1
|
Campesan S, Del Popolo I, Marcou K, Straatman-Iwanowska A, Repici M, Boytcheva KV, Cotton VE, Allcock N, Rosato E, Kyriacou CP, Giorgini F. Bypassing mitochondrial defects rescues Huntington's phenotypes in Drosophila. Neurobiol Dis 2023; 185:106236. [PMID: 37495179 DOI: 10.1016/j.nbd.2023.106236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/06/2023] [Accepted: 07/22/2023] [Indexed: 07/28/2023] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disease with limited treatment options. Human and animal studies have suggested that metabolic and mitochondrial dysfunctions contribute to HD pathogenesis. Here, we use high-resolution respirometry to uncover defective mitochondrial oxidative phosphorylation and electron transfer capacity when a mutant huntingtin fragment is targeted to neurons or muscles in Drosophila and find that enhancing mitochondrial function can ameliorate these defects. In particular, we find that co-expression of parkin, an E3 ubiquitin ligase critical for mitochondrial dynamics and homeostasis, produces significant enhancement of mitochondrial respiration when expressed either in neurons or muscles, resulting in significant rescue of neurodegeneration, viability and longevity in HD model flies. Targeting mutant HTT to muscles results in larger mitochondria and higher mitochondrial mass, while co-expression of parkin increases mitochondrial fission and decreases mass. Furthermore, directly addressing HD-mediated defects in the fly's mitochondrial electron transport system, by rerouting electrons to either bypass mitochondrial complex I or complexes III-IV, significantly increases mitochondrial respiration and results in a striking rescue of all phenotypes arising from neuronal mutant huntingtin expression. These observations suggest that bypassing impaired mitochondrial respiratory complexes in HD may have therapeutic potential for the treatment of this devastating disorder.
Collapse
Affiliation(s)
- Susanna Campesan
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK.
| | - Ivana Del Popolo
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Kyriaki Marcou
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Anna Straatman-Iwanowska
- Electron Microscopy Facility, Core Biotechnology Services, Adrian Building, University of Leicester, University Road, Leicester LE1 7RH, Leicestershire, UK
| | - Mariaelena Repici
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK; School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Kalina V Boytcheva
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Victoria E Cotton
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Natalie Allcock
- Electron Microscopy Facility, Core Biotechnology Services, Adrian Building, University of Leicester, University Road, Leicester LE1 7RH, Leicestershire, UK
| | - Ezio Rosato
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Charalambos P Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK.
| |
Collapse
|
2
|
Jiang A, Handley RR, Lehnert K, Snell RG. From Pathogenesis to Therapeutics: A Review of 150 Years of Huntington's Disease Research. Int J Mol Sci 2023; 24:13021. [PMID: 37629202 PMCID: PMC10455900 DOI: 10.3390/ijms241613021] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative genetic disorder caused by an expanded polyglutamine-coding (CAG) trinucleotide repeat in the huntingtin (HTT) gene. HD behaves as a highly penetrant dominant disorder likely acting through a toxic gain of function by the mutant huntingtin protein. Widespread cellular degeneration of the medium spiny neurons of the caudate nucleus and putamen are responsible for the onset of symptomology that encompasses motor, cognitive, and behavioural abnormalities. Over the past 150 years of HD research since George Huntington published his description, a plethora of pathogenic mechanisms have been proposed with key themes including excitotoxicity, dopaminergic imbalance, mitochondrial dysfunction, metabolic defects, disruption of proteostasis, transcriptional dysregulation, and neuroinflammation. Despite the identification and characterisation of the causative gene and mutation and significant advances in our understanding of the cellular pathology in recent years, a disease-modifying intervention has not yet been clinically approved. This review includes an overview of Huntington's disease, from its genetic aetiology to clinical presentation and its pathogenic manifestation. An updated view of molecular mechanisms and the latest therapeutic developments will also be discussed.
Collapse
Affiliation(s)
- Andrew Jiang
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand; (R.R.H.); (K.L.); (R.G.S.)
| | | | | | | |
Collapse
|
3
|
Norris SA, Tian L, Williams EL, Perlmutter JS. Transient dystonia correlates with parkinsonism after 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine in nonhuman primates. DYSTONIA 2023; 2:11019. [PMID: 37711667 PMCID: PMC10501383 DOI: 10.3389/dyst.2023.11019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Unilateral internal carotid artery 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) infusion in non-human primates produces transient contralateral hemi-dystonia followed by stable contralateral hemi-parkinsonism; the relationship between dystonia and parkinsonism remains unclear. We hypothesized that transient dystonia severity following MPTP correlates with parkinsonism severity. In male Macaca nemestrina (n = 3) and M. fascicularis (n = 17) we administered unilateral intra-carotid MPTP, then correlated validated blinded ratings of transient peak dystonia and delayed parkinsonism. We also correlated dystonia severity with post-mortem measures of residual striatal dopamine and nigral neuron counts obtained a mean 53 ± 15 days following MPTP, after resolution of dystonia but during stable parkinsonism. Median latency to dystonia onset was 1 day, and peak severity 2.5 days after MPTP; total dystonia duration was 13.5 days. Parkinsonism peaked a median of 19.5 days after MPTP, remaining nearly constant thereafter. Peak dystonia severity highly correlated with parkinsonism severity (r[18] = 0.82, p < 0.001). Residual cell counts in lesioned nigra correlated linearly with peak dystonia scores (r[18] = -0.68, p=<0.001). Dystonia was not observed in monkeys without striatal dopamine depletion (n = 2); dystonia severity correlated with striatal dopamine depletion when residual nigral cell loss was less than 50% ([11] r = -0.83, p < 0.001) but spanned a broad range with near complete striatal dopamine depletion, when nigral cell loss was greater than 50%. Our data indicate that residual striatal dopamine may not reflect dystonia severity. We speculate on mechanisms of transient dystonia followed by parkinsonism that may be studied using this particular NHP MPTP model to better understand relationships of transient dystonia to nigrostriatal injury and parkinsonism.
Collapse
Affiliation(s)
- S. A. Norris
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - L. Tian
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - E. L. Williams
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - J. S. Perlmutter
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neuroscience, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Physical Therapy, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Occupational Therapy, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
4
|
Bénit P, Goncalves J, El Khoury R, Rak M, Favier J, Gimenez-Roqueplo AP, Rustin P. Succinate Dehydrogenase, Succinate, and Superoxides: A Genetic, Epigenetic, Metabolic, Environmental Explosive Crossroad. Biomedicines 2022; 10:1788. [PMID: 35892689 PMCID: PMC9394281 DOI: 10.3390/biomedicines10081788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Research focused on succinate dehydrogenase (SDH) and its substrate, succinate, culminated in the 1950s accompanying the rapid development of research dedicated to bioenergetics and intermediary metabolism. This allowed researchers to uncover the implication of SDH in both the mitochondrial respiratory chain and the Krebs cycle. Nowadays, this theme is experiencing a real revival following the discovery of the role of SDH and succinate in a subset of tumors and cancers in humans. The aim of this review is to enlighten the many questions yet unanswered, ranging from fundamental to clinically oriented aspects, up to the danger of the current use of SDH as a target for a subclass of pesticides.
Collapse
Affiliation(s)
- Paule Bénit
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| | - Judith Goncalves
- Paris Centre de Recherche Cardiovasculaire (PARCC), Inserm, Université Paris Cité, F-75015 Paris, France; (J.G.); (J.F.)
| | - Riyad El Khoury
- Department of Pathology and Laboratory Medicine, Neuromuscular Diagnostic Laboratory, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Malgorzata Rak
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| | - Judith Favier
- Paris Centre de Recherche Cardiovasculaire (PARCC), Inserm, Université Paris Cité, F-75015 Paris, France; (J.G.); (J.F.)
| | - Anne-Paule Gimenez-Roqueplo
- Département de Médecine Génomique des Tumeurs et des Cancers, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, F-75015 Paris, France;
| | - Pierre Rustin
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| |
Collapse
|
5
|
Mustafa AM, Rabie MA, Zaki HF, Shaheen AM. Inhibition of Brain GTP Cyclohydrolase I Attenuates 3-Nitropropionic Acid-Induced Striatal Toxicity: Involvement of Mas Receptor/PI3k/Akt/CREB/ BDNF Axis. Front Pharmacol 2022; 12:740966. [PMID: 35002694 PMCID: PMC8727546 DOI: 10.3389/fphar.2021.740966] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
GTP cyclohydrolase I (GTPCH I) is the rate-limiting enzyme for tetrahydrobiopterin (BH4) biosynthesis; the latter is an essential factor for iNOS activation that contributes neuronal loss in Huntington’s disease (HD). The aim of the study was to investigate the neuroprotective effect of 2,4-diamino-6-hydroxypyrimidine (DAHP), GTPCH I enzyme inhibitor, against neuronal loss in 3-nitropropinic acid (3-NP)-induced HD in rats and to reveal the possible involved mechanisms mediated through PI3K/Akt axis and its correlation to Mas receptor (MasR). Rats received 3-NP (10 mg/kg/day; i.p.) with or without administration of DAHP (0.5 g/kg/day; i.p.) or wortmannin (WM), a PI3K inhibitor, (15 μg/kg/day; i.v.) for 14 days. DAHP improved cognitive, memory, and motor abnormalities induced by 3-NP, as confirmed by striatal histopathological specimens and immunohistochemical examination of GFAP. Moreover, DAHP treatment inhibited GTPCH I activity, resulting in decreased BH4 levels and iNOS activation. Also, DAHP upregulated the protein expression of survival protein; p85/p55 (pY458/199)-PI3K and pS473-Akt that, in turn, boosted the activation of striatal neurotrophic factors and receptor, pS133-CREB, BDNF and pY515-TrKB, which positively affect MasR protein expression and improve mitochondrial dysfunction, as indicated by enhancing both SDH and PGC-1α levels. Indeed, DAHP attenuates oxidative stress by increasing SOD activity and Nrf2 expression in addition to reducing neuro-inflammatory status by inhibiting NF-κB p65 and TNF-α expression. Interestingly, all the previous effects were blocked by co-administration of WM with DAHP. In conclusion, DAHP exerts neuroprotective effect against neuronal loss induced by 3-NP administration via inhibition of GTPCH I and iNOS activity and activation of MasR/PI3K/Akt/CREB/BDNF/TrKB axis besides its antioxidant and anti-inflammatory effect.
Collapse
Affiliation(s)
- Aya M Mustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aya M Shaheen
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
6
|
Klonarakis M, De Vos M, Woo E, Ralph L, Thacker JS, Gil-Mohapel J. The three sisters of fate: Genetics, pathophysiology and outcomes of animal models of neurodegenerative diseases. Neurosci Biobehav Rev 2022; 135:104541. [DOI: 10.1016/j.neubiorev.2022.104541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/28/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023]
|
7
|
Dietrich P, Alli S, Mulligan MK, Cox R, Ashbrook DG, Williams RW, Dragatsis I. Identification of cyclin D1 as a major modulator of 3-nitropropionic acid-induced striatal neurodegeneration. Neurobiol Dis 2022; 162:105581. [PMID: 34871739 PMCID: PMC8717869 DOI: 10.1016/j.nbd.2021.105581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/14/2021] [Accepted: 12/02/2021] [Indexed: 01/03/2023] Open
Abstract
Mitochondria dysfunction occurs in the aging brain as well as in several neurodegenerative disorders and predisposes neuronal cells to enhanced sensitivity to neurotoxins. 3-nitropropionic acid (3-NP) is a naturally occurring plant and fungal neurotoxin that causes neurodegeneration predominantly in the striatum by irreversibly inhibiting the tricarboxylic acid respiratory chain enzyme, succinate dehydrogenase (SDH), the main constituent of the mitochondria respiratory chain complex II. Significantly, although 3-NP-induced inhibition of SDH occurs in all brain regions, neurodegeneration occurs primarily and almost exclusively in the striatum for reasons still not understood. In rodents, 3-NP-induced striatal neurodegeneration depends on the strain background suggesting that genetic differences among genotypes modulate toxicant variability and mechanisms that underlie 3-NP-induced neuronal cell death. Using the large BXD family of recombinant inbred (RI) strains we demonstrate that variants in Ccnd1 - the gene encoding cyclin D1 - of the DBA/2 J parent underlie the resistance to 3-NP-induced striatal neurodegeneration. In contrast, the Ccnd1 variant inherited from the widely used C57BL/6 J parental strain confers sensitivity. Given that cellular stress triggers induction of cyclin D1 expression followed by cell-cycle re-entry and consequent neuronal cell death, we sought to determine if the C57BL/6 J and DBA/2 J Ccnd1 variants are differentially modulated in response to 3-NP. We confirm that 3-NP induces cyclin D1 expression in striatal neuronal cells of C57BL/6 J, but this response is blunted in the DBA/2 J. We further show that striatal-specific alternative processing of a highly conserved 3'UTR negative regulatory region of Ccnd1 co-segregates with the C57BL/6 J parental Ccnd1 allele in BXD strains and that its differential processing accounts for sensitivity or resistance to 3-NP. Our results indicate that naturally occurring Ccnd1 variants may play a role in the variability observed in neurodegenerative disorders involving mitochondria complex II dysfunction and point to cyclin D1 as a possible therapeutic target.
Collapse
Affiliation(s)
- Paula Dietrich
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, TN 38163, USA,Corresponding authors: ,
| | - Shanta Alli
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, TN 38163, USA
| | - Megan K. Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee, Health Science Center, Memphis, TN 38163, USA
| | - Rachel Cox
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, TN 38163, USA,The University of Tennessee, Knoxville, TN 37996, USA
| | - David G. Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee, Health Science Center, Memphis, TN 38163, USA
| | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee, Health Science Center, Memphis, TN 38163, USA
| | - Ioannis Dragatsis
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, TN 38163, USA,Corresponding authors: ,
| |
Collapse
|
8
|
Nguyen VTT, König S, Eggert S, Endres K, Kins S. The role of mycotoxins in neurodegenerative diseases: current state of the art and future perspectives of research. Biol Chem 2021; 403:3-26. [PMID: 34449171 DOI: 10.1515/hsz-2021-0214] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/16/2021] [Indexed: 01/02/2023]
Abstract
Mycotoxins are fungal metabolites that can cause various diseases in humans and animals. The adverse health effects of mycotoxins such as liver failure, immune deficiency, and cancer are well-described. However, growing evidence suggests an additional link between these fungal metabolites and neurodegenerative diseases. Despite the wealth of these initial reports, reliable conclusions are still constrained by limited access to human patients and availability of suitable cell or animal model systems. This review summarizes knowledge on mycotoxins associated with neurodegenerative diseases and the assumed underlying pathophysiological mechanisms. The limitations of the common in vivo and in vitro experiments to identify the role of mycotoxins in neurotoxicity and thereby in neurodegenerative diseases are elucidated and possible future perspectives to further evolve this research field are presented.
Collapse
Affiliation(s)
- Vu Thu Thuy Nguyen
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Untere Zahlbacher Str. 8, D-55131 Mainz, Germany
| | - Svenja König
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| | - Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Untere Zahlbacher Str. 8, D-55131 Mainz, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| |
Collapse
|
9
|
Darby S, Sanchez LC, Mallicote MF, House AM, Plummer CE, Nadruz V, Benmoha RH, Roberts SM, Derendorf H, Silva-Sanchez C, Claire J, MacKay RJ. Plasma l-indospicine and 3-nitropropionic acid in ponies fed creeping indigo: Comparison with results from an episode of presumptive creeping indigo toxicosis. Equine Vet J 2020; 54:145-152. [PMID: 33369767 DOI: 10.1111/evj.13415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/20/2020] [Accepted: 12/17/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Creeping indigo (Indigofera spicata) toxicosis is an emerging problem among horses in Florida and bordering states. OBJECTIVES To quantify the putative toxins l-indospicine (IND) and 3-nitropropionic acid (NPA) in creeping indigo collected from multiple sites and to measure plasma toxin concentrations in ponies fed creeping indigo and horses with presumptive creeping indigo toxicosis. STUDY DESIGN Experimental descriptive study with descriptive observational field investigation. METHODS Air-dried creeping indigo was assayed for IND and NPA content. Five ponies were fed chopped creeping indigo containing 1 mg/kg/day of IND and trace amounts of NPA for 5 days, then observed for 28 days. Blood samples from these ponies and from horses involved in a presumptive creeping indigo toxicosis were assayed for IND and NPA. RESULTS IND in creeping indigo plants was 0.4-3.5 mg/g dry matter whereas NPA was <0.01 to 0.03 mg/g. During creeping indigo feeding, clinical and laboratory signs were unchanged except for significant weight loss (median 6%, range 2%-9%; p = .04) and significant increase from baseline plasma protein concentration (median 16 g/L, range 8-25 g/L; p < .001). These changes could not definitively be ascribed to creeping indigo ingestion. Plasma IND rose to 3.9 ± 0.52 mg/L on day 6. Pharmacokinetic modelling indicated an elimination half-life of 25 days and a steady state plasma concentration of 22 mg/L. Plasma IND concentration in sick horses during an incident of creeping indigo toxicosis was approximately twice that of clinically normal pasture mates. Plasma NPA was <0.05 mg/L in all samples. MAIN LIMITATIONS Creeping indigo used in the feeding trial may not be representative of plants involved in creeping indigo toxicosis. There was no control group without creeping indigo in the feeding trial. CONCLUSIONS Indospicine can be detected in blood of horses consuming creeping indigo and the toxin accumulates in tissues and clears slowly. The role of NPA in the neurological signs of this syndrome is unclear.
Collapse
Affiliation(s)
- Shannon Darby
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - L Chris Sanchez
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Martha F Mallicote
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Amanda M House
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Caryn E Plummer
- Departments of Small and Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Veridiana Nadruz
- Kansas State University Veterinary Health Center, Manhattan, KS, USA
| | - Rachel H Benmoha
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Stephen M Roberts
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Hartmut Derendorf
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Cecilia Silva-Sanchez
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Jami Claire
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Robert J MacKay
- Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| |
Collapse
|
10
|
Neueder A, Orth M. Mitochondrial biology and the identification of biomarkers of Huntington's disease. Neurodegener Dis Manag 2020; 10:243-255. [PMID: 32746707 DOI: 10.2217/nmt-2019-0033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Apart from finding novel compounds for treating Huntington's disease (HD) an important challenge at present consists in finding reliable read-outs or biomarkers that reflect key biological processes involved in HD pathogenesis. The core elements of HD biology, for example, HTT RNA levels or protein species can serve as biomarker, as could measures from biological systems or pathways in which Huntingtin plays an important role. Here we review the evidence for the involvement of mitochondrial biology in HD. The most consistent findings pertain to mitochondrial quality control, for example, fission/fusion. However, a convincing mitochondrial signature with biomarker potential is yet to emerge. This requires more research including in peripheral sources of human material, such as blood, or skeletal muscle.
Collapse
Affiliation(s)
| | - Michael Orth
- Department of Neurology, Ulm University, Ulm, Germany.,SwissHuntington's Disease Centre, Neurozentrum Siloah, Worbstr. 312, 3073 Gümligenbei Bern, Switzerland
| |
Collapse
|
11
|
Wiprich MT, Zanandrea R, Altenhofen S, Bonan CD. Influence of 3-nitropropionic acid on physiological and behavioral responses in zebrafish larvae and adults. Comp Biochem Physiol C Toxicol Pharmacol 2020; 234:108772. [PMID: 32353558 DOI: 10.1016/j.cbpc.2020.108772] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 12/13/2022]
Abstract
Long-term treatment with 3-nitropropionic acid (3-NPA), a toxin derived from plants and fungi, may reproduce symptoms and biochemical characteristics of Huntington's disease (HD). Our study evaluated the effects of 3-NPA on the physiological and behavioral responses in zebrafish larvae and adults. Larvae exposed to 0.1, 0.2, or 0.5 mM 3-NPA exhibited an increase in heart rate at 2- and 5-days post-fertilization (dpf). There was a decrease in the ocular distance at 5 dpf with 0.05 mM 3-NPA treatment. However, 3-NPA did not alter larval locomotor parameters. Adult zebrafish received 3-NPA intraperitoneal injections (a total of seven injections at doses 10, 20, or 60 mg/kg every 96 h) and showed a decrease in body weight , locomotion and aggressive behavior. No changes were observed in anxiety-like behavior and social interaction between 3-NPA-exposed animals and control groups. However, 3-NPA-treated animals (at 60 mg/kg) demonstrated impaired long-term aversive memory. Overall, 3-NPA exposure induced morphological and heart rate alterations in zebrafish larvae. Additionally, our study showed behavioral changes in zebrafish that were submitted to long-term 3-NPA treatment, which could be related to HD symptoms.
Collapse
Affiliation(s)
- Melissa Talita Wiprich
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rodrigo Zanandrea
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Stefani Altenhofen
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Cerebrais, Excitotoxicidade e Neuroproteção, Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Cerebrais, Excitotoxicidade e Neuroproteção, Porto Alegre, RS, Brazil.
| |
Collapse
|
12
|
Age-related cognitive decline in baboons: modeling the prodromal phase of Alzheimer's disease and related dementias. Aging (Albany NY) 2020; 12:10099-10116. [PMID: 32427127 PMCID: PMC7346018 DOI: 10.18632/aging.103272] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/01/2020] [Indexed: 12/12/2022]
Abstract
The aging of brain cells and synaptic loss are the major underlying pathophysiological processes contributing to the progressive decline in cognitive functions and Alzheimer’s disease. The difference in cognitive performances observed between adult and aged subjects across species highlights the decline of brain systems with age. The inflection point in age-related cognitive decline is important for our understanding of the pathophysiology of neurodegenerative diseases and for timing therapeutic interventions. Humans and nonhuman primates share many similarities including age-dependent changes in gene expression and decline in neural and immune functions. Given these evolutionary conserved organ systems, complex human-like behavioral and age-dependent changes may be modeled and monitored longitudinally in nonhuman primates. We integrated three clinically relevant outcome measures to investigate the effect of age on cognition, motor function and diurnal activity in aged baboons. We provide evidence of a naturally-occurring age-dependent precipitous decline in movement planning, in learning novel tasks, in simple discrimination and in motivation. These results suggest that baboons aged ~20 years (equivalent to ~60 year old humans) may offer a relevant model for the prodromal phase of Alzheimer’s disease and related dementias to investigate mechanisms involved in the precipitous decline in cognitive functions and to develop early therapeutic interventions
Collapse
|
13
|
Camberos-Luna L, Massieu L. Therapeutic strategies for ketosis induction and their potential efficacy for the treatment of acute brain injury and neurodegenerative diseases. Neurochem Int 2019; 133:104614. [PMID: 31785349 DOI: 10.1016/j.neuint.2019.104614] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
The therapeutic use of ketone bodies (KB) against acute brain injury and neurodegenerative disorders has lately been suggested by many studies. Several mechanisms responsible for the protective action of KB have been described, including metabolic, anti-inflammatory and epigenetic. However, it is still not clear whether a specific mechanism of action can be associated with a particular neurological disorder. Different strategies to induce ketosis including the ketogenic diet (KD), caloric restriction (CR), intermittent fasting (IF), as well as the administration of medium chain triglycerides (MCTs), exogenous ketones or KB derivatives, have been used in animal models of brain injury and in humans. They have shown different degrees of success to prevent neuronal damage, motor alterations and cognitive decline. However, more investigation is needed in order to establish safe protocols for clinical application. Throughout the present review, we describe the different approaches that have been used to elevate blood KB and discuss their effectiveness considering their advantages and limitations, as tested in models of brain injury, neurodegeneration and clinical research. We also describe the mechanisms of action of KB in non-pathologic conditions and in association with their protective effect against neuronal damage in acute neurological disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Lucy Camberos-Luna
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, CP 04510, Mexico.
| | - Lourdes Massieu
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, CP 04510, Mexico.
| |
Collapse
|
14
|
Lavisse S, Williams S, Lecourtois S, van Camp N, Guillermier M, Gipchtein P, Jan C, Goutal S, Eymin L, Valette J, Delzescaux T, Perrier AL, Hantraye P, Aron Badin R. Longitudinal characterization of cognitive and motor deficits in an excitotoxic lesion model of striatal dysfunction in non-human primates. Neurobiol Dis 2019; 130:104484. [PMID: 31132407 DOI: 10.1016/j.nbd.2019.104484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
As research progresses in the understanding of the molecular and cellular mechanisms underlying neurodegenerative diseases like Huntington's disease (HD) and expands towards preclinical work for the development of new therapies, highly relevant animal models are increasingly needed to test new hypotheses and to validate new therapeutic approaches. In this light, we characterized an excitotoxic lesion model of striatal dysfunction in non-human primates (NHPs) using cognitive and motor behaviour assessment as well as functional imaging and post-mortem anatomical analyses. NHPs received intra-striatal stereotaxic injections of quinolinic acid bilaterally in the caudate nucleus and unilaterally in the left sensorimotor putamen. Post-operative MRI scans showed atrophy of the caudate nucleus and a large ventricular enlargement in all 6 NHPs that correlated with post-mortem measurements. Behavioral analysis showed deficits in 2 analogues of the Wisconsin card sorting test (perseverative behavior) and in an executive task, while no deficits were observed in a visual recognition or an episodic memory task at 6 months following surgery. Spontaneous locomotor activity was decreased after lesion and the incidence of apomorphine-induced dyskinesias was significantly increased at 3 and 6 months following lesion. Positron emission tomography scans obtained at end-point showed a major deficit in glucose metabolism and D2 receptor density limited to the lesioned striatum of all NHPs compared to controls. Post-mortem analyses revealed a significant loss of medium-sized spiny neurons in the striatum, a loss of neurons and fibers in the globus pallidus, a unilateral decrease in dopaminergic neurons of the substantia nigra and a loss of neurons in the motor and dorsolateral prefrontal cortex. Overall, we show that this robust NHP model presents specific behavioral (learning, execution and retention of cognitive tests) and metabolic functional deficits that, to the best of our knowledge, are currently not mimicked in any available large animal model of striatal dysfunction. Moreover, we used non-invasive, translational techniques like behavior and imaging to quantify such deficits and found that they correlate to a significant cell loss in the striatum and its main input and output structures. This model can thus significantly contribute to the pre-clinical longitudinal evaluation of the ability of new therapeutic cell, gene or pharmacotherapy approaches in restoring the functionality of the striatal circuitry.
Collapse
Affiliation(s)
- Sonia Lavisse
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Susannah Williams
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Sophie Lecourtois
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Nadja van Camp
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Martine Guillermier
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Pauline Gipchtein
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Caroline Jan
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Sébastien Goutal
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Leopold Eymin
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Julien Valette
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Thierry Delzescaux
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Anselme L Perrier
- Inserm U861, I-STEM, AFM, Corbeil-Essonnes 91100, cedex, France; UEVE U861, I-STEM, AFM, Corbeil-Essonnes 91100, cedex, France.
| | - Philippe Hantraye
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Romina Aron Badin
- MIRCen, CEA/IBFJ/DRF/LMN, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France; UMR CEA CNRS 9199-Université Paris Saclay, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
15
|
Interleukin-1β Protects Neurons against Oxidant-Induced Injury via the Promotion of Astrocyte Glutathione Production. Antioxidants (Basel) 2018; 7:antiox7080100. [PMID: 30044427 PMCID: PMC6115796 DOI: 10.3390/antiox7080100] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/04/2018] [Accepted: 07/21/2018] [Indexed: 01/13/2023] Open
Abstract
Interleukin-1β (IL-1β), a key cytokine that drives neuroinflammation in the Central Nervous System (CNS), is enhanced in many neurological diseases/disorders. Although IL-1β contributes to and/or sustains pathophysiological processes in the CNS, we recently demonstrated that IL-1β can protect cortical astrocytes from oxidant injury in a glutathione (GSH)-dependent manner. To test whether IL-1β could similarly protect neurons against oxidant stress, near pure neuronal cultures or mixed cortical cell cultures containing neurons and astrocytes were exposed to the organic peroxide, tert-butyl hydroperoxide (t-BOOH), following treatment with IL-1β or its vehicle. Neurons and astrocytes in mixed cultures, but not pure neurons, were significantly protected from the toxicity of t-BOOH following treatment with IL-1β in association with enhanced GSH production/release. IL-1β failed to increase the GSH levels or to provide protection against t-BOOH toxicity in chimeric mixed cultures consisting of IL-1R1+/+ neurons plated on top of IL-1R1−/− astrocytes. The attenuation of GSH release via block of multidrug resistance-associated protein 1 (MRP1) transport also abrogated the protective effect of IL-1β. These protective effects were not strictly an in vitro phenomenon as we found an increased striatal vulnerability to 3-nitropropionic acid-mediated oxidative stress in IL-1R1 null mice. Overall, our data indicate that IL-1β protects neurons against oxidant injury and that this likely occurs in a non-cell-autonomous manner that relies on an increase in astrocyte GSH production and release.
Collapse
|
16
|
Aron Badin R. Nonhuman Primate Models of Huntington's Disease and Their Application in Translational Research. Methods Mol Biol 2018; 1780:267-284. [PMID: 29856024 DOI: 10.1007/978-1-4939-7825-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Huntington's disease (HD) is a monogenic, autosomal dominant inherited fatal disease that affects 1 in 10,000 people worldwide. Given its unique genetic characteristics, HD would appear as one of the most straightforward neurodegenerative diseases to replicate in animal models. Indeed, mutations in the HTT gene have been used to generate a variety of animal models that display differential pathologies and have significantly increased our understanding of the pathological mechanisms of HD. However, decades of efforts have also shown the complexity of recapitulating the human condition in other species. Here we describe the three different types of models that have been generated in nonhuman primate species, stating their advantages and limitations and attempt to give a critical perspective of their translational value to test the efficacy of novel therapeutic strategies. Obtaining construct, phenotypic, and predictive validity has proven to be challenging in most animal models of human diseases. In HD in particular, it is hard to assess the predictive validity of a new therapeutic strategy when no effective "benchmark" treatment is available in the clinic. In this light, only phenotypic/face validity and construct validity are discussed.
Collapse
Affiliation(s)
- Romina Aron Badin
- Commissariat à l'Energie Atomique (CEA), Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses, France.
- Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France.
| |
Collapse
|
17
|
Emborg ME. Nonhuman Primate Models of Neurodegenerative Disorders. ILAR J 2017; 58:190-201. [PMID: 28985333 PMCID: PMC5886328 DOI: 10.1093/ilar/ilx021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 05/18/2017] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's (AD), Huntington's (HD), and Parkinson's (PD) disease are age-related neurodegenerative disorders characterized by progressive neuronal cell death. Although each disease has particular pathologies and symptoms, accumulated evidence points to similar mechanisms of neurodegeneration, including inflammation, oxidative stress, and protein aggregation. A significant body of research is ongoing to understand how these pathways affect each other and what ultimately triggers the onset of the disease. Experiments in nonhuman primates (NHPs) account for only 5% of all research in animals. Yet the impact of NHP studies for clinical translation is much greater, especially for neurodegenerative disorders, as NHPs have a complex cognitive and motor functions and highly developed neuroanatomy. New NHP models are emerging to better understand pathology and improve the platform in which to test novel therapies. The goal of this report is to review NHP models of AD, HD, and PD in the context of the current understanding of these diseases and their contribution to the development of novel therapies.
Collapse
Affiliation(s)
- Marina E Emborg
- Marina E. Emborg, MD, PhD, is the director of the Preclinical Parkinson’s Research Program at the Wisconsin National Primate Research Center and an associate professor in the department of Medical Physics at the University of Wisconsin in Madison, Wisconsin.
| |
Collapse
|
18
|
Alarcón-Herrera N, Flores-Maya S, Bellido B, García-Bores AM, Mendoza E, Ávila-Acevedo G, Hernández-Echeagaray E. Protective effects of chlorogenic acid in 3-nitropropionic acid induced toxicity and genotoxicity. Food Chem Toxicol 2017; 109:1018-1025. [DOI: 10.1016/j.fct.2017.04.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/29/2017] [Accepted: 04/30/2017] [Indexed: 01/28/2023]
|
19
|
Mochel F. Triheptanoin for the treatment of brain energy deficit: A 14-year experience. J Neurosci Res 2017; 95:2236-2243. [PMID: 28688166 DOI: 10.1002/jnr.24111] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 06/10/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022]
Abstract
Triheptanoin is an odd-chain triglyceride with anaplerotic properties-that is, replenishing the pool of metabolic intermediates in the Krebs cycle. Unlike even-chain fatty acids metabolized to acetyl-CoA only, triheptanoin can indeed provide both acetyl-CoA and propionyl-CoA, two key carbon sources for the Krebs cycle. Triheptanoin was initially used in patients with long-chain fatty acid oxidation disorders. The first demonstration of the possible benefit of triheptanoin for brain energy deficit came from a patient with pyruvate carboxylase deficiency, a severe metabolic disease that affects anaplerosis in the brain. In an open-label study, triheptanoin was then shown to decrease nonepileptic paroxysmal manifestations by 90% in patients with glucose transporter 1 deficiency syndrome, a disease that affects glucose transport into the brain. 31 P magnetic resonance spectroscopy studies also indicated that triheptanoin was able to correct bioenergetics in the brain of patients with Huntington disease, a neurodegenerative disease associated with brain energy deficit. Altogether, these studies indicate that triheptanoin can be a treatment for brain energy deficit related to altered anaplerosis and/or glucose metabolism. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fanny Mochel
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,AP-HP, Pitié-Salpêtrière University Hospital, Department of Genetics, Paris, France.,University Pierre and Marie Curie, Neurometabolic Research Group, Paris, France
| |
Collapse
|
20
|
Suganya SN, Sumathi T. Effect of rutin against a mitochondrial toxin, 3-nitropropionicacid induced biochemical, behavioral and histological alterations-a pilot study on Huntington's disease model in rats. Metab Brain Dis 2017; 32:471-481. [PMID: 27928694 DOI: 10.1007/s11011-016-9929-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 11/08/2016] [Indexed: 12/13/2022]
Abstract
Dietary compounds like flavonoids may offer protection against neurodegeneration. Huntington's disease (HD) is a neurodegenerative disorder characterized by symptoms like chorea and dementia. 3-Nitropropionic acid (3-NP), a Succinate dehydrogenase (SDH) inhibitor produces behavioral, biochemical and histological changes in the striatum, mimics HD in animals and humans. The present study was designed to examine the protective activity of Rutin (RT), a primary flavonoid from citrus fruits, green tea on 3-NP induced experimental model of HD in rats. Rats were pretreated with Rutin, a potent antioxidant (25 and 50 mg/kg b.w.) orally prior to the intraperitoneally (i.p.) administration of 3-NP (10 mg/kg b.w.) for 14 days. Behavioral assessments were carried out on 5th, 10th and 15th day after 3-NP treatment. Body weight, biochemical and histological studies were analyzed on 15th day. Systemic administration of 3-NP significantly reduced the body weight, locomotor activities (Rota rod, Open field test), memory (Morris water maze) and antioxidants such as Glutathione (GSH) levels, activities of Superoxide dismutase (SOD), Catalase (CAT), Glutathione peroxidase (GPx), Glutathione-S-transferase (GST), Glutathione reductase (GR). 3-NP also produces striatal damage by increased the levels of lipid peroxides, nitrite, Glial Fibrillary Acidic Protein (GFAP) and activity of Acetylcholine esterase (AchE). Thus, Rutin treatment of 25 and 50 mg/kg b.w. has significantly restored all the biochemical, behavioral and histological alterations caused by the 3-NP through its antioxidant activity. The findings of our study indicates that Rutin may have an important role in protecting the striatum from oxidative/nitrosative insults caused by 3-NP. These results suggest that RT might be a drug of choice to treat HD.
Collapse
Affiliation(s)
- Sarumani Natarajan Suganya
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600113, India
| | - Thangarajan Sumathi
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, 600113, India.
| |
Collapse
|
21
|
He Y, Akumuo RC, Yang Y, Hewett SJ. Mice deficient in L-12/15 lipoxygenase show increased vulnerability to 3-nitropropionic acid neurotoxicity. Neurosci Lett 2017; 643:65-69. [PMID: 28229935 DOI: 10.1016/j.neulet.2017.02.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 01/21/2023]
Abstract
Considerable evidence supports a contributory role for leukocyte-type 12/15 Lipoxygenase (L-12/15 LO) in mediating hippocampal and cortical neuronal injury in models of Alzheimer's disease and stroke. Whether L-12/15 LO contributes to neuronal injury in a model of Huntington's disease (HD) has yet to be determined. HD is characterized by marked striatal neuronal loss, which can be mimicked in humans and animals by inhibition of mitochondrial complex II using 3-Nitropropionic acid (3-NP). Herein, we compared histological and behavioral outcomes between mice that were wild-type or null for L-12/15 LO following systemic injection of 3NP. We found that mice deficient in L-12/15 LO had a higher incidence of striatal lesions coincident with an increase in morbidity as compared to their wild-type littermate controls. This could not be explained by differential metabolism of 3-NP as striatal succinate dehydrogenase activity was inhibited to the same extent in both genotypes. The present results show that deleting L-12/15 LO is detrimental to the striatum in the setting of chronic, systemic 3-NP exposure and are consistent with the overall conclusion that region-specific effects may determine the ultimate outcome of L-12/15 LO activation in the setting of brain injury.
Collapse
Affiliation(s)
- Yan He
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY 13210, United States
| | - Rita C Akumuo
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY 13210, United States
| | - Yuan Yang
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY 13210, United States
| | - Sandra J Hewett
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY 13210, United States.
| |
Collapse
|
22
|
Polyzos AA, McMurray CT. The chicken or the egg: mitochondrial dysfunction as a cause or consequence of toxicity in Huntington's disease. Mech Ageing Dev 2017; 161:181-197. [PMID: 27634555 PMCID: PMC5543717 DOI: 10.1016/j.mad.2016.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/07/2016] [Accepted: 09/12/2016] [Indexed: 01/30/2023]
Abstract
Mitochondrial dysfunction and ensuing oxidative damage is typically thought to be a primary cause of Huntington's disease, Alzheimer's disease, and Parkinson disease. There is little doubt that mitochondria (MT) become defective as neurons die, yet whether MT defects are the primary cause or a detrimental consequence of toxicity remains unanswered. Oxygen consumption rate (OCR) and glycolysis provide sensitive and informative measures of the functional status MT and the cells metabolic regulation, yet these measures differ depending on the sample source; species, tissue type, age at measurement, and whether MT are measured in purified form or in a cell. The effects of these various parameters are difficult to quantify and not fully understood, but clearly have an impact on interpreting the bioenergetics of MT or their failure in disease states. A major goal of the review is to discuss issues and coalesce detailed information into a reference table to help in assessing mitochondrial dysfunction as a cause or consequence of Huntington's disease.
Collapse
Affiliation(s)
- Aris A Polyzos
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd., Berkeley, CA 94720, USA.
| | - Cynthia T McMurray
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd., Berkeley, CA 94720, USA.
| |
Collapse
|
23
|
Dihydromyricetin Ameliorates 3NP-induced Behavioral Deficits and Striatal Injury in Rats. J Mol Neurosci 2016; 60:267-75. [PMID: 27501707 DOI: 10.1007/s12031-016-0801-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 07/14/2016] [Indexed: 01/29/2023]
Abstract
Oxidative stress is closely involved in neurodegenerative diseases. The present study aimed to examine the effect of anti-oxidant DHM (dihydromyricetin) on 3NP (3-nitropropionic acid) -induced behavioral deficits of experimental rats and striatal histopathological injury by using behavioral, imaging, biochemistry, histochemistry and molecular biology technologies. The experimental results showed that both motor dysfunctions and learning and memory impairments induced by 3NP were significantly reduced after DHM treatment. 3NP-induced striatal metabolic abnormality was also remarkably improved by DHM treatment, showed as the increased glucose metabolism in PET/CT scan, decreased MDA (malondialdehyde) and increased SOD (superoxide dismutase) activity in enzyme histochemical staining. In addition, the cell apoptosis was evidently detected in the striatum of the 3NP group, while in the 3NP + DHM group, the number of apoptotic cells was remarkably reduced. 3NP treatment obviously induced down-regulation of Bcl-2, and up-regulations of Bax and Cleaved Caspase-3, while these changes were significantly reversed by DHM treatment. The present results suggested that DHM showed its protective effect by anti-oxidant and anti-apoptosis mechanisms.
Collapse
|
24
|
Chan F, Lax NZ, Davies CH, Turnbull DM, Cunningham MO. Neuronal oscillations: A physiological correlate for targeting mitochondrial dysfunction in neurodegenerative diseases? Neuropharmacology 2016; 102:48-58. [DOI: 10.1016/j.neuropharm.2015.10.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/19/2015] [Accepted: 10/24/2015] [Indexed: 12/21/2022]
|
25
|
Kim HL, Lee MY, Shin YJ, Song DW, Park J, Chang BS, Lee JH. Increased Expression of Osteopontin in the Degenerating Striatum of Rats Treated with Mitochondrial Toxin 3-Nitropropionic Acid: A Light and Electron Microscopy Study. Acta Histochem Cytochem 2015; 48:135-43. [PMID: 26633905 PMCID: PMC4652028 DOI: 10.1267/ahc.15010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/04/2015] [Indexed: 12/16/2022] Open
Abstract
The mycotoxin 3-nitropropionic acid (3NP) is an irreversible inhibitor that induces neuronal damage by inhibiting mitochondrial complex II. Neurodegeneration induced by 3NP, which is preferentially induced in the striatum, is caused by an excess influx and accumulation of calcium in mitochondria. Osteopontin (OPN) is a glycosylated phosphoprotein and plays a role in the regulation of calcium precipitation in the injured brain. The present study was designed to examine whether induction of OPN protein is implicated in the pathogenesis of 3NP-induced striatal neurodegeneration. We observed overlapping regional expression of OPN, the neurodegeneration marker Fluoro-Jade B, and the microglial marker ionized calcium-binding adaptor molecule 1 (Iba1) in the 3NP-lesioned striatum. OPN expression was closely associated with the mitochondrial marker NADH dehydrogenase (ubiquinone) flavoprotein 2 in the damaged striatum. In addition, immunoelectron microscopy demonstrated that OPN protein was specifically localized to the inner membrane and matrix of the mitochondria in degenerating striatal neurons, and cell fragments containing OPN-labeled mitochondria were also present within activated brain macrophages. Thus, our study revealed that OPN expression is associated with mitochondrial dysfunction produced by 3NP-induced alteration of mitochondrial calcium homeostasis, suggesting that OPN is involved in the pathogenesis of striatal degeneration by 3NP administration.
Collapse
Affiliation(s)
- Hong-Lim Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine, Konkuk University
- Integrative Research Support Center, College of Medicine, Catholic University
| | - Mun-Yong Lee
- Department of Anatomy, College of Medicine, Catholic University
| | - Yoo-Jin Shin
- Department of Anatomy, College of Medicine, Catholic University
| | - Doo-Won Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University
| | - Jieun Park
- Integrative Research Support Center, College of Medicine, Catholic University
| | | | - Jong-Hwan Lee
- Department of Veterinary Anatomy, College of Veterinary Medicine, Konkuk University
| |
Collapse
|
26
|
Pellman JJ, Hamilton J, Brustovetsky T, Brustovetsky N. Ca(2+) handling in isolated brain mitochondria and cultured neurons derived from the YAC128 mouse model of Huntington's disease. J Neurochem 2015; 134:652-67. [PMID: 25963273 PMCID: PMC4516671 DOI: 10.1111/jnc.13165] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 01/10/2023]
Abstract
We investigated Ca(2+) handling in isolated brain synaptic and non-synaptic mitochondria and in cultured striatal neurons from the YAC128 mouse model of Huntington's disease. Both synaptic and non-synaptic mitochondria from 2- and 12-month-old YAC128 mice had larger Ca(2+) uptake capacity than mitochondria from YAC18 and wild-type FVB/NJ mice. Synaptic mitochondria from 12-month-old YAC128 mice had further augmented Ca(2+) capacity compared with mitochondria from 2-month-old YAC128 mice and age-matched YAC18 and FVB/NJ mice. This increase in Ca(2+) uptake capacity correlated with an increase in the amount of mutant huntingtin protein (mHtt) associated with mitochondria from 12-month-old YAC128 mice. We speculate that this may happen because of mHtt-mediated sequestration of free fatty acids thereby increasing resistance of mitochondria to Ca(2+)-induced damage. In experiments with striatal neurons from YAC128 and FVB/NJ mice, brief exposure to 25 or 100 μM glutamate produced transient elevations in cytosolic Ca(2+) followed by recovery to near resting levels. Following recovery of cytosolic Ca(2+), mitochondrial depolarization with FCCP produced comparable elevations in cytosolic Ca(2+), suggesting similar Ca(2+) release and, consequently, Ca(2+) loads in neuronal mitochondria from YAC128 and FVB/NJ mice. Together, our data argue against a detrimental effect of mHtt on Ca(2+) handling in brain mitochondria of YAC128 mice. We demonstrate that mutant huntingtin (mHtt) binds to brain synaptic and nonsynaptic mitochondria and the amount of mitochondria-bound mHtt correlates with increased mitochondrial Ca(2+) uptake capacity. We propose that this may happen due to mHtt-mediated sequestration of free fatty acids thereby increasing resistance of mitochondria to Ca(2+)-induced damage.
Collapse
Affiliation(s)
- Jessica J. Pellman
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN 46202, USA
| | - James Hamilton
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN 46202, USA
| | - Tatiana Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN 46202, USA
| | - Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN 46202, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis IN 46202, USA
| |
Collapse
|
27
|
Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WKD, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson's disease. Prog Neurobiol 2015. [PMID: 26209473 DOI: 10.1016/j.pneurobio.2015.07.002] [Citation(s) in RCA: 347] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.
Collapse
Affiliation(s)
- Matthieu F Bastide
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wassilios G Meissner
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | - Barbara Picconi
- Laboratory of Neurophysiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Stefania Fasano
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Pierre-Olivier Fernagut
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michael Feyder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cristina Alcacer
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yunmin Ding
- Department of Neurology, Columbia University, New York, USA
| | - Riccardo Brambilla
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and National Parkinson Foundation Centre of Excellence, University of British Columbia, Vancouver, Canada
| | - Mathieu Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michel Engeln
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Sylvia Navailles
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Philippe De Deurwaerdère
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wai Kin D Ko
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Laurent Groc
- Univ. de Bordeaux, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France; CNRS, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France
| | - Maria-Cruz Rodriguez
- Department of Neurology, Hospital Universitario Donostia and Neuroscience Unit, Bio Donostia Research Institute, San Sebastian, Spain
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, CA 94025, USA
| | - Michele Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - Dominique Guehl
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - François Tison
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | | | - Un Jung Kang
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kathy Steece-Collier
- Michigan State University, College of Human Medicine, Department of Translational Science and Molecular Medicine & The Udall Center of Excellence in Parkinson's Disease Research, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Susan Fox
- Morton & Gloria Shulman Movement Disorders Center, Toronto Western Hospital, Toronto, Ontario M4T 2S8, Canada
| | - Manolo Carta
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Erwan Bézard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Motac Neuroscience Ltd, Manchester, UK.
| |
Collapse
|
28
|
Chan AWS, Jiang J, Chen Y, Li C, Prucha MS, Hu Y, Chi T, Moran S, Rahim T, Li S, Li X, Zola SM, Testa CM, Mao H, Villalba R, Smith Y, Zhang X, Bachevalier J. Progressive cognitive deficit, motor impairment and striatal pathology in a transgenic Huntington disease monkey model from infancy to adulthood. PLoS One 2015; 10:e0122335. [PMID: 25966278 PMCID: PMC4428630 DOI: 10.1371/journal.pone.0122335] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/10/2015] [Indexed: 02/02/2023] Open
Abstract
One of the roadblocks to developing effective therapeutics for Huntington disease (HD) is the lack of animal models that develop progressive clinical traits comparable to those seen in patients. Here we report a longitudinal study that encompasses cognitive and motor assessment, and neuroimaging of a group of transgenic HD and control monkeys from infancy to adulthood. Along with progressive cognitive and motor impairment, neuroimaging revealed a progressive reduction in striatal volume. Magnetic resonance spectroscopy at 48 months of age revealed a decrease of N-acetylaspartate (NAA), further suggesting neuronal damage/loss in the striatum. Postmortem neuropathological analyses revealed significant neuronal loss in the striatum. Our results indicate that HD monkeys share similar disease patterns with HD patients, making them potentially suitable as a preclinical HD animal model.
Collapse
Affiliation(s)
- Anthony W. S. Chan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| | - Jie Jiang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Yiju Chen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Chunxia Li
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Melinda S. Prucha
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Yijuan Hu
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, Georgia, United States of America
| | - Tim Chi
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Sean Moran
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Tayeb Rahim
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Shihua Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Xiaojiang Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Stuart M. Zola
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Claudia M. Testa
- Department of Neurology and Parkinson’s and Movement Disorders Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Rosa Villalba
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Yoland Smith
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Xiaodong Zhang
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Jocelyne Bachevalier
- Department of Psychology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Center, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
29
|
Malik J, Choudhary S, Kumar P. Protective effect of Convolvulus pluricaulis standardized extract and its fractions against 3-nitropropionic acid-induced neurotoxicity in rats. PHARMACEUTICAL BIOLOGY 2015; 53:1448-1457. [PMID: 25853968 DOI: 10.3109/13880209.2014.984856] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Convolvulus pluricaulis Chois. (Convolvulaceae), a well-known Ayurvedic "Medhya Rasayana" (nervine tonic), is extensively used for different central nervous system (CNS) disorders. OBJECTIVE The objective of this study was to evaluate the protective effect of standardized hydro-methanol extract of C. pluricaulis (CPE) and its fractions, namely ethyl acetate (EAE), butanol (BE), and aqueous (AE), against 3-nitropropionic acid (3-NP) induced neurotoxicity in rats. MATERIALS AND METHODS The extract of the whole plant was standardized on the basis of scopoletin content (0.014%) using thin layer chromatography densitometric analysis. CPE (100 and 200 mg/kg) and its fractions, namely EAE (15 and 30 mg/kg), BE (25 and 50 mg/kg), and AE (50 and 100 mg/kg) were administered orally for 20 d. Their protective effect against 3-NP (10 mg/kg, i.p. for 14 d) was assessed by the effect on various behavioral parameters, namely body weight, locomotor activity, grip strength, gait pattern, and the effect on cognitive dysfunction. Biochemical parameters for oxidative damage were also assessed in the striatum and cortex regions of the brain. RESULTS Administration of 3-NP induced HD-like symptoms that were indicated by reduced body weight, locomotor activity, memory, grip strength, and oxidative defense. CPE (200 mg/kg), EAE (30 mg/kg), and BE (50 mg/kg) significantly (p < 0.001) attenuated 3-NP induced reduction in locomotor activity, grip strength, memory, body weight, and oxidative defense in comparison with 3-NP-treated animals on 10 and 15 d. CONCLUSION The present study suggested that CPE has a protective action against 3-NP-induced neurotoxicity and can be further explored for its efficacy against Huntington's disease.
Collapse
Affiliation(s)
- Jai Malik
- University Institute of Pharmaceutical Sciences, Panjab University , Chandigarh , India
| | | | | |
Collapse
|
30
|
Durand E, Petit O, Tremblay L, Zimmer C, Sgambato-Faure V, Chassain C, Laurent M, Pereira B, Silberberg C, Durif F. Social behavioral changes in MPTP-treated monkey model of Parkinson's disease. Front Behav Neurosci 2015; 9:42. [PMID: 25767440 PMCID: PMC4341564 DOI: 10.3389/fnbeh.2015.00042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/05/2015] [Indexed: 11/26/2022] Open
Abstract
Parkinsonian patients experience not only the physical discomfort of motor disorders but also the considerable psychological distress caused by cognitive deficits and behavioral disorders. These two factors can result in a disruption of social relationships during the symptomatic and even the presymptomatic motor states of the disease. However, it remains difficult, if not impossible, to evaluate social relationships in presymptomatic patients. The present study focused on the evaluation of social relationships within a group of female long-tailed macaques during presymptomatic and symptomatic motor states induced by Chronic Low-Dose (CLD) and then Chronic High-Dose (CHD) systemic administration of 1-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP). Dopaminergic denervation within basal ganglia and cortical areas was evaluated using Positron Emission Tomography (PET) scans with (18)F-DOPA (6-[18F]-fluoro-L-3,4-dihydroxyphenylalanine) radiotracer. Interestingly, social behavioral changes could be identified in the presymptomatic motor state before any motor and/or cognitive impairment occurred. Stronger effects were observed in subordinate animals compared to dominant animals. From baseline state to CLD-presymptomatic motor state, the frequency of emitted affiliative and aggressive behaviors increased. From CLD-presymptomatic to CHD-presymptomatic motor states, the frequency of the three categories of social behaviors (aggressive, submissive and affiliative) decreased. At this time, quantitative data analysis in PET scans highlighted a dopaminergic denervation in the insula and the posterior caudate nucleus. Finally, the frequency of the three categories of social behaviors decreased during the stable-symptomatic motor state compared to baseline and presymptomatic motor states; this was also associated with motor and cognitive disorders and a dopaminergic denervation in all the evaluated cortical and subcortical structures.
Collapse
Affiliation(s)
- Elodie Durand
- Université d'Auvergne Clermont 1, UFR Médecine, EA 7280 (NPsy-Sydo)Clermont-Ferrand, France
| | - Odile Petit
- Département Ecologie, Physiologie et Ethologie, Institut Pluridisciplinaire Hubert Curien, UMR 7178, CNRS-UDSStrasbourg, France
| | - Léon Tremblay
- Centre de Neurosciences Cognitives, UMR 5229 CNRS-Université Lyon 1Bron, France
| | - Cédric Zimmer
- Département Ecologie, Physiologie et Ethologie, Institut Pluridisciplinaire Hubert Curien, UMR 7178, CNRS-UDSStrasbourg, France
| | | | - Carine Chassain
- CHU Clermont Ferrand, Gabriel MontpiedClermont-Ferrand, France
| | - Marlène Laurent
- Université d'Auvergne Clermont 1, UFR Médecine, EA 7280 (NPsy-Sydo)Clermont-Ferrand, France
| | - Bruno Pereira
- CHU Clermont Ferrand, Gabriel MontpiedClermont-Ferrand, France
| | - Céline Silberberg
- Université d'Auvergne Clermont 1, UFR Médecine, EA 7280 (NPsy-Sydo)Clermont-Ferrand, France
| | - Franck Durif
- Université d'Auvergne Clermont 1, UFR Médecine, EA 7280 (NPsy-Sydo)Clermont-Ferrand, France
| |
Collapse
|
31
|
Hering T, Birth N, Taanman JW, Orth M. Selective striatal mtDNA depletion in end-stage Huntington's disease R6/2 mice. Exp Neurol 2015; 266:22-9. [PMID: 25682918 DOI: 10.1016/j.expneurol.2015.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/20/2015] [Accepted: 02/05/2015] [Indexed: 12/21/2022]
Abstract
In Huntington's disease (HD) the striatum and cortex seem particularly vulnerable. Mitochondrial dysfunction can also cause neurodegeneration with prominent striatal involvement very similar to HD. We first examined if mitochondrial biogenesis, mitochondrial DNA (mtDNA) transcription, and the implications for mitochondrial respiratory chain (MRC) assembly and function differ between the striatum and cortex compared with the whole brain average in the healthy mouse brain. We then examined the effects of the mutant huntingtin transgene in end-stage R6/2 mice. In wild-type mice, mitochondrial mass (citrate synthase levels, mtDNA copy number) was higher in the striatum than in the cortex or whole brain average. PGC-1α and TFAM mRNA levels were also higher in the striatum than the whole brain average and cortex. mRNA reserve for MRC Complex proteins was higher in the striatum and cortex. In addition, in the cortex a greater part of mitochondrial mass was dedicated to the generation of ATP by oxidative phosphorylation than in the striatum or on average in the brain. In the HD transgenic striatum there was selective mtDNA depletion without evidence that this translated to abnormalities of steady-state MRC function. Our data indicate that in mice the striatum differs from the cortex, or whole brain average, in potentially important aspects of mitochondrial biology. This may contribute to the increased vulnerability of the striatum to insults such as the HD mutation, causing selective striatal mtDNA depletion in end-stage R6/2 mice.
Collapse
Affiliation(s)
- Tanja Hering
- Department of Neurology, Ulm University, Germany
| | | | - Jan-Willem Taanman
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, UK
| | - Michael Orth
- Department of Neurology, Ulm University, Germany
| |
Collapse
|
32
|
Mendoza E, Miranda-Barrientos J, Vázquez-Roque R, Morales-Herrera E, Ruelas A, De la Rosa G, Flores G, Hernández-Echeagaray E. In vivo mitochondrial inhibition alters corticostriatal synaptic function and the modulatory effects of neurotrophins. Neuroscience 2014; 280:156-70. [DOI: 10.1016/j.neuroscience.2014.09.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 09/09/2014] [Indexed: 01/09/2023]
|
33
|
Kannike K, Sepp M, Zuccato C, Cattaneo E, Timmusk T. Forkhead transcription factor FOXO3a levels are increased in Huntington disease because of overactivated positive autofeedback loop. J Biol Chem 2014; 289:32845-57. [PMID: 25271153 PMCID: PMC4239633 DOI: 10.1074/jbc.m114.612424] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Huntington disease (HD) is a fatal autosomal dominant neurodegenerative disorder caused by an increased number of CAG repeats in the HTT gene coding for huntingtin. Decreased neurotrophic support and increased mitochondrial and excitotoxic stress have been reported in HD striatal and cortical neurons. The members of the class O forkhead (FOXO) transcription factor family, including FOXO3a, act as sensor proteins that are activated upon decreased survival signals and/or increased cellular stress. Using immunocytochemical screening in mouse striatal Hdh7/7 (wild type), Hdh7/109 (heterozygous for HD mutation), and Hdh109/109 (homozygous for HD mutation) cells, we identified FOXO3a as a differentially regulated transcription factor in HD. We report increased nuclear FOXO3a levels in mutant Hdh cells. Additionally, we show that treatment with mitochondrial toxin 3-nitropropionic acid results in enhanced nuclear localization of FOXO3a in wild type Hdh7/7 cells and in rat primary cortical neurons. Furthermore, mRNA levels of Foxo3a are increased in mutant Hdh cells compared with wild type cells and in 3-nitropropionic acid-treated primary neurons compared with untreated neurons. A similar increase was observed in the cortex of R6/2 mice and HD patient post-mortem caudate tissue compared with controls. Using chromatin immunoprecipitation and reporter assays, we demonstrate that FOXO3a regulates its own transcription by binding to the conserved response element in Foxo3a promoter. Altogether, the findings of this study suggest that FOXO3a levels are increased in HD cells as a result of overactive positive feedback loop.
Collapse
Affiliation(s)
- Kaja Kannike
- From the Department of Gene Technology, Tallinn University of Technology, Tallinn 12618, Estonia and
| | - Mari Sepp
- From the Department of Gene Technology, Tallinn University of Technology, Tallinn 12618, Estonia and
| | - Chiara Zuccato
- the Department of Pharmacological Sciences and Center for Stem Cell Research, University of Milan, Milano 20133, Italy
| | - Elena Cattaneo
- the Department of Pharmacological Sciences and Center for Stem Cell Research, University of Milan, Milano 20133, Italy
| | - Tõnis Timmusk
- From the Department of Gene Technology, Tallinn University of Technology, Tallinn 12618, Estonia and
| |
Collapse
|
34
|
A role for human mitochondrial complex II in the production of reactive oxygen species in human skin. Redox Biol 2014; 2:1016-22. [PMID: 25460738 PMCID: PMC4215388 DOI: 10.1016/j.redox.2014.08.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 08/16/2014] [Accepted: 08/25/2014] [Indexed: 11/20/2022] Open
Abstract
The mitochondrial respiratory chain is a major generator of cellular oxidative stress, thought to be an underlying cause of the carcinogenic and ageing process in many tissues including skin. Previous studies of the relative contributions of the respiratory chain (RC) complexes I, II and III towards production of reactive oxygen species (ROS) have focussed on rat tissues and certainly not on human skin which is surprising as this tissue is regularly exposed to UVA in sunlight, a potent generator of cellular oxidative stress. In a novel approach we have used an array of established specific metabolic inhibitors and DHR123 fluorescence to study the relative roles of the mitochondrial RC complexes in cellular ROS production in 2 types of human skin cells. These include additional enhancement of ROS production by exposure to physiological levels of UVA. The effects within epidermal and dermal derived skin cells are compared to other tissue cell types as well as those harbouring a compromised mitochondrial status (Rho-zero A549). The results show that the complex II inhibitor, TTFA, was the only RC inhibitor to significantly increase UVA-induced ROS production in both skin cell types (P<0.05) suggesting that the role of human skin complex II in terms of influencing ROS production is more important than previously thought particularly in comparison to liver cells. Interestingly, two-fold greater maximal activity of complex II enzyme was observed in both skin cell types compared to liver (P<0.001). The activities of RC enzymes appear to decrease with increasing age and telomere length is correlated with ageing. Our study showed that the level of maximal complex II activity was higher in the MRC5/hTERT (human lung fibroblasts transfected with telomerase) cells than the corresponding wild type cells (P=0.0012) which can be considered (in terms of telomerase activity) as models of younger and older cells respectively. We examined the influence of mitochondrial complex II on ROS production in human skin. Past studies have focussed on ROS production from mitochondrial complexes I and III. DHR123 fluorescence was used following individual complex inhibition and UVA exposure. Only complex II inhibition significantly increased ROS levels in both skin cell types. Complex II had a two-fold greater activity in skin cells compared to liver cells.
Collapse
|
35
|
Efficacy of selective PDE4D negative allosteric modulators in the object retrieval task in female cynomolgus monkeys (Macaca fascicularis). PLoS One 2014; 9:e102449. [PMID: 25050979 PMCID: PMC4106781 DOI: 10.1371/journal.pone.0102449] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 05/19/2014] [Indexed: 11/19/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) signalling plays an important role in synaptic plasticity and information processing in the hippocampal and basal ganglia systems. The augmentation of cAMP signalling through the selective inhibition of phosphodiesterases represents a viable strategy to treat disorders associated with dysfunction of these circuits. The phosphodiesterase (PDE) type 4 inhibitor rolipram has shown significant pro-cognitive effects in neurological disease models, both in rodents and primates. However, competitive non-isoform selective PDE4 inhibitors have a low therapeutic index which has stalled their clinical development. Here, we demonstrate the pro-cognitive effects of selective negative allosteric modulators (NAMs) of PDE4D, D159687 and D159797 in female Cynomolgous macaques, in the object retrieval detour task. The efficacy displayed by these NAMs in a primate cognitive task which engages the corticostriatal circuitry, together with their suitable pharmacokinetic properties and safety profiles, suggests that clinical development of these allosteric modulators should be considered for the treatment of a variety of brain disorders associated with cognitive decline.
Collapse
|
36
|
Choi YJ, Kim NH, Lim MS, Lee HJ, Kim SS, Chun W. Geldanamycin attenuates 3‑nitropropionic acid‑induced apoptosis and JNK activation through the expression of HSP 70 in striatal cells. Int J Mol Med 2014; 34:24-34. [PMID: 24756698 PMCID: PMC4072345 DOI: 10.3892/ijmm.2014.1747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/14/2014] [Indexed: 11/05/2022] Open
Abstract
Although selective striatal cell death is a characteristic hallmark in the pathogenesis of Huntington's disease (HD), the underlying mechanism of striatal susceptibility remains to be clarified. Heat shock proteins (HSPs) have been reported to suppress the aggregate formation of mutant huntingtin and concurrent striatal cell death. In a previous study, we observed that heat shock transcription factor 1 (HSF1), a major transcription factor of HSPs, significantly attenuated 3‑nitropropionic acid (3NP)‑induced reactive oxygen species (ROS) production and apoptosis through the expression of HSP 70 in striatal cells. To investigate the differential roles of HSPs in 3NP‑induced striatal cell death, the effect of geldanamycin (GA), an HSP 90 inhibitor, was examined in 3NP‑stimulated striatal cells. GA significantly attenuated 3NP‑induced striatal apoptosis and ROS production with an increased expression of HSP 70. Triptolide (TL), an HSP 70 inhibitor, abolished GA‑mediated protective effects in 3NP‑stimulated striatal cells. To understand the underlying mechanism by which GA‑mediated HSP 70 protects striatal cells against 3NP stimulation, the involvement of various signaling pathways was examined. GA significantly attenuated 3NP‑induced c‑Jun N‑terminal kinase (JNK) phosphorylation and subsequent c‑Jun phosphorylation in striatal cells. Taken together, the present study demonstrated that GA exhibits protective properties against 3NP‑induced apoptosis and JNK activation via the induction of HSP 70 in striatal cells, suggesting that expression of HSP 70 may be a valuable therapeutic target in the treatment of HD.
Collapse
Affiliation(s)
- Yong-Joon Choi
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Nam Ho Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Man Sup Lim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Sung Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Gangwon 200‑701, Republic of Korea
| |
Collapse
|
37
|
Wilson BK, Hess EJ. Animal models for dystonia. Mov Disord 2014; 28:982-9. [PMID: 23893454 DOI: 10.1002/mds.25526] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 03/19/2013] [Accepted: 04/29/2013] [Indexed: 01/28/2023] Open
Abstract
Symptomatic animal models have clinical features consistent with human disorders and are often used to identify the anatomical and physiological processes involved in the expression of symptoms and to experimentally demonstrate causality where it would be infeasible in the patient population. Rodent and primate models of dystonia have identified basal ganglia abnormalities, including alterations in striatal GABAergic (ie, transmitting or secreting γ-aminobutyric acid) and dopaminergic transmission. Symptomatic animal models have also established the critical role of the cerebellum in dystonia, particularly abnormal glutamate signaling and aberrant Purkinje cell activity. Further, experiments suggest that the basal ganglia and cerebellum are nodes in an integrated network that is dysfunctional in dystonia. The knowledge gained from experiments in symptomatic animal models may serve as the foundation for the development of novel therapeutic interventions to treat dystonia. © 2013 Movement Disorder Society.
Collapse
Affiliation(s)
- Bethany K Wilson
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | |
Collapse
|
38
|
Lehéricy S, Tijssen MAJ, Vidailhet M, Kaji R, Meunier S. The anatomical basis of dystonia: current view using neuroimaging. Mov Disord 2014; 28:944-57. [PMID: 23893451 DOI: 10.1002/mds.25527] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 04/06/2013] [Accepted: 05/02/2013] [Indexed: 12/15/2022] Open
Abstract
This review will consider the knowledge that neuroimaging studies have provided to the understanding of the anatomy of dystonia. Major advances have occurred in the use of neuroimaging for dystonia in the past 2 decades. At present, the most developed imaging approaches include whole-brain or region-specific studies of structural or diffusion changes, functional imaging using fMRI or positron emission tomography (PET), and metabolic imaging using fluorodeoxyglucose PET. These techniques have provided evidence that regions other than the basal ganglia are involved in dystonia. In particular, there is increasing evidence that primary dystonia can be viewed as a circuit disorder, involving the basal ganglia-thalamo-cortical and cerebello-thalamo-cortical pathways. This suggests that a better understanding of the dysfunction in each region in the network and their interactions are important topics to address. Current views of interpretation of imaging data as cause or consequence of dystonia, and the postmortem correlates of imaging data are presented. The application of imaging as a tool to monitor therapy and its use as an outcome measure will be discussed. © 2013 Movement Disorder Society.
Collapse
Affiliation(s)
- Stéphane Lehéricy
- Institut du Cerveau et de la Moelle (ICM) epiniere, Centre de NeuroImagerie de Recherche (CENIR), Paris, France.
| | | | | | | | | |
Collapse
|
39
|
Thangarajan S, Deivasigamani A, Natarajan SS, Krishnan P, Mohanan SK. Neuroprotective activity ofL-theanine on 3-nitropropionic acid-induced neurotoxicity in rat striatum. Int J Neurosci 2014; 124:673-84. [DOI: 10.3109/00207454.2013.872642] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Chakraborty J, Rajamma U, Mohanakumar KP. A mitochondrial basis for Huntington's disease: therapeutic prospects. Mol Cell Biochem 2013; 389:277-91. [PMID: 24374792 DOI: 10.1007/s11010-013-1951-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 12/19/2013] [Indexed: 01/12/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant disease, with overt movement dysfunctions. Despite focused research on the basis of neurodegeneration in HD for last few decades, the mechanism for the site-specific lesion of neurons in the brain is not clear. All the explanations that partially clarify the phenomenon of neurodegeneration leads to one organelle, mitochondrion, which is severely affected in HD at the level of electron transport chain, Ca(2+) buffering efficiency and morphology. But, with the existing knowledge, it is not clear whether the cell death processes in HD initiate from mitochondria, though the Huntingtin (Htt) aggregates show close proximity to this organelle, or do some extracellular stimuli like TNFα or FasL trigger the process. Mainly because of the disparity in the different available experimental models, the results are quite confusing or at least inconsistent to a great extent. The fact remains that the mutant Htt protein was seen to be associated with mitochondria directly, and as the striatum is highly enriched with dopamine and glutamate, it may make the striatal mitochondria more vulnerable because of the presence of dopa-quinones, and due to an imbalance in Ca(2+). The current therapeutic strategies are based on symptomatic relief, and, therefore, mainly target neurotransmitter(s) and their receptors to modulate behavioral outputs, but none of them targets mitochondria or try to address the basic molecular events that cause neurons to die in discrete regions of the brain, which could probably be resulting from grave mitochondrial dysfunctions. Therefore, targeting mitochondria for their protection, while addressing symptomatic recovery, holds a great potential to tone down the progression of the disease, and to provide better relief to the patients and caretakers.
Collapse
Affiliation(s)
- J Chakraborty
- Laboratory of Clinical and Experimental Neuroscience, Division of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Rooms 117&119, 4, Raja S. C. Mullick Road, Kolkata, 700 032, India
| | | | | |
Collapse
|
41
|
Pereira GJS, Tressoldi N, Hirata H, Bincoletto C, Smaili SS. Autophagy as a Neuroprotective Mechanism Against 3-Nitropropionic Acid-Induced Murine Astrocyte Cell Death. Neurochem Res 2013; 38:2418-26. [DOI: 10.1007/s11064-013-1154-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 09/16/2013] [Indexed: 11/29/2022]
|
42
|
Clabough EBD. Huntington's disease: the past, present, and future search for disease modifiers. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2013; 86:217-33. [PMID: 23766742 PMCID: PMC3670441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant genetic disorder that specifically causes neurodegeneration of striatal neurons, resulting in a triad of symptoms that includes emotional, cognitive, and motor disturbances. The HD mutation causes a polyglutamine repeat expansion within the N-terminal of the huntingtin (Htt) protein. This expansion causes aggregate formation within the cytosol and nucleus due to the presence of misfolded mutant Htt, as well as altered interactions with Htt's multiple binding partners, and changes in post-translational Htt modifications. The present review charts efforts toward a therapy that delays age of onset or slows symptom progression in patients affected by HD, as there is currently no effective treatment. Although silencing Htt expression appears promising as a disease modifying treatment, it should be attempted with caution in light of Htt's essential roles in neural maintenance and development. Other therapeutic targets include those that boost aggregate dissolution, target excitotoxicity and metabolic issues, and supplement growth factors.
Collapse
Affiliation(s)
- Erin B D Clabough
- Randolph-Macon College, Department of Biology, Ashland, Virginia 23005, USA.
| |
Collapse
|
43
|
Aron Badin R, Hantraye P. Designing primate models to assess the prodromal phase of Huntington's disease. Rev Neurol (Paris) 2012; 168:802-5. [DOI: 10.1016/j.neurol.2012.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
44
|
Ruiz M, Déglon N. Viral-mediated overexpression of mutant huntingtin to model HD in various species. Neurobiol Dis 2011; 48:202-11. [PMID: 21889981 DOI: 10.1016/j.nbd.2011.08.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 08/11/2011] [Accepted: 08/18/2011] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an expansion of CAG repeats in the huntingtin (Htt) gene. Despite intensive efforts devoted to investigating the mechanisms of its pathogenesis, effective treatments for this devastating disease remain unavailable. The lack of suitable models recapitulating the entire spectrum of the degenerative process has severely hindered the identification and validation of therapeutic strategies. The discovery that the degeneration in HD is caused by a mutation in a single gene has offered new opportunities to develop experimental models of HD, ranging from in vitro models to transgenic primates. However, recent advances in viral-vector technology provide promising alternatives based on the direct transfer of genes to selected sub-regions of the brain. Rodent studies have shown that overexpression of mutant human Htt in the striatum using adeno-associated virus or lentivirus vectors induces progressive neurodegeneration, which resembles that seen in HD. This article highlights progress made in modeling HD using viral vector gene transfer. We describe data obtained with of this highly flexible approach for the targeted overexpression of a disease-causing gene. The ability to deliver mutant Htt to specific tissues has opened pathological processes to experimental analysis and allowed targeted therapeutic development in rodent and primate pre-clinical models.
Collapse
Affiliation(s)
- Marta Ruiz
- Atomic Energy Commission (CEA), Institute of Biomedical Imaging (I2BM), Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses, France
| | | |
Collapse
|
45
|
Kelps KA, Turchan-Cholewo J, Hascup ER, Taylor TL, Gash DM, Gerhardt GA, Bradley LH. Evaluation of the physical and in vitro protective activity of three synthetic peptides derived from the pro- and mature GDNF sequence. Neuropeptides 2011; 45:213-8. [PMID: 21507484 PMCID: PMC3091812 DOI: 10.1016/j.npep.2011.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 03/21/2011] [Accepted: 03/22/2011] [Indexed: 12/12/2022]
Abstract
Recently, a small 11-amino acid amidated peptide, dopamine neuron stimulating peptide-11 (DNSP-11), was shown to exert neurotrophic-like actions on primary dopaminergic neurons and in parkinsonian rat models. This suggests smaller neurotrophic-like molecules may be deliverable and modifiable for therapeutic use. Here we evaluate the molecular and cellular protection properties of DNSP-11 and two other amidated-peptides, a 5-mer (DNSP-5) and a 17-mer (DNSP-17), hypothesized to be endoproteolytically processed from the pro- and mature glial cell line-derived neurotrophic factor (GDNF) protein sequence, respectively. Far-UV circular dichroism spectra show that the three DNSPs are soluble and act independently in vitro. Reverse phase HPLC and mass spectrometry analysis show that the three peptides are stable for one month at a variety of storage and experimental conditions. To gain insight into their biodistribution properties in the brain, we used affinity chromatography to show that DNSP-17 binds heparin equally as tight as GDNF, whereas DNSP-5 and DNSP-11 do not bind heparin, which should facilitate their delivery in vivo. Finally, we present data showing that DNSP-11 provides dose-dependent protection of HEK-293 cells from staurosporine and 3-nitropropionate (3-NP) cytotoxicity, thereby supporting its broad mitochondrial-protective properties.
Collapse
Affiliation(s)
- Kristen A. Kelps
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Jadwiga Turchan-Cholewo
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Erin R. Hascup
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Tiffany L. Taylor
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Don M. Gash
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Greg A. Gerhardt
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| | - Luke H. Bradley
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, U.S.A
- Department of Molecular & Cellular Biochemistry and the Center of Structural Biology, University of Kentucky College of Medicine, Lexington, KY, U.S.A
| |
Collapse
|
46
|
Neychev VK, Gross RE, Lehéricy S, Hess EJ, Jinnah HA. The functional neuroanatomy of dystonia. Neurobiol Dis 2011; 42:185-201. [PMID: 21303695 DOI: 10.1016/j.nbd.2011.01.026] [Citation(s) in RCA: 320] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 01/08/2011] [Accepted: 01/28/2011] [Indexed: 10/18/2022] Open
Abstract
Dystonia is a neurological disorder characterized by involuntary twisting movements and postures. There are many different clinical manifestations, and many different causes. The neuroanatomical substrates for dystonia are only partly understood. Although the traditional view localizes dystonia to basal ganglia circuits, there is increasing recognition that this view is inadequate for accommodating a substantial portion of available clinical and experimental evidence. A model in which several brain regions play a role in a network better accommodates the evidence. This network model accommodates neuropathological and neuroimaging evidence that dystonia may be associated with abnormalities in multiple different brain regions. It also accommodates animal studies showing that dystonic movements arise with manipulations of different brain regions. It is consistent with neurophysiological evidence suggesting defects in neural inhibitory processes, sensorimotor integration, and maladaptive plasticity. Finally, it may explain neurosurgical experience showing that targeting the basal ganglia is effective only for certain subpopulations of dystonia. Most importantly, the network model provides many new and testable hypotheses with direct relevance for new treatment strategies that go beyond the basal ganglia. This article is part of a Special Issue entitled "Advances in dystonia".
Collapse
|
47
|
Abstract
Huntington disease (HD) is an autosomal dominant neurodegenerative disease with complete penetrance. Although the understanding of the cellular mechanisms that drive neurodegeneration in HD and account for the characteristic pattern of neuronal vulnerability is incomplete, defects in energy metabolism, particularly mitochondrial function, represent a common thread in studies of HD pathogenesis in humans and animal models. Here we review the clinical, biochemical, and molecular evidence of an energy deficit in HD and discuss the mechanisms underlying mitochondrial and related alterations.
Collapse
Affiliation(s)
- Fanny Mochel
- INSERM UMR S975, Institut du Cerveau et de la Moelle,
AP-HP, Département de Génétique, and
Unité Fonctionnelle Neurométabolique, Hôpital La Salpêtrière, Paris, France.
Université Pierre et Marie Curie, Paris, France.
Department of Neurology, University of Texas Southwestern Medical Center and VA North Texas Medical Center, Dallas, Texas, USA.
Neuromuscular Center, Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, USA
| | - Ronald G. Haller
- INSERM UMR S975, Institut du Cerveau et de la Moelle,
AP-HP, Département de Génétique, and
Unité Fonctionnelle Neurométabolique, Hôpital La Salpêtrière, Paris, France.
Université Pierre et Marie Curie, Paris, France.
Department of Neurology, University of Texas Southwestern Medical Center and VA North Texas Medical Center, Dallas, Texas, USA.
Neuromuscular Center, Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, USA
| |
Collapse
|
48
|
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder that currently has no cure. In order to develop effective treatment, an understanding of HD pathogenesis and the evaluation of therapeutic efficacy of novel medications with the aid of animal models are critical steps. Transgenic animals sharing similar genetic defects that lead to HD have provided important discoveries in HD mechanisms that cell models are not able to replicate, which include psychiatric impairment, cognitive behavioral impact, and motor functions. Although transgenic HD rodent models have been widely used in HD research, it is clear that an animal model with comparable physiology to man, similar genetic defects that lead to HD, and the ability to develop similar cognitive and behavioral impairments is critical for explaining HD pathogenesis and the development of cures. Compared to HD rodents, HD transgenic nonhuman primates have not only developed comparable neuropathology but also present HD clinical features such as rigidity, seizure, dystonia, bradykinesia, and chorea that no other animal model has been able to replicate. Distinctive degenerating neurons and the accumulation of neuropil aggregates observed in HD monkey brain strongly support the hypothesis that the unique neuropathogenic events seen in HD monkey brain recapitulate HD in man. The latest development of transgenic HD primates has opened a new era of animal modeling that better represents human genetic disorders such as HD, which will accelerate the development of diagnostic tools and identifying novel biomarkers through longitudinal studies including gene expression and metabolite profiling, and noninvasive imaging. Furthermore, novel treatments with predictable efficacy in human patients can be developed using HD monkeys because of comparable neuropathology and clinical features.
Collapse
Affiliation(s)
- Shang-Hsun Yang
- Department of Physiology, National Cheng Kung University Medical College, 1, University Road, Tainan, 70101, Taiwan,
| | | |
Collapse
|
49
|
Nitric oxide mechanism in the protective effect of antidepressants against 3-nitropropionic acid-induced cognitive deficit, glutathione and mitochondrial alterations in animal model of Huntington's disease. Behav Pharmacol 2010; 21:217-30. [PMID: 20480544 DOI: 10.1097/fbp.0b013e32833a5bf4] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Huntington's disease (HD), a basal ganglia disorder, is characterized not only by a spectrum of motor deficits, but also by emotional, cognitive and psychiatric manifestations. Cognitive impairment is one of the serious manifestations of this disease in the later stage of life. Although there is currently no cure for HD, there has been a surge of clinical trials involving patients with HD over the past 5 years. However, cognitive measures have generally been lacking from these trials. The beneficial effect of antidepressants in HD has been suggested in recent clinical trials. However, their mechanism of action is still not clear. Therefore, this study was designed to elucidate and compare the mechanistic role of different classes of antidepressants (sertraline, venlafaxine, imipramine and trazodone) against 3-nitropropionic acid (3-NP)-induced cognitive impairment, oxidative stress (glutathione) and mitochondrial dysfunction in rat hippocampus. Systemic treatment with 3-NP (10 mg/kg for 14 days) significantly impaired memory performance (both in the Morris water maze and elevated plus maze escape retention test), oxidative defence (glutathione redox status) and mitochondrial enzyme complex activities in rat hippocampus. Sertraline, venlafaxine, imipramine and trazodone treatments significantly improved performance in both cognitive tasks and glutathione redox status, and restored mitochondrial enzyme complex activities, as compared with the 3-NP treated group. L-arginine (50 mg/kg) pretreatment for 14 days together with a subeffective dose of sertraline (10 mg/kg), venlafaxine (10 mg/kg), imipramine (10 mg/kg) or trazodone (10 mg/kg) partially attenuated their protective effects. Further, G-nitro-L-Arginine-Methyl Ester (10 mg/kg) pretreatment together with subeffective dose of sertraline (10 mg/kg), venlafaxine (10 mg/kg), imipramine (10 mg/kg) and trazodone (10 mg/kg) significantly enhanced their efficacy. The results of this study suggest that nitric oxide modulation is involved in their protective effect of antidepressants against 3-NP induced cognitive dysfunction in rats.
Collapse
|
50
|
Wu CL, Hwang CS, Chen SD, Yin JH, Yang DI. Neuroprotective mechanisms of brain-derived neurotrophic factor against 3-nitropropionic acid toxicity: therapeutic implications for Huntington's disease. Ann N Y Acad Sci 2010; 1201:8-12. [PMID: 20649532 DOI: 10.1111/j.1749-6632.2010.05628.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
3-Nitropropionic acid (3-NP) is an irreversible inhibitor of mitochondrial succinate dehydrogenase that has been used to explore the molecular mechanisms of cell death associated with mitochondrial dysfunction and neurodegeneration for Huntington's disease (HD). Brain-derived neurotrophic factor (BDNF) is a neurotrophin that may regulate neuronal survival and differentiation. Experimental evidence derived from both clinical as well as basic research suggests a close association between BDNF deficiency and HD pathogenesis. In this review, we focus on recent progress in the molecular mechanisms responsible for the BDNF-mediated neuroprotective effects against mitochondrial dysfunction induced by 3-NP. Delineation of BDNF-mediated neuroprotective actions against 3-NP toxicity may add in the development of therapeutic intervention for HD where mitochondrial dysfunction is known to play a crucial role in pathogenesis of this devastating disease.
Collapse
Affiliation(s)
- Chia-Lin Wu
- Institute of Brain Science and Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|