC128 transgenic mice. CNS Neurosci Ther 2012; 18:77-86. [PMID: 21501423 DOI: 10.1111/j.1755-5949.2011.00246.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Huntington’s disease (HD) is a hereditary neurodegenerative disorder caused by an unstable expansion of cytosine-adenine-guanine (CAG) repeats in the HD gene. The symptoms include cognitive dysfunction and severe motor impairment with loss of voluntary movement coordination that is later replaced by bradykinesia and rigidity. The neuropathology is characterized by neuronal loss mainly in the striatum and cortex, and the appearance of neuronal intranuclear inclusions of mutant huntingtin. The mechanisms responsible for neurodegeneration are still not fully understood although excitotoxicity and a consequent increase in intracellular calcium concentration as well as the activation of caspases and calapins are known to play a key role. There is currently no satisfactory treatment or cure for this disease. The YAC128 transgenic mice express the full-length human HD gene with 128 CAG repeats and constitute a unique model for the study of HD as they replicate the slow and biphasic progression of behavioral deficits characteristic of the human condition and show striatal neuronal loss. As such, these transgenic mice have been an invaluable model not only for the elucidation of the neurodegenerative pathways in HD, but also for the screening and development of new therapeutic approaches. Here, I will review the unique characteristics of this transgenic HD model and will provide a summary of the therapies that have been tested in these mice, namely: potentiation of the protective roles of wild-type huntingtin and mutant huntingtin aggregation, transglutaminase inhibition, inhibition of glutamate- and dopamine-induced toxicity, apoptosis inhibition, use of essential fatty acids, and the novel approach of intrabody gene therapy. The insights obtained from these and future studies will help identify potential candidates for clinical trials and will ultimately contribute to the discovery of a successful treatment for this devastating neurodegenerative disorder.
Collapse
Affiliation(s)
- Joana M Gil-Mohapel
- Division of Medical Sciences, Island Medical Program, University of Victoria, British Columbia, Canada.
| |
Collapse
|
50
|
Ritch JJ, Valencia A, Alexander J, Sapp E, Gatune L, Sangrey GR, Sinha S, Scherber CM, Zeitlin S, Sadri-Vakili G, Irimia D, Difiglia M, Kegel KB. Multiple phenotypes in Huntington disease mouse neural stem cells. Mol Cell Neurosci 2012; 50:70-81. [PMID: 22508027 DOI: 10.1016/j.mcn.2012.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 03/09/2012] [Accepted: 03/29/2012] [Indexed: 11/25/2022] Open
Abstract
Neural stem (NS) cells are a limitless resource, and thus superior to primary neurons for drug discovery provided they exhibit appropriate disease phenotypes. Here we established NS cells for cellular studies of Huntington's disease (HD). HD is a heritable neurodegenerative disease caused by a mutation resulting in an increased number of glutamines (Q) within a polyglutamine tract in Huntingtin (Htt). NS cells were isolated from embryonic wild-type (Htt(7Q/7Q)) and "knock-in" HD (Htt(140Q/140Q)) mice expressing full-length endogenous normal or mutant Htt. NS cells were also developed from mouse embryonic stem cells that were devoid of Htt (Htt(-/-)), or knock-in cells containing human exon1 with an N-terminal FLAG epitope tag and with 7Q or 140Q inserted into one of the mouse alleles (Htt(F7Q/7Q) and Htt(F140Q/7Q)). Compared to Htt(7Q/7Q) NS cells, HD Htt(140Q/140Q) NS cells showed significantly reduced levels of cholesterol, increased levels of reactive oxygen species (ROS), and impaired motility. The heterozygous Htt(F140Q/7Q) NS cells had increased ROS and decreased motility compared to Htt(F7Q/7Q). These phenotypes of HD NS cells replicate those seen in HD patients or in primary cell or in vivo models of HD. Huntingtin "knock-out" NS cells (Htt(-/-)) also had impaired motility, but in contrast to HD cells had increased cholesterol. In addition, Htt(140Q/140Q) NS cells had higher phospho-AKT/AKT ratios than Htt(7Q/7Q) NS cells in resting conditions and after BDNF stimulation, suggesting mutant htt affects AKT dependent growth factor signaling. Upon differentiation, the Htt(7Q/7Q) and Htt(140Q/140Q) generated numerous Beta(III)-Tubulin- and GABA-positive neurons; however, after 15 days the cellular architecture of the differentiated Htt(140Q/140Q) cultures changed compared to Htt(7Q/7Q) cultures and included a marked increase of GFAP-positive cells. Our findings suggest that NS cells expressing endogenous mutant Htt will be useful for study of mechanisms of HD and drug discovery.
Collapse
Affiliation(s)
- James J Ritch
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|