1
|
Nassrallah WB, Cheng J, Mackay JP, Hogg PW, Raymond LA. Mechanisms of synapse-to-nucleus calcium signalling in striatal neurons and impairments in Huntington's disease. J Neurochem 2024; 168:2671-2689. [PMID: 38770573 DOI: 10.1111/jnc.16132] [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: 11/14/2023] [Revised: 04/15/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Huntington's disease (HD) is a monogenic disorder with autosomal dominant inheritance. In HD patients, neurons in the striatum and cortex degenerate, leading to motor, psychiatric and cognitive disorders. Dysregulated synaptic function and calcium handling are common in many neurodegenerative diseases, including HD. N-methyl-D-aspartate (NMDA) receptor function is enhanced in HD at extrasynaptic sites, altering the balance of calcium-dependent neuronal survival versus death signalling pathways. Endoplasmic reticulum (ER) calcium handling is also abnormal in HD. The ER, which is continuous with the nuclear envelope, is purportedly involved in nuclear calcium signalling; based on this, we hypothesised that nuclear calcium signalling is altered in HD. We explored this hypothesis with calcium imaging techniques, including simultaneous epifluorescent imaging of cytosolic and nuclear calcium using jRCaMP1b and GCaMP3 sensors, respectively, in striatal spiny projection neurons in cortical-striatal co-cultures from the YAC128 mouse model of HD. Our data show contributions from a variety of calcium channels to nuclear calcium signalling. NMDA receptors (NMDARs) play an essential role in initiating action potential-dependent calcium signalling to the nucleus, and ryanodine receptors (RyR) contribute to both cytosolic and nuclear calcium signals. Unlike previous reports in glutamatergic hippocampal and cortical neurons, we found that in GABAergic striatal neurons, L-type voltage-gated calcium channels (CaV) contribute to cytosolic, but not nuclear calcium signalling. Calcium imaging also suggests impairments in nuclear calcium signalling in HD striatal neurons, where spontaneous action potential-dependent calcium transients in the nucleus were smaller in YAC128 striatal neurons compared to those of wild-type (WT). Our results elucidate mechanisms involved in action potential-dependent nuclear calcium signalling in GABAergic striatal neurons, and have revealed a clear deficit in this signalling in HD.
Collapse
Affiliation(s)
- Wissam B Nassrallah
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Judy Cheng
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - James P Mackay
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter W Hogg
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lynn A Raymond
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Khayachi A, Abuzgaya M, Liu Y, Jiao C, Dejgaard K, Schorova L, Kamesh A, He Q, Cousineau Y, Pietrantonio A, Farhangdoost N, Castonguay CE, Chaumette B, Alda M, Rouleau GA, Milnerwood AJ. Akt and AMPK activators rescue hyperexcitability in neurons from patients with bipolar disorder. EBioMedicine 2024; 104:105161. [PMID: 38772282 PMCID: PMC11134542 DOI: 10.1016/j.ebiom.2024.105161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Bipolar disorder (BD) is a multifactorial psychiatric illness affecting ∼1% of the global adult population. Lithium (Li), is the most effective mood stabilizer for BD but works only for a subset of patients and its mechanism of action remains largely elusive. METHODS In the present study, we used iPSC-derived neurons from patients with BD who are responsive (LR) or not (LNR) to lithium. Combined electrophysiology, calcium imaging, biochemistry, transcriptomics, and phosphoproteomics were employed to provide mechanistic insights into neuronal hyperactivity in BD, investigate Li's mode of action, and identify alternative treatment strategies. FINDINGS We show a selective rescue of the neuronal hyperactivity phenotype by Li in LR neurons, correlated with changes to Na+ conductance. Whole transcriptome sequencing in BD neurons revealed altered gene expression pathways related to glutamate transmission, alterations in cell signalling and ion transport/channel activity. We found altered Akt signalling as a potential therapeutic effect of Li in LR neurons from patients with BD, and that Akt activation mimics Li effect in LR neurons. Furthermore, the increased neural network activity observed in both LR & LNR neurons from patients with BD were reversed by AMP-activated protein kinase (AMPK) activation. INTERPRETATION These results suggest potential for new treatment strategies in BD, such as Akt activators in LR cases, and the use of AMPK activators for LNR patients with BD. FUNDING Supported by funding from ERA PerMed, Bell Brain Canada Mental Research Program and Brain & Behavior Research Foundation.
Collapse
Affiliation(s)
- Anouar Khayachi
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| | - Malak Abuzgaya
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Yumin Liu
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Chuan Jiao
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Kurt Dejgaard
- McIntyre Institute, Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Lenka Schorova
- McGill University Health Center Research Institute, Montréal, Quebec, Canada
| | - Anusha Kamesh
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Qin He
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Yuting Cousineau
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Alessia Pietrantonio
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Nargess Farhangdoost
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Charles-Etienne Castonguay
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Boris Chaumette
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France; GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France; Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada; Department of Human Genetics, McGill University, Montréal, Quebec, Canada.
| | - Austen J Milnerwood
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
3
|
Wang Y, Ramandi D, Sepers MD, Mackay JP, Raymond LA. Age- and region-dependent cortical excitability in the zQ175 Huntington disease mouse model. Hum Mol Genet 2024; 33:387-399. [PMID: 37947186 PMCID: PMC10877458 DOI: 10.1093/hmg/ddad191] [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: 07/17/2023] [Revised: 09/29/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
The neurodegenerative disorder, Huntington disease (HD), manifests as disorders of movement, cognition and mood. Although studies report abnormal corticostriatal synaptic function early in HD mouse models, less is known about cortical-cortical activity across brain regions and disease stages. Recently, we reported enhanced mesoscale spread of cortical responses to sensory stimulation in vivo at early-manifest stages of two HD mouse models. Here, we investigated cortical excitability of zQ175 HD-model mice compared to their wild-type littermates across different cell types, ages and/or cortical regions using ex vivo electrophysiology. Cortical pyramidal neurons (CPNs) in somatosensory cortex of zQ175 mice showed intrinsic hyper-excitability at 3-4 months, but hypo-excitability at early-manifest stage (8-9 months); reduced frequency of spontaneous excitatory postsynaptic currents (sEPSCs) was seen at both ages. In contrast, motor cortex CPNs in early-manifest zQ175 mice showed increased intrinsic excitability and sEPSC frequency. Large-amplitude excitatory discharges recorded from CPNs in early-manifest zQ175 mice showed increased frequency only in somatosensory cortex, suggesting the intrinsic hypo-excitability of these CPNs may be compensatory against cortical network hyper-excitability. Similarly, in early-manifest zQ175 mice, region-dependent differences were seen in fast-spiking interneurons (FSIs): somatosensory but not motor FSIs from early-manifest zQ175 mice had reduced intrinsic excitability. Moreover, CPNs showed decreased frequency of spontaneous inhibitory postsynaptic currents and increased excitatory-inhibitory (E-I) balance of evoked synaptic currents in somatosensory cortex. Aberrant large-amplitude discharges and reduced inhibitory drive may therefore underlie E-I imbalances that result in circuit changes and synaptic dysfunction in early-manifest HD.
Collapse
Affiliation(s)
- Yundi Wang
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - Daniel Ramandi
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
- Graduate Program in Cell and Developmental Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 2A1, Canada
| | - Marja D Sepers
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - James P Mackay
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - Lynn A Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| |
Collapse
|
4
|
Nassrallah WB, Ramandi D, Cheng J, Oh J, Mackay J, Sepers MD, Lau D, Bading H, Raymond LA. Activin A targets extrasynaptic NMDA receptors to ameliorate neuronal and behavioral deficits in a mouse model of Huntington disease. Neurobiol Dis 2023; 189:106360. [PMID: 37992785 DOI: 10.1016/j.nbd.2023.106360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/08/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023] Open
Abstract
Cortical-striatal synaptic dysfunction, including enhanced toxic signaling by extrasynaptic N-methyl-d-aspartate receptors (eNMDARs), precedes neurodegeneration in Huntington disease (HD). A previous study showed Activin A, whose transcription is upregulated by calcium influx via synaptic NMDARs, suppresses eNMDAR signaling. Therefore, we examined the role of Activin A in the YAC128 HD mouse model, comparing it to wild-type controls. We found decreased Activin A secretion in YAC128 cortical-striatal co-cultures, while Activin A overexpression in this model rescued altered eNMDAR expression. Striatal overexpression of Activin A in vivo improved motor learning on the rotarod task, and normalized striatal neuronal eNMDAR-mediated currents, membrane capacitance and spontaneous excitatory postsynaptic current frequency in the YAC128 mice. These results support the therapeutic potential of Activin A signaling and targeting eNMDARs to restore striatal neuronal health and ameliorate behavioral deficits in HD.
Collapse
Affiliation(s)
- Wissam B Nassrallah
- Graduate Program in Neuroscience, University of British Columbia, Canada; University of British Columbia, Vancouver, BC, Canada; Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Daniel Ramandi
- Graduate Program in Cell and Developmental Biology, University of British Columbia, Canada; Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Judy Cheng
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Jean Oh
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - James Mackay
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Marja D Sepers
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - David Lau
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada; Department of Medicine, Division of Neurology, University of British Columbia, Canada.
| |
Collapse
|
5
|
Sharma R, Neupane C, Pham TL, Lee M, Lee S, Lee SY, Nam MH, Kim CS, Park JB. Tonic Activation of NR2D-Containing NMDARs Exacerbates Dopaminergic Neuronal Loss in MPTP-Injected Parkinsonian Mice. J Neurosci 2023; 43:7730-7744. [PMID: 37726169 PMCID: PMC10648527 DOI: 10.1523/jneurosci.1955-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
NR2D subunit-containing NMDA receptors (NMDARs) gradually disappear during brain maturation but can be recruited by pathophysiological stimuli in the adult brain. Here, we report that 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication recruited NR2D subunit-containing NMDARs that generated an Mg2+-resistant tonic NMDA current (INMDA) in dopaminergic (DA) neurons in the midbrain of mature male mice. MPTP selectively generated an Mg2+-resistant tonic INMDA in DA neurons in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA). Consistently, MPTP increased NR2D but not NR2B expression in the midbrain regions. Pharmacological or genetic NR2D interventions abolished the generation of Mg2+-resistant tonic INMDA in SNpc DA neurons, and thus attenuated subsequent DA neuronal loss and gait deficits in MPTP-treated mice. These results show that extrasynaptic NR2D recruitment generates Mg2+-resistant tonic INMDA and exacerbates DA neuronal loss, thus contributing to MPTP-induced Parkinsonism. The state-dependent NR2D recruitment could be a novel therapeutic target for mitigating cell type-specific neuronal death in neurodegenerative diseases.SIGNIFICANCE STATEMENT NR2D subunit-containing NMDA receptors (NMDARs) are widely expressed in the brain during late embryonic and early postnatal development, and then downregulated during brain maturation and preserved at low levels in a few regions of the adult brain. Certain stimuli can recruit NR2D subunits to generate tonic persistent NMDAR currents in nondepolarized neurons in the mature brain. Our results show that MPTP intoxication recruits NR2D subunits in midbrain dopaminergic (DA) neurons, which leads to tonic NMDAR current-promoting dopaminergic neuronal death and consequent abnormal gait behavior in the MPTP mouse model of Parkinson's disease (PD). This is the first study to indicate that extrasynaptic NR2D recruitment could be a target for preventing neuronal death in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ramesh Sharma
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Chiranjivi Neupane
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Thuy Linh Pham
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Miae Lee
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Cuk-Seong Kim
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jin Bong Park
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| |
Collapse
|
6
|
Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Mares-Barbosa TB, Patrón-Soberano A, Howe AG, Portales-Pérez DP, Miquelajáuregui Graf A, Estrada-Sánchez AM. Revealing the contribution of astrocytes to glutamatergic neuronal transmission. Front Cell Neurosci 2023; 16:1037641. [PMID: 36744061 PMCID: PMC9893894 DOI: 10.3389/fncel.2022.1037641] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Research on glutamatergic neurotransmission has focused mainly on the function of presynaptic and postsynaptic neurons, leaving astrocytes with a secondary role only to ensure successful neurotransmission. However, recent evidence indicates that astrocytes contribute actively and even regulate neuronal transmission at different levels. This review establishes a framework by comparing glutamatergic components between neurons and astrocytes to examine how astrocytes modulate or otherwise influence neuronal transmission. We have included the most recent findings about the role of astrocytes in neurotransmission, allowing us to understand the complex network of neuron-astrocyte interactions. However, despite the knowledge of synaptic modulation by astrocytes, their contribution to specific physiological and pathological conditions remains to be elucidated. A full understanding of the astrocyte's role in neuronal processing could open fruitful new frontiers in the development of therapeutic applications.
Collapse
Affiliation(s)
- Ares Orlando Cuellar-Santoyo
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Victor Manuel Ruiz-Rodríguez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Teresa Belem Mares-Barbosa
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | - Araceli Patrón-Soberano
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Andrew G. Howe
- Intelligent Systems Laboratory, HRL Laboratories, LLC, Malibu, CA, United States
| | - Diana Patricia Portales-Pérez
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | | | - Ana María Estrada-Sánchez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| |
Collapse
|
7
|
Sawant N, Morton H, Kshirsagar S, Reddy AP, Reddy PH. Mitochondrial Abnormalities and Synaptic Damage in Huntington's Disease: a Focus on Defective Mitophagy and Mitochondria-Targeted Therapeutics. Mol Neurobiol 2021; 58:6350-6377. [PMID: 34519969 DOI: 10.1007/s12035-021-02556-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022]
Abstract
Huntington's disease (HD) is a fatal and pure genetic disease with a progressive loss of medium spiny neurons (MSN). HD is caused by expanded polyglutamine repeats in the exon 1 of HD gene. Clinically, HD is characterized by chorea, seizures, involuntary movements, dystonia, cognitive decline, intellectual impairment, and emotional disturbances. Several years of intense research revealed that multiple cellular changes, including defective axonal transport, protein-protein interactions, defective bioenergetics, calcium dyshomeostasis, NMDAR activation, synaptic damage, mitochondrial abnormalities, and selective loss of medium spiny neurons are implicated in HD. Recent research on mutant huntingtin (mHtt) and mitochondria has found that mHtt interacts with the mitochondrial division protein, dynamin-related protein 1 (DRP1), enhances GTPase DRP1 enzymatic activity, and causes excessive mitochondrial fragmentation and abnormal distribution, leading to defective axonal transport of mitochondria and selective synaptic degeneration. Recent research also revealed that failure to remove dead and/or dying mitochondria is an early event in the disease progression. Currently, efforts are being made to reduce abnormal protein interactions and enhance synaptic mitophagy as therapeutic strategies for HD. The purpose of this article is to discuss recent research in HD progression. This article also discusses recent developments of cell and mouse models, cellular changes, mitochondrial abnormalities, DNA damage, bioenergetics, oxidative stress, mitophagy, and therapeutics strategies in HD.
Collapse
Affiliation(s)
- Neha Sawant
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hallie Morton
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neurology, Department of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Internal Medicine, Cell Biology & Biochemistry, Public Health and School of Health Professions, Texas Tech University Health Sciences Center, Neuroscience & Pharmacology3601 4th Street, NeurologyLubbock, TX, 79430, USA.
| |
Collapse
|
8
|
C57BL/6 Background Attenuates mHTT Toxicity in the Striatum of YAC128 Mice. Int J Mol Sci 2021; 22:ijms222312664. [PMID: 34884469 PMCID: PMC8657915 DOI: 10.3390/ijms222312664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/05/2022] Open
Abstract
Mouse models are frequently used to study Huntington’s disease (HD). The onset and severity of neuronal and behavioral pathologies vary greatly between HD mouse models, which results from different huntingtin expression levels and different CAG repeat length. HD pathology appears to depend also on the strain background of mouse models. Thus, behavioral deficits of HD mice are more severe in the FVB than in the C57BL/6 background. Alterations in medium spiny neuron (MSN) morphology and function have been well documented in young YAC128 mice in the FVB background. Here, we tested the relevance of strain background for mutant huntingtin (mHTT) toxicity on the cellular level by investigating HD pathologies in YAC128 mice in the C57BL/6 background (YAC128/BL6). Morphology, spine density, synapse function and membrane properties were not or only subtly altered in MSNs of 12-month-old YAC128/BL6 mice. Despite the mild cellular phenotype, YAC128/BL6 mice showed deficits in motor performance. More pronounced alterations in MSN function were found in the HdhQ150 mouse model in the C57BL/6 background (HdhQ150/BL6). Consistent with the differences in HD pathology, the number of inclusion bodies was considerably lower in YAC128/BL6 mice than HdhQ150/BL6 mice. This study highlights the relevance of strain background for mHTT toxicity in HD mouse models.
Collapse
|
9
|
Jabłońska M, Grzelakowska K, Wiśniewski B, Mazur E, Leis K, Gałązka P. Pridopidine in the treatment of Huntington's disease. Rev Neurosci 2021; 31:441-451. [PMID: 32083454 DOI: 10.1515/revneuro-2019-0085] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/06/2019] [Indexed: 01/26/2023]
Abstract
Huntington's disease (HD) is a highly common inherited monogenic neurodegenerative disease, and the gene responsible for its development is located in the 4p16.3 chromosome. The product of that gene mutation is an abnormal huntingtin (Htt) protein that disrupts the neural conduction, thus leading to motor and cognitive disorders. The disease progresses to irreversible changes in the central nervous system (CNS). Although only a few drugs are available to symptomatic treatment, 'dopamine stabilizers' (as represented by the pridopidine) may be the new treatment options. The underlying causes of HD are dopaminergic conduction disorders. Initially, the disease is hyperkinetic (chorea) until it eventually reaches the hypokinetic phase. Studies confirmed a correlation between the amount of dopamine in the CNS and the stage of the disease. Pridopidine has the capacity to be a dopamine buffer, which could increase or decrease the dopamine content depending on the disease phase. A research carried out on animal models demonstrated the protective effect of pridopidine on nerve cells thanks to its ability to alter the cortical glutamatergic signaling through the N-methyl-D-aspartate (NMDA) receptors. Studies on dopamine stabilizers also reported that pridopidine has a 100-fold greater affinity for the sigma-1 receptor than for the D2 receptor. Disturbances in the activity of sigma-1 receptors occur in neurodegenerative diseases, including HD. Their interaction with pridopidine results in the neuroprotective effect, which is manifested as an increase in the plasticity of synaptic neurons and prevention of their atrophy within the striatum. To determine the effectiveness of pridopidine in the treatment of HD, large multicenter randomized studies such as HART, MermaiHD, and PRIDE-HD were carried out.
Collapse
Affiliation(s)
- Magdalena Jabłońska
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Klaudyna Grzelakowska
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Bartłomiej Wiśniewski
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Ewelina Mazur
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Kamil Leis
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Przemysław Gałązka
- Department of General and Oncological Pediatric Surgery, Antoni Jurasz University Hospital No. 1 in Bydgoszcz, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| |
Collapse
|
10
|
Kadgien CA, Kamesh A, Milnerwood AJ. Endosomal traffic and glutamate synapse activity are increased in VPS35 D620N mutant knock-in mouse neurons, and resistant to LRRK2 kinase inhibition. Mol Brain 2021; 14:143. [PMID: 34530877 PMCID: PMC8447518 DOI: 10.1186/s13041-021-00848-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Vacuolar protein sorting 35 (VPS35) regulates neurotransmitter receptor recycling from endosomes. A missense mutation (D620N) in VPS35 leads to autosomal-dominant, late-onset Parkinson's disease. Here, we study the basic neurobiology of VPS35 and Parkinson's disease mutation effects in the D620N knock-in mouse and the effect of leucine-rich repeat kinase 2 (LRRK2) inhibition on synaptic phenotypes. The study was conducted using a VPS35 D620N knock-in mouse that expresses VPS35 at endogenous levels. Protein levels, phosphorylation states, and binding ratios in brain lysates from knock-in mice and wild-type littermates were assayed by co-immunoprecipitation and western blot. Dendritic protein co-localization, AMPA receptor surface expression, synapse density, and glutamatergic synapse activity in primary cortical cultures from knock-in and wild-type littermates were assayed using immunocytochemistry and whole-cell patch clamp electrophysiology. In brain tissue, we confirm VPS35 forms complexes with LRRK2 and AMPA-type glutamate receptor GluA1 subunits, in addition to NMDA-type glutamate receptor GluN1 subunits and D2-type dopamine receptors. Receptor and LRRK2 binding was unaltered in D620N knock-in mice, but we confirm the mutation results in reduced binding of VPS35 with WASH complex member FAM21, and increases phosphorylation of the LRRK2 kinase substrate Rab10, which is reversed by LRRK2 kinase inhibition in vivo. In cultured cortical neurons from knock-in mice, pRab10 is also increased, and reversed by LRRK2 inhibition. The mutation also results in increased endosomal recycling protein cluster density (VPS35-FAM21 co-clusters and Rab11 clusters), glutamate transmission, and GluA1 surface expression. LRRK2 kinase inhibition, which reversed Rab10 hyper-phosphorylation, did not rescue elevated glutamate release or surface GluA1 expression in knock-in neurons, but did alter AMPAR traffic in wild-type cells. The results improve our understanding of the cell biology of VPS35, and the consequences of the D620N mutation in developing neuronal networks. Together the data support a chronic synaptopathy model for latent neurodegeneration, providing phenotypes and candidate pathophysiological stresses that may drive eventual transition to late-stage parkinsonism in VPS35 PD. The study demonstrates the VPS35 mutation has effects that are independent of ongoing LRRK2 kinase activity, and that LRRK2 kinase inhibition alters basal physiology of glutamate synapses in vitro.
Collapse
Affiliation(s)
- Chelsie A Kadgien
- Graduate Program in Neuroscience and Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Anusha Kamesh
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Austen J Milnerwood
- Graduate Program in Neuroscience and Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada.
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada.
| |
Collapse
|
11
|
Iuliano M, Seeley C, Sapp E, Jones EL, Martin C, Li X, DiFiglia M, Kegel-Gleason KB. Disposition of Proteins and Lipids in Synaptic Membrane Compartments Is Altered in Q175/Q7 Huntington's Disease Mouse Striatum. Front Synaptic Neurosci 2021; 13:618391. [PMID: 33815086 PMCID: PMC8013775 DOI: 10.3389/fnsyn.2021.618391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
Dysfunction at synapses is thought to be an early change contributing to cognitive, psychiatric and motor disturbances in Huntington's disease (HD). In neurons, mutant Huntingtin collects in aggregates and distributes to the same sites as wild-type Huntingtin including on membranes and in synapses. In this study, we investigated the biochemical integrity of synapses in HD mouse striatum. We performed subcellular fractionation of striatal tissue from 2 and 6-month old knock-in Q175/Q7 HD and Q7/Q7 mice. Compared to striata of Q7/Q7 mice, proteins including GLUT3, Na+/K+ ATPase, NMDAR 2b, PSD95, and VGLUT1 had altered distribution in Q175/Q7 HD striata of 6-month old mice but not 2-month old mice. These proteins are found on plasma membranes and pre- and postsynaptic membranes supporting hypotheses that functional changes at synapses contribute to cognitive and behavioral symptoms of HD. Lipidomic analysis of mouse fractions indicated that compared to those of wild-type, fractions 1 and 2 of 6 months Q175/Q7 HD had altered levels of two species of PIP2, a phospholipid involved in synaptic signaling, increased levels of cholesterol ester and decreased cardiolipin species. At 2 months, increased levels of species of acylcarnitine, phosphatidic acid and sphingomyelin were measured. EM analysis showed that the contents of fractions 1 and 2 of Q7/Q7 and Q175/Q7 HD striata had a mix of isolated synaptic vesicles, vesicle filled axon terminals singly or in clusters, and ER and endosome-like membranes. However, those of Q175/Q7 striata contained significantly fewer and larger clumps of particles compared to those of Q7/Q7. Human HD postmortem putamen showed differences from control putamen in subcellular distribution of two proteins (Calnexin and GLUT3). Our biochemical, lipidomic and EM analysis show that the presence of the HD mutation conferred age dependent disruption of localization of synaptic proteins and lipids important for synaptic function. Our data demonstrate concrete biochemical changes suggesting altered integrity of synaptic compartments in HD mice that may mirror changes in HD patients and presage cognitive and psychiatric changes that occur in premanifest HD.
Collapse
|
12
|
Kuhlmann N, Wagner Valladolid M, Quesada-Ramírez L, Farrer MJ, Milnerwood AJ. Chronic and Acute Manipulation of Cortical Glutamate Transmission Induces Structural and Synaptic Changes in Co-cultured Striatal Neurons. Front Cell Neurosci 2021; 15:569031. [PMID: 33679324 PMCID: PMC7930618 DOI: 10.3389/fncel.2021.569031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
In contrast to the prenatal topographic development of sensory cortices, striatal circuit organization is slow and requires the functional maturation of cortical and thalamic excitatory inputs throughout the first postnatal month. While mechanisms regulating synapse development and plasticity are quite well described at excitatory synapses of glutamatergic neurons in the neocortex, comparatively little is known of how this translates to glutamate synapses onto GABAergic neurons in the striatum. Here we investigate excitatory striatal synapse plasticity in an in vitro system, where glutamate can be studied in isolation from dopamine and other neuromodulators. We examined pre-and post-synaptic structural and functional plasticity in GABAergic striatal spiny projection neurons (SPNs), co-cultured with glutamatergic cortical neurons. After synapse formation, medium-term (24 h) TTX silencing increased the density of filopodia, and modestly decreased dendritic spine density, when assayed at 21 days in vitro (DIV). Spine reductions appeared to require residual spontaneous activation of ionotropic glutamate receptors. Conversely, chronic (14 days) TTX silencing markedly reduced spine density without any observed increase in filopodia density. Time-dependent, biphasic changes to the presynaptic marker Synapsin-1 were also observed, independent of residual spontaneous activity. Acute silencing (3 h) did not affect presynaptic markers or postsynaptic structures. To induce rapid, activity-dependent plasticity in striatal neurons, a chemical NMDA receptor-dependent “long-term potentiation (LTP)” paradigm was employed. Within 30 min, this increased spine and GluA1 cluster densities, and the percentage of spines containing GluA1 clusters, without altering the presynaptic signal. The results demonstrate that the growth and pruning of dendritic protrusions is an active process, requiring glutamate receptor activity in striatal projection neurons. Furthermore, NMDA receptor activation is sufficient to drive glutamatergic structural plasticity in SPNs, in the absence of dopamine or other neuromodulators.
Collapse
Affiliation(s)
- Naila Kuhlmann
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Lucía Quesada-Ramírez
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Austen J Milnerwood
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
13
|
Schmidt ME, Caron NS, Aly AE, Lemarié FL, Dal Cengio L, Ko Y, Lazic N, Anderson L, Nguyen B, Raymond LA, Hayden MR. DAPK1 Promotes Extrasynaptic GluN2B Phosphorylation and Striatal Spine Instability in the YAC128 Mouse Model of Huntington Disease. Front Cell Neurosci 2020; 14:590569. [PMID: 33250715 PMCID: PMC7674490 DOI: 10.3389/fncel.2020.590569] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022] Open
Abstract
Huntington disease (HD) is a devastating neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. Disrupted cortico-striatal transmission is an early event that contributes to neuronal spine and synapse dysfunction primarily in striatal medium spiny neurons, the most vulnerable cell type in the disease, but also in neurons of other brain regions including the cortex. Although striatal and cortical neurons eventually degenerate, these synaptic and circuit changes may underlie some of the earliest motor, cognitive, and psychiatric symptoms. Moreover, synaptic dysfunction and spine loss are hypothesized to be therapeutically reversible before neuronal death occurs, and restoration of normal synaptic function may delay neurodegeneration. One of the earliest synaptic alterations to occur in HD mouse models is enhanced striatal extrasynaptic NMDA receptor expression and activity. This activity is mediated primarily through GluN2B subunit-containing receptors and is associated with increased activation of cell death pathways, inhibition of survival signaling, and greater susceptibility to excitotoxicity. Death-associated protein kinase 1 (DAPK1) is a pro-apoptotic kinase highly expressed in neurons during development. In the adult brain, DAPK1 becomes re-activated and recruited to extrasynaptic NMDAR complexes during neuronal death, where it phosphorylates GluN2B at S1303, amplifying toxic receptor function. Approaches to reduce DAPK1 activity have demonstrated benefit in animal models of stroke, Alzheimer's disease, Parkinson's disease, and chronic stress, indicating that DAPK1 may be a novel target for neuroprotection. Here, we demonstrate that dysregulation of DAPK1 occurs early in the YAC128 HD mouse model, and contributes to elevated extrasynaptic GluN2B S1303 phosphorylation. Inhibition of DAPK1 normalizes extrasynaptic GluN2B phosphorylation and surface expression, and completely prevents YAC128 striatal spine loss in cortico-striatal co-culture, thus validating DAPK1 as a potential target for synaptic protection in HD and warranting further development of DAPK1-targeted therapies for neurodegeneration.
Collapse
Affiliation(s)
- Mandi E. Schmidt
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Nicholas S. Caron
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Amirah E. Aly
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Fanny L. Lemarié
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Louisa Dal Cengio
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yun Ko
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Nikola Lazic
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Lisa Anderson
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Betty Nguyen
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A. Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Michael R. Hayden
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Kuhlmann N, Milnerwood AJ. A Critical LRRK at the Synapse? The Neurobiological Function and Pathophysiological Dysfunction of LRRK2. Front Mol Neurosci 2020; 13:153. [PMID: 32973447 PMCID: PMC7482583 DOI: 10.3389/fnmol.2020.00153] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/22/2020] [Indexed: 12/25/2022] Open
Abstract
Since the discovery of LRRK2 mutations causal to Parkinson's disease (PD) in the early 2000s, the LRRK2 protein has been implicated in a plethora of cellular processes in which pathogenesis could occur, yet its physiological function remains elusive. The development of genetic models of LRRK2 PD has helped identify the etiological and pathophysiological underpinnings of the disease, and may identify early points of intervention. An important role for LRRK2 in synaptic function has emerged in recent years, which links LRRK2 to other genetic forms of PD, most notably those caused by mutations in the synaptic protein α-synuclein. This point of convergence may provide useful clues as to what drives dysfunction in the basal ganglia circuitry and eventual death of substantia nigra (SN) neurons. Here, we discuss the evolution and current state of the literature placing LRRK2 at the synapse, through the lens of knock-out, overexpression, and knock-in animal models. We hope that a deeper understanding of LRRK2 neurobiology, at the synapse and beyond, will aid the eventual development of neuroprotective interventions for PD, and the advancement of useful treatments in the interim.
Collapse
Affiliation(s)
- Naila Kuhlmann
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Austen J Milnerwood
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Crevier-Sorbo G, Rymar VV, Crevier-Sorbo R, Sadikot AF. Thalamostriatal degeneration contributes to dystonia and cholinergic interneuron dysfunction in a mouse model of Huntington's disease. Acta Neuropathol Commun 2020; 8:14. [PMID: 32033588 PMCID: PMC7007676 DOI: 10.1186/s40478-020-0878-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/03/2020] [Indexed: 01/18/2023] Open
Abstract
Huntington’s disease (HD) is an autosomal dominant trinucleotide repeat disorder characterized by choreiform movements, dystonia and striatal neuronal loss. Amongst multiple cellular processes, abnormal neurotransmitter signalling and decreased trophic support from glutamatergic cortical afferents are major mechanisms underlying striatal degeneration. Recent work suggests that the thalamostriatal (TS) system, another major source of glutamatergic input, is abnormal in HD although its phenotypical significance is unknown. We hypothesized that TS dysfunction plays an important role in generating motor symptoms and contributes to degeneration of striatal neuronal subtypes. Our results using the R6/2 mouse model of HD indicate that neurons of the parafascicular nucleus (PF), the main source of TS afferents, degenerate at an early stage. PF lesions performed prior to motor dysfunction or striatal degeneration result in an accelerated dystonic phenotype and are associated with premature loss of cholinergic interneurons. The progressive loss of striatal medium spiny neurons and parvalbumin-positive interneurons observed in R6/2 mice is unaltered by PF lesions. Early striatal cholinergic ablation using a mitochondrial immunotoxin provides evidence for increased cholinergic vulnerability to cellular energy failure in R6/2 mice, and worsens the dystonic phenotype. The TS system therefore contributes to trophic support of striatal interneuron subtypes in the presence of neurodegenerative stress, and TS deafferentation may be a novel cell non-autonomous mechanism contributing to the pathogenesis of HD. Furthermore, behavioural experiments demonstrate that the TS system and striatal cholinergic interneurons are key motor-network structures involved in the pathogenesis of dystonia. This work suggests that treatments aimed at rescuing the TS system may preserve important elements of striatal structure and function and provide symptomatic relief in HD.
Collapse
|
16
|
Waters S, Tedroff J, Ponten H, Klamer D, Sonesson C, Waters N. Pridopidine: Overview of Pharmacology and Rationale for its Use in Huntington's Disease. J Huntingtons Dis 2019; 7:1-16. [PMID: 29480206 PMCID: PMC5836399 DOI: 10.3233/jhd-170267] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite advances in understanding the pathophysiology of Huntington’s disease (HD), there are currently no effective pharmacological agents available to treat core symptoms or to stop or prevent the progression of this hereditary neurodegenerative disorder. Pridopidine, a novel small molecule compound, has demonstrated potential for both symptomatic treatment and disease modifying effects in HD. While pridopidine failed to achieve its primary efficacy outcomes (Modified motor score) in two trials (MermaiHD and HART) there were consistent effects on secondary outcomes (TMS). In the most recent study (PrideHD) pridiopidine did not differ from placebo on TMS, possibly due to a large enduring placebo effect. This review describes the process, based on in vivo systems response profiling, by which pridopidine was discovered and discusses its pharmacological profile, aiming to provide a model for the system-level effects, and a rationale for the use of pridopidine in patients affected by HD. Considering the effects on brain neurochemistry, gene expression and behaviour in vivo, pridopidine displays a unique effect profile. A hallmark feature in the behavioural pharmacology of pridopidine is its state-dependent inhibition or activation of dopamine-dependent psychomotor functions. Such effects are paralleled by strengthening of synaptic connectivity in cortico-striatal pathways suggesting pridopidine has potential to modify phenotypic expression as well as progression of HD. The preclinical pharmacological profile is discussed with respect to the clinical results for pridopidine, and proposals are made for further investigation, including preclinical and clinical studies addressing disease progression and effects at different stages of HD.
Collapse
Affiliation(s)
- Susanna Waters
- Department of Pharmacology, University of Gothenburg, Gothenburg, Sweden.,Integrative Research Laboratories AB, Gothenburg, Sweden.,Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joakim Tedroff
- Department of Clinical Neurosciences, Karolinska Institute, Stockholm, Sweden.,Integrative Research Laboratories AB, Gothenburg, Sweden
| | - Henrik Ponten
- Department of Pharmacology, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Klamer
- Department of Pharmacology, University of Gothenburg, Gothenburg, Sweden
| | - Clas Sonesson
- Integrative Research Laboratories AB, Gothenburg, Sweden
| | | |
Collapse
|
17
|
Ambroziak W, Fourie C, Montgomery JM. SAP97-mediated rescue of NMDA receptor surface distribution in a neuronal model of Huntington's disease. Hippocampus 2019; 28:707-723. [PMID: 30067285 DOI: 10.1002/hipo.22995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/22/2018] [Accepted: 05/29/2018] [Indexed: 01/10/2023]
Abstract
Huntington's disease (HD) is a genetic neurodegenerative disorder caused by an expansion of the CAG repeat tract in the HTT gene, leading to motor, cognitive, and psychiatric symptoms. At the cellular level, NMDA-type glutamate receptors are upregulated at glutamatergic extrasynaptic sites in HD, triggering cell death signaling pathways and driving HD neurodegeneration. Extrasynaptic and synaptic glutamate receptor trafficking and surface distribution are regulated by the α and β N-terminal isoforms of SAP97, a postsynaptic density protein localized at glutamatergic synapses. Here we examined the surface distribution of NMDARs and AMPARs in a cellular model of HD, and whether the manipulation of individual SAP97 isoforms can regulate receptor distribution in diseased neurons. Using dSTORM super-resolution imaging, we reveal that mutant HTT drives the elevation of extrasynaptic NMDAR clusters located 100-500 nm from the postsynaptic density. This was accompanied by a decline in synaptic NMDAR-mediated currents while surface NMDAR-mediated currents remained unchanged. These effects were induced within 3 days of mutant HTT expression in rat hippocampal neurons in vitro, and were specific for NMDARs and not observed with AMPARs. Intriguingly, upregulation of either α- or βSAP97 expression increased synaptic and/or perisynaptic NMDAR localization and prevented the shift of NMDARs to extrasynaptic sites in mutant HTT neurons. This was accompanied by the rescue of normal synaptic NMDAR-mediated currents. Taken together, our high-resolution data reveals plasticity in surface NMDAR localization driven by mutant HTT and identifies the similar but independent roles of SAP97 N-terminal isoforms in maintaining normal synaptic function in pathological states.
Collapse
Affiliation(s)
- Wojciech Ambroziak
- Department of Physiology and Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Chantelle Fourie
- Department of Physiology and Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology and Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
Singh R, Ganeshpurkar A, Kumar D, Kumar D, Kumar A, Singh SK. Identifying potential GluN2B subunit containing N-Methyl-D-aspartate receptor inhibitors: an integrative in silico and molecular modeling approach. J Biomol Struct Dyn 2019; 38:2533-2545. [PMID: 31232189 DOI: 10.1080/07391102.2019.1635530] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs), a class of ligand-gated ion channels, are involved in non-selective cation transport across the membrane. These are contained in glutamatergic synapse and produce excitatory effects leading to synaptic plasticity and memory function. GluN1-GluN2B, a subtype of NMDAR(s), has significant role in neurodegeneration, amyloid β (Aβ) induced synaptic dysfunction and loss. Thus, targeting and inhibiting GluN1-GluN2B may be effective in the management of neurodegenerative diseases including Alzheimer's disease. In the present study, ligand and structure-based approaches were tried to identify the inhibitors. The pharmacophore, developed from co-crystallised ifenprodil, afforded virtual hits, which were further subjected through drug likeliness and PAINS filters to remove interfering compounds. Further comprehensive docking studies, free energy calculations and ADMET studies resulted in two virtual leads. The leads, ZINC257261614 and ZINC95977857 displayed good docking scores of -12.90 and -12.20 Kcal/mol and free binding energies of -60.83 and -61.83 Kcal/mol, respectively. The compounds were having acceptable predicted ADMET profiles and were subjected to molecular dynamic (MD) studies. The MD simulation produced stable complexes of these ligands with GluN1-GluN2B subunit having protein and ligand RMSD in acceptable limit. AbbreviationsADAlzheimer's diseaseADMEAbsorption distribution metabolism and excretionATDAmino terminal domainBBBBlood-brain barrierCNSCentral nervous systemCREBcAMP response element binding proteinCTDCarboxy-terminal domainGluGlutamateGMQEGlobal model quality estimationHTVSHigh throughput virtual screeningHIAHuman intestinal absorptionLGALamarckian genetic algorithmMDMolecular dynamicsMM-GBSAMolecular mechanics, the Generalised Born model for Solvent AccessibilityNMDARN-methyl-D-aspartate receptorsPAINSPan assay interference compoundsRMSDRoot-mean square deviationRMSFRoot-mean-square fluctuationSMARTSSMILES arbitrary target specificationSPstandard precisionXPextra precisionCommunicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ravi Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Ankit Ganeshpurkar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Devendra Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharti Vidyapeeth University, Pune, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
19
|
Smith‐Dijak AI, Sepers MD, Raymond LA. Alterations in synaptic function and plasticity in Huntington disease. J Neurochem 2019; 150:346-365. [DOI: 10.1111/jnc.14723] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/28/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Amy I. Smith‐Dijak
- Graduate Program in Neuroscience the University of British Columbia Vancouver British Columbia Canada
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| | - Marja D. Sepers
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| | - Lynn A. Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
20
|
Smith-Dijak AI, Nassrallah WB, Zhang LYJ, Geva M, Hayden MR, Raymond LA. Impairment and Restoration of Homeostatic Plasticity in Cultured Cortical Neurons From a Mouse Model of Huntington Disease. Front Cell Neurosci 2019; 13:209. [PMID: 31156395 PMCID: PMC6532531 DOI: 10.3389/fncel.2019.00209] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Huntington disease (HD) is an inherited neurodegenerative disorder caused by a mutation in the huntingtin gene. The onset of symptoms is preceded by synaptic dysfunction. Homeostatic synaptic plasticity (HSP) refers to processes that maintain the stability of networks of neurons, thought to be required to enable new learning and cognitive flexibility. One type of HSP is synaptic scaling, in which the strength of all of the synapses onto a cell increases or decreases following changes in the cell’s level of activity. Several pathways implicated in synaptic scaling are dysregulated in HD, including brain-derived neurotrophic factor (BDNF) and calcium signaling. Here, we investigated whether HSP is disrupted in cortical neurons from an HD mouse model. We treated cultured cortical neurons from wild-type (WT) FVB/N or YAC128 HD mice with tetrodotoxin (TTX) for 48 h to silence action potentials and then recorded miniature excitatory postsynaptic currents. In WT cultures, these increased in both amplitude and frequency after TTX treatment, and further experiments showed that this was a result of insertion of AMPA receptors and formation of new synapses, respectively. Manipulation of BDNF concentration in the culture medium revealed that BDNF signaling contributed to these changes. In contrast to WT cortical neurons, YAC128 cultures showed no response to action potential silencing. Strikingly, we were able to restore the TTX-induced changes in YAC128 cultures by treating them with pridopidine, a drug which enhances BDNF signaling through stimulation of the sigma-1 receptor (S1R), and with the S1R agonist 3-PPP. These data provide evidence for disruption of HSP in cortical neurons from an HD mouse model that is restored by stimulation of S1R. Our results suggest a potential new direction for developing therapy to mitigate cognitive deficits in HD.
Collapse
Affiliation(s)
- Amy I Smith-Dijak
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, BC, Canada.,Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| | - Wissam B Nassrallah
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, BC, Canada.,Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| | - Lily Y J Zhang
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| | - Michal Geva
- Research and Development, Teva Pharmaceutical Industries Ltd., Netanya, Israel
| | - Michael R Hayden
- Research and Development, Teva Pharmaceutical Industries Ltd., Netanya, Israel.,Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Kang R, Wang L, Sanders SS, Zuo K, Hayden MR, Raymond LA. Altered Regulation of Striatal Neuronal N-Methyl-D-Aspartate Receptor Trafficking by Palmitoylation in Huntington Disease Mouse Model. Front Synaptic Neurosci 2019; 11:3. [PMID: 30846936 PMCID: PMC6393405 DOI: 10.3389/fnsyn.2019.00003] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) play a critical role in synaptic signaling, and alterations in the synaptic/extrasynaptic NMDAR balance affect neuronal survival. Studies have shown enhanced extrasynaptic GluN2B-type NMDAR (2B-NMDAR) activity in striatal neurons in the YAC128 mouse model of Huntington disease (HD), resulting in increased cell death pathway activation contributing to striatal vulnerability to degeneration. However, the mechanism(s) of altered GluN2B trafficking remains unclear. Previous work shows that GluN2B palmitoylation on two C-terminal cysteine clusters regulates 2B-NMDAR trafficking to the surface membrane and synapses in cortical neurons. Notably, two palmitoyl acyltransferases (PATs), zDHHC17 and zDHHC13, also called huntingtin-interacting protein 14 (HIP14) and HIP14-like (HIP14L), directly interact with the huntingtin protein (Htt), and mutant Htt disrupts this interaction. Here, we investigated whether GluN2B palmitoylation is involved in enhanced extrasynaptic surface expression of 2B-NMDARs in YAC128 striatal neurons and whether this process is regulated by HIP14 or HIP14L. We found reduced GluN2B palmitoylation in YAC128 striatum, specifically on cysteine cluster II. Consistent with that finding, the palmitoylation-deficient GluN2B Cysteine cluster II mutant exhibited enhanced, extrasynaptic surface expression in striatal neurons from wild-type mice, mimicking increased extrasynaptic 2B-NMDAR observed in YAC128 cultures. We also found that HIP14L palmitoylated GluN2B cysteine cluster II. Moreover, GluN2B palmitoylation levels were reduced in striatal tissue from HIP14L-deficient mice, and siRNA-mediated HIP14L knockdown in cultured neurons enhanced striatal neuronal GluN2B surface expression and susceptibility to NMDA toxicity. Thus, altered regulation of GluN2B palmitoylation levels by the huntingtin-associated PAT HIP14L may contribute to the cell death-signaling pathways underlying HD.
Collapse
Affiliation(s)
- Rujun Kang
- Department of Psychiatry, Brain Research Centre and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| | - Liang Wang
- Department of Psychiatry, Brain Research Centre and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| | - Shaun S Sanders
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Kurt Zuo
- Department of Psychiatry, Brain Research Centre and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| | - Michael R Hayden
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Brain Research Centre and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
22
|
Hsu YT, Chang YG, Chern Y. Insights into GABA Aergic system alteration in Huntington's disease. Open Biol 2018; 8:rsob.180165. [PMID: 30518638 PMCID: PMC6303784 DOI: 10.1098/rsob.180165] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant progressive neurodegenerative disease that is characterized by a triad of motor, psychiatric and cognitive impairments. There is still no effective therapy to delay or halt the disease progress. The striatum and cortex are two particularly affected brain regions that exhibit dense reciprocal excitatory glutamate and inhibitory gamma-amino butyric acid (GABA) connections. Imbalance between excitatory and inhibitory signalling is known to greatly affect motor and cognitive processes. Emerging evidence supports the hypothesis that disrupted GABAergic circuits underlie HD pathogenesis. In the present review, we focused on the multiple defects recently found in the GABAergic inhibitory system, including altered GABA level and synthesis, abnormal subunit composition and distribution of GABAA receptors and aberrant GABAA receptor-mediated signalling. In particular, the important role of cation–chloride cotransporters (i.e. NKCC1 and KCC2) is discussed. Recent studies also suggest that neuroinflammation contributes significantly to the abnormal GABAergic inhibition in HD. Thus, GABAA receptors and cation–chloride cotransporters are potential therapeutic targets for HD. Given the limited availability of therapeutic treatments for HD, a better understanding of GABAergic dysfunction in HD could provide novel therapeutic opportunities.
Collapse
Affiliation(s)
- Yi-Ting Hsu
- PhD Program for Translational Medicine, China Medical University and Academia Sinica, Taiwan, Republic of China.,Department of Neurology, China Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Ya-Gin Chang
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan, Republic of China.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yijuang Chern
- PhD Program for Translational Medicine, China Medical University and Academia Sinica, Taiwan, Republic of China .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
23
|
Boussicault L, Kacher R, Lamazière A, Vanhoutte P, Caboche J, Betuing S, Potier MC. CYP46A1 protects against NMDA-mediated excitotoxicity in Huntington's disease: Analysis of lipid raft content. Biochimie 2018; 153:70-79. [DOI: 10.1016/j.biochi.2018.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
|
24
|
Schmidt ME, Buren C, Mackay JP, Cheung D, Dal Cengio L, Raymond LA, Hayden MR. Altering cortical input unmasks synaptic phenotypes in the YAC128 cortico-striatal co-culture model of Huntington disease. BMC Biol 2018; 16:58. [PMID: 29945611 PMCID: PMC6020351 DOI: 10.1186/s12915-018-0526-3] [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: 01/12/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Huntington disease (HD) is a fatal neurodegenerative disorder caused by a CAG expansion in the huntingtin (HTT) gene, leading to selective and progressive neuronal death predominantly in the striatum. Mutant HTT expression causes dysfunctional cortico-striatal (CS) transmission, loss of CS synapses, and striatal medium spiny neuron (MSN) dendritic spine instability prior to neuronal death. Co-culturing cortical and striatal neurons in vitro promotes the formation of functional CS synapses and is a widely used approach to elucidate pathogenic mechanisms of HD and to validate potential synapto-protective therapies. A number of relevant in vivo synaptic phenotypes from the YAC128 HD mouse model, which expresses full-length transgenic human mutant HTT, are recapitulated in CS co-culture by 21 days in vitro (DIV). However, striatal spine loss, which occurs in HD patients and in vivo animal models, has been observed in YAC128 CS co-culture in some studies but not in others, leading to difficulties in reproducing and interpreting results. Here, we investigated whether differences in the relative proportion of cortical and striatal neurons alter YAC128 synaptic phenotypes in this model. RESULTS YAC128 MSNs in 1:1 CS co-culture exhibited impaired dendritic length and complexity compared to wild-type, whereas reducing cortical input using a 1:3 CS ratio revealed a dramatic loss of YAC128 MSN dendritic spines. Chimeric experiments determined that this spine instability was primarily cell autonomous, depending largely on mutant HTT expression in striatal neurons. Moreover, we found that spontaneous electrophysiological MSN activity correlated closely with overall dendritic length, with no differences observed between genotypes in 1:3 co-cultures despite significant YAC128 spine loss. Finally, limiting cortical input with a 1:3 CS ratio impaired the basal survival of YAC128 neurons at DIV21, and this was partially selective for dopamine- and cAMP-regulated phosphoprotein 32-positive MSNs. CONCLUSIONS Our findings reconcile previous discordant reports of spine loss in this model, and improve the utility and reliability of the CS co-culture for the development of novel therapeutic strategies for HD.
Collapse
Affiliation(s)
- Mandi E Schmidt
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, V5Z 4H4, Canada
| | - Caodu Buren
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 4834-2255 Wesbrook Mall, Vancouver, V6T 1Z3, Canada.,Present address: The Hospital for Sick Children, 555 University Avenue, Toronto, M5G 1X8, Canada
| | - James P Mackay
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 4834-2255 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - Daphne Cheung
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, V5Z 4H4, Canada
| | - Louisa Dal Cengio
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, V5Z 4H4, Canada
| | - Lynn A Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 4834-2255 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, V5Z 4H4, Canada.
| |
Collapse
|
25
|
Abstract
Huntington's disease (HD) is one of the most common tandem repeat disorders and presents as a unique trilogy of cognitive, psychiatric and motor symptoms. One of the major mysteries of HD is why it selectively affects specific neuronal populations. A new article in BMC Biology provides a piece in the puzzle of pathogenesis. By demonstrating the delicate relationship between cortical and striatal neurons, it provokes broader questions of how we might understand HD as a disorder of synapses, neural circuits and systems biology.
Collapse
Affiliation(s)
- Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
26
|
Saavedra A, García-Díaz Barriga G, Pérez-Navarro E, Alberch J. Huntington's disease: novel therapeutic perspectives hanging in the balance. Expert Opin Ther Targets 2018; 22:385-399. [PMID: 29671352 DOI: 10.1080/14728222.2018.1465930] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Huntington's disease (HD), an autosomal dominant neurodegenerative disorder caused by an expansion of CAG repeats in the huntingtin gene, has long been characterized by the presence of motor symptoms due to the loss of striatal projection neurons. Cognitive dysfunction and neuropsychiatric symptoms are also present and they occur in the absence of cell death in most mouse models, pointing to neuronal dysfunction and abnormal synaptic plasticity as causative mechanisms. Areas covered: Here, we focus on those common mechanisms altered by the presence of mutant huntingtin affecting corticostriatal and hippocampal function as therapeutic targets that could prove beneficial to ameliorate both cognitive and motor function in HD. Specifically, we discuss the importance of reestablishing the balance in (1) synaptic/extrasynaptic N-methyl-D-aspartate receptor signaling, (2) mitochondrial dynamics/trafficking, (3) TrkB/p75NTR signaling, and (4) transcriptional activity. Expert opinion: Mutant huntingtin has a broad impact on multiple cellular processes, which makes it very challenging to design a curative therapeutic strategy. As we point out here, novel therapeutic interventions should look for multi-purpose drugs targeting common and early affected processes leading to corticostriatal and hippocampal dysfunction that additionally operate in a feedforward vicious cycle downstream the activation of extrasynaptic N-methyl-D-aspartate receptor.
Collapse
Affiliation(s)
- Ana Saavedra
- a Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències , Universitat de Barcelona , Barcelona , Spain.,b Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,c Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain
| | - Gerardo García-Díaz Barriga
- a Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències , Universitat de Barcelona , Barcelona , Spain.,b Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,c Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain
| | - Esther Pérez-Navarro
- a Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències , Universitat de Barcelona , Barcelona , Spain.,b Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,c Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain
| | - Jordi Alberch
- a Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències , Universitat de Barcelona , Barcelona , Spain.,b Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,c Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain
| |
Collapse
|
27
|
Kunkanjanawan T, Carter R, Ahn KS, Yang J, Parnpai R, Chan AWS. Induced Pluripotent HD Monkey Stem Cells Derived Neural Cells for Drug Discovery. SLAS DISCOVERY 2016; 22:696-705. [PMID: 28027448 DOI: 10.1177/2472555216685044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by an expansion of CAG trinucleotide repeat (polyglutamine [polyQ]) in the huntingtin ( HTT) gene, which leads to the formation of mutant HTT (mHTT) protein aggregates. In the nervous system, an accumulation of mHTT protein results in glutamate-mediated excitotoxicity, proteosome instability, and apoptosis. Although HD pathogenesis has been extensively studied, effective treatment of HD has yet to be developed. Therapeutic discovery research in HD has been reported using yeast, cells derived from transgenic animal models and HD patients, and induced pluripotent stem cells from patients. A transgenic nonhuman primate model of HD (HD monkey) shows neuropathological, behavioral, and molecular changes similar to an HD patient. In addition, neural progenitor cells (NPCs) derived from HD monkeys can be maintained in culture and differentiated to neural cells with distinct HD cellular phenotypes including the formation of mHTT aggregates, intranuclear inclusions, and increased susceptibility to oxidative stress. Here, we evaluated the potential application of HD monkey NPCs and neural cells as an in vitro model for HD drug discovery research.
Collapse
Affiliation(s)
- Tanut Kunkanjanawan
- 1 Yerkes National Primate Research Center, Atlanta, GA, USA.,2 Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.,3 Embryo Technology and Stem Cell Research Center, School of Biotechnology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Richard Carter
- 1 Yerkes National Primate Research Center, Atlanta, GA, USA.,2 Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Kwan-Sung Ahn
- 1 Yerkes National Primate Research Center, Atlanta, GA, USA.,2 Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jinjing Yang
- 1 Yerkes National Primate Research Center, Atlanta, GA, USA.,2 Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Rangsun Parnpai
- 3 Embryo Technology and Stem Cell Research Center, School of Biotechnology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Anthony W S Chan
- 1 Yerkes National Primate Research Center, Atlanta, GA, USA.,2 Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
28
|
Ingram T, Chakrabarti L. Proteomic profiling of mitochondria: what does it tell us about the ageing brain? Aging (Albany NY) 2016; 8:3161-3179. [PMID: 27992860 PMCID: PMC5270661 DOI: 10.18632/aging.101131] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/01/2016] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction is evident in numerous neurodegenerative and age-related disorders. It has also been linked to cellular ageing, however our current understanding of the mitochondrial changes that occur are unclear. Functional studies have made some progress reporting reduced respiration, dynamic structural modifications and loss of membrane potential, though there are conflicts within these findings. Proteomic analyses, together with functional studies, are required in order to profile the mitochondrial changes that occur with age and can contribute to unravelling the complexity of the ageing phenotype. The emergence of improved protein separation techniques, combined with mass spectrometry analyses has allowed the identification of age and cell-type specific mitochondrial changes in energy metabolism, antioxidants, fusion and fission machinery, chaperones, membrane proteins and biosynthesis pathways. Here, we identify and review recent data from the analyses of mitochondria from rodent brains. It is expected that knowledge gained from understanding age-related mitochondrial changes of the brain should lead to improved biomarkers of normal ageing and also age-related disease progression.
Collapse
Affiliation(s)
- Thomas Ingram
- SVMS, Faculty of Medicine, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Lisa Chakrabarti
- SVMS, Faculty of Medicine, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| |
Collapse
|
29
|
Majláth Z, Török N, Toldi J, Vécsei L. Memantine and Kynurenic Acid: Current Neuropharmacological Aspects. Curr Neuropharmacol 2016; 14:200-9. [PMID: 26564141 PMCID: PMC4825950 DOI: 10.2174/1570159x14666151113123221] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/22/2015] [Accepted: 12/03/2015] [Indexed: 12/31/2022] Open
Abstract
Glutamatergic neurotransmission, of special importance in the human brain, is implicated in key brain functions such as synaptic plasticity and memory. The excessive activation of N-methyl- D-aspartate (NMDA) receptors may result in excitotoxic neuronal damage; this process has been implicated in the pathomechanism of different neurodegenerative disorders, such as Alzheimer’s disease (AD). Memantine is an uncompetitive antagonist of NMDA receptors with a favorable pharmacokinetic profile, and is therefore clinically well tolerated. Memantine is approved for the treatment of AD, but may additionally be beneficial for other dementia forms and pain conditions. Kynurenic acid
(KYNA) is an endogenous antagonist of NMDA receptors which has been demonstrated under experimental conditions to be neuroprotective. The development of a well-tolerated NMDA antagonist may offer a novel therapeutic option for the treatment of neurodegenerative disease and pain syndromes. KYNA may be a valuable candidate for future drug development.
Collapse
Affiliation(s)
| | | | | | - László Vécsei
- Department of Neurology, Faculty of Medicine, University of Szeged, Albert Szent-Györgyi Clinical Center, Semmelweis u. 6. H-6725 Szeged, Hungary.
| |
Collapse
|
30
|
Raymond LA. Striatal synaptic dysfunction and altered calcium regulation in Huntington disease. Biochem Biophys Res Commun 2016; 483:1051-1062. [PMID: 27423394 DOI: 10.1016/j.bbrc.2016.07.058] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/11/2016] [Indexed: 11/30/2022]
Abstract
Synaptic dysfunction and altered calcium homeostasis in the brain is common to many neurodegenerative disorders. Among these, Huntington disease (HD), which is inherited in an autosomal dominant fashion, can serve as a model for investigating these mechanisms. HD generally manifests in middle age as a disorder of movement, mood and cognition. An expanded polymorphic CAG repeat in the HTT gene results in progressive neurodegeneration that impacts striatal spiny projection neurons (SPNs) earliest and most severely. Striatal SPNs receive massive glutamatergic input from cortex and thalamus, and these excitatory synapses are a focus for early changes that can trigger aberrant downstream signaling to disrupt synaptic plasticity and lead to later degeneration. Mitochondrial dysfunction and altered intracellular calcium-induced calcium release and sequestration mechanisms add to the impairments in circuit function that may underlie prodromal cognitive and subtle motor deficits. These mechanisms and implications for developing disease-modifying therapy will be reviewed here.
Collapse
Affiliation(s)
- Lynn A Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 4834-2255 Wesbrook Mall, Vancouver, BC, Canada, V6T 1Z3.
| |
Collapse
|
31
|
Enhanced Store-Operated Calcium Entry Leads to Striatal Synaptic Loss in a Huntington's Disease Mouse Model. J Neurosci 2016; 36:125-41. [PMID: 26740655 DOI: 10.1523/jneurosci.1038-15.2016] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED In Huntington's disease (HD), mutant Huntingtin (mHtt) protein causes striatal neuron dysfunction, synaptic loss, and eventual neurodegeneration. To understand the mechanisms responsible for synaptic loss in HD, we developed a corticostriatal coculture model that features age-dependent dendritic spine loss in striatal medium spiny neurons (MSNs) from YAC128 transgenic HD mice. Age-dependent spine loss was also observed in vivo in YAC128 MSNs. To understand the causes of spine loss in YAC128 MSNs, we performed a series of mechanistic studies. We previously discovered that mHtt protein binds to type 1 inositol (1,4,5)-trisphosphate receptor (InsP3R1) and increases its sensitivity to activation by InsP3. We now report that the resulting increase in steady-state InsP3R1 activity reduces endoplasmic reticulum (ER) Ca(2+) levels. Depletion of ER Ca(2+) leads to overactivation of the neuronal store-operated Ca(2+) entry (nSOC) pathway in YAC128 MSN spines. The synaptic nSOC pathway is controlled by the ER resident protein STIM2. We discovered that STIM2 expression is elevated in aged YAC128 striatal cultures and in YAC128 mouse striatum. Knock-down of InsP3R1 expression by antisense oligonucleotides or knock-down or knock-out of STIM2 resulted in normalization of nSOC and rescue of spine loss in YAC128 MSNs. The selective nSOC inhibitor EVP4593 was identified in our previous studies. We now demonstrate that EVP4593 reduces synaptic nSOC and rescues spine loss in YAC128 MSNs. Intraventricular delivery of EVP4593 in YAC128 mice rescued age-dependent striatal spine loss in vivo. Our results suggest EVP4593 and other inhibitors of the STIM2-dependent nSOC pathway as promising leads for HD therapeutic development. SIGNIFICANCE STATEMENT In Huntington's disease (HD) mutant Huntingtin (mHtt) causes early corticostriatal synaptic dysfunction and eventual neurodegeneration of medium spine neurons (MSNs) through poorly understood mechanisms. We report here that corticostriatal cocultures prepared from YAC128 HD mice feature age-dependent MSN spine loss, mirroring YAC128 MSN spine loss in vivo. This finding establishes a system for mechanistic studies of synaptic instability in HD. We use it to demonstrate that sensitization of type 1 inositol (1,4,5)-trisphosphate receptors by mHtt, which depletes endoplasmic reticulum calcium, contributes to synaptotoxic enhancement of STIM2-dependent store-operated calcium (SOC) entry. Treatment with EVP4593, a neuroprotective inhibitor of neuronal SOC channels, rescues YAC128 MSN spine loss both in vitro and in vivo. These results suggest that enhanced neuronal SOC causes synaptic loss in HD-afflicted MSNs.
Collapse
|
32
|
Buren C, Tu G, Parsons MP, Sepers MD, Raymond LA. Influence of cortical synaptic input on striatal neuronal dendritic arborization and sensitivity to excitotoxicity in corticostriatal coculture. J Neurophysiol 2016; 116:380-90. [PMID: 27121581 DOI: 10.1152/jn.00933.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/25/2016] [Indexed: 11/22/2022] Open
Abstract
Corticostriatal cocultures are utilized to recapitulate the cortex-striatum connection in vitro as a convenient model to investigate the development, function, and regulation of synapses formed between cortical and striatal neurons. However, optimization of this dissociated neuronal system to more closely reproduce in vivo circuits has not yet been explored. We studied the effect of varying the plating ratio of cortical to striatal neurons on striatal spiny projection neuron (SPN) characteristics in primary neuronal cocultures. Despite the large difference in cortical-striatal neuron ratio (1:1 vs. 1:3) at day of plating, by 18 days in vitro the difference became modest (∼25% lower cortical-striatal neuron ratio in 1:3 cocultures) and the neuronal density was lower in the 1:3 cocultures, indicating enhanced loss of striatal SPNs. Comparing SPNs in cocultures plated at a 1:1 vs. 1:3 ratio, we found that resting membrane potential, input resistance, current injection-induced action potential firing rates, and input-output curves were similar in the two conditions. However, SPNs in the cocultures plated at the lower cortical ratio exhibited reduced membrane capacitance along with significantly shorter total dendritic length, decreased dendritic complexity, and fewer excitatory synapses, consistent with their trend toward reduced miniature excitatory postsynaptic current frequency. Strikingly, the proportion of NMDA receptors found extrasynaptically in recordings from SPNs was significantly higher in the less cortical coculture. Consistently, SPNs in cocultures with reduced cortical input showed decreased basal pro-survival signaling through cAMP response element binding protein and enhanced sensitivity to NMDA-induced apoptosis. Altogether, our study indicates that abundance of cortical input regulates SPN dendritic arborization and survival/death signaling.
Collapse
Affiliation(s)
- Caodu Buren
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; and
| | - Gaqi Tu
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Matthew P Parsons
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; and
| | - Marja D Sepers
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; and
| | - Lynn A Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; and
| |
Collapse
|
33
|
Real-time imaging of glutamate clearance reveals normal striatal uptake in Huntington disease mouse models. Nat Commun 2016; 7:11251. [PMID: 27052848 PMCID: PMC4829692 DOI: 10.1038/ncomms11251] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/07/2016] [Indexed: 12/17/2022] Open
Abstract
It has become well accepted that Huntington disease (HD) is associated with impaired glutamate uptake, resulting in a prolonged time-course of extracellular glutamate that contributes to excitotoxicity. However, the data supporting this view come largely from work in synaptosomes, which may overrepresent nerve-terminal uptake over astrocytic uptake. Here, we quantify real-time glutamate dynamics in HD mouse models by high-speed imaging of an intensity-based glutamate-sensing fluorescent reporter (iGluSnFR) and electrophysiological recordings of synaptically activated transporter currents in astrocytes. These techniques reveal a disconnect between the results obtained in synaptosomes and those in situ. Exogenous glutamate uptake is impaired in synaptosomes, whereas real-time measures of glutamate clearance in the HD striatum are normal or even accelerated, particularly in the aggressive R6/2 model. Our results highlight the importance of quantifying glutamate dynamics under endogenous release conditions, and suggest that the widely cited uptake impairment in HD does not contribute to pathogenesis. Huntington disease (HD) has been linked via biochemical uptake assays to impaired glutamate clearance and resultant excitotoxicity. Here, utilizing a fluorescent reporter, the authors measure real-time glutamate dynamics in mouse model HD brain slices and find normal or even accelerated glutamate clearance.
Collapse
|
34
|
Impaired development of cortico-striatal synaptic connectivity in a cell culture model of Huntington's disease. Neurobiol Dis 2015; 87:80-90. [PMID: 26711622 DOI: 10.1016/j.nbd.2015.12.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/13/2015] [Accepted: 12/18/2015] [Indexed: 11/21/2022] Open
Abstract
Huntington's disease (HD) is a genetically inherited neurodegenerative disease caused by a mutation in the gene encoding the huntingtin protein. This mutation results in progressive cell death that is particularly striking in the striatum. Recent evidence indicates that early HD is initially a disease of the synapse, in which subtle alterations in synaptic neurotransmission, particularly at the cortico-striatal (C-S) synapse, can be detected well in advance of cell death. Here, we used a cell culture model in which striatal neurons are co-cultured with cortical neurons, and monitored the development of C-S connectivity up to 21days in vitro (DIV) in cells cultured from either the YAC128 mouse model of HD or the background strain, FVB/N (wild-type; WT) mice. Our data demonstrate that while C-S connectivity in WT co-cultures develops rapidly and continuously from DIV 7 to 21, YAC128 C-S connectivity shows no significant growth from DIV 14 onward. Morphological and electrophysiological data suggest that a combination of pre- and postsynaptic mechanisms contribute to this effect, including a reduction in both the postsynaptic dendritic arborization and the size and replenishment rate of the presynaptic readily releasable pool of excitatory vesicles. Moreover, a chimeric culture strategy confirmed that the most robust impairment in C-S connectivity was only observed when mutant huntingtin was expressed both pre- and postsynaptically. In all, our data demonstrate a progressive HD synaptic phenotype in this co-culture system that may be exploited as a platform for identifying promising therapeutic strategies to prevent early HD-associated synaptopathy.
Collapse
|
35
|
Differential changes in thalamic and cortical excitatory synapses onto striatal spiny projection neurons in a Huntington disease mouse model. Neurobiol Dis 2015; 86:62-74. [PMID: 26621114 DOI: 10.1016/j.nbd.2015.11.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/09/2015] [Accepted: 11/23/2015] [Indexed: 01/18/2023] Open
Abstract
Huntington disease (HD), a neurodegenerative disorder caused by CAG repeat expansion in the gene encoding huntingtin, predominantly affects the striatum, especially the spiny projection neurons (SPN). The striatum receives excitatory input from cortex and thalamus, and the role of the former has been well-studied in HD. Here, we report that mutated huntingtin alters function of thalamostriatal connections. We used a novel thalamostriatal (T-S) coculture and an established corticostriatal (C-S) coculture, generated from YAC128 HD and WT (FVB/NJ background strain) mice, to investigate excitatory neurotransmission onto striatal SPN. SPN in T-S coculture from WT mice showed similar mini-excitatory postsynaptic current (mEPSC) frequency and amplitude as in C-S coculture; however, both the frequency and amplitude were significantly reduced in YAC128 T-S coculture. Further investigation in T-S coculture showed similar excitatory synapse density in WT and YAC128 SPN dendrites by immunostaining, suggesting changes in total dendritic length or probability of release as possible explanations for mEPSC frequency changes. Synaptic N-methyl-D-aspartate receptor (NMDAR) current was similar, but extrasynaptic current, associated with cell death signaling, was enhanced in YAC128 SPN in T-S coculture. Employing optical stimulation of cortical versus thalamic afferents and recording from striatal SPN in brain slice, we found increased glutamate release probability and reduced AMPAR/NMDAR current ratios in thalamostriatal synapses, most prominently in YAC128. Enhanced extrasynaptic NMDAR current in YAC128 SPN was apparent with both cortical and thalamic stimulation. We conclude that thalamic afferents to the striatum are affected early, prior to an overt HD phenotype; however, changes in NMDAR localization in SPN are independent of the source of glutamatergic input.
Collapse
|
36
|
Nahavandi S, Tang SY, Baratchi S, Soffe R, Nahavandi S, Kalantar-zadeh K, Mitchell A, Khoshmanesh K. Microfluidic platforms for the investigation of intercellular signalling mechanisms. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:4810-26. [PMID: 25238429 DOI: 10.1002/smll.201401444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 06/27/2014] [Indexed: 05/02/2023]
Abstract
Intercellular signalling has been identified as a highly complex process, responsible for orchestrating many physiological functions. While conventional methods of investigation have been useful, their limitations are impeding further development. Microfluidics offers an opportunity to overcome some of these limitations. Most notably, microfluidic systems can emulate the in-vivo environments. Further, they enable exceptionally precise control of the microenvironment, allowing complex mechanisms to be selectively isolated and studied in detail. There has thus been a growing adoption of microfluidic platforms for investigation of cell signalling mechanisms. This review provides an overview of the different signalling mechanisms and discusses the methods used to study them, with a focus on the microfluidic devices developed for this purpose.
Collapse
Affiliation(s)
- Sofia Nahavandi
- Faculty of Medicine, Dentistry, & Health Sciences, The University of Melbourne, VIC 3010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Ranju V, Sathiya S, Kalaivani P, Priya RJ, Saravana Babu C. Memantine exerts functional recovery by improving BDNF and GDNF expression in 3-nitropropionic acid intoxicated mice. Neurosci Lett 2014; 586:1-7. [PMID: 25475686 DOI: 10.1016/j.neulet.2014.11.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/18/2014] [Accepted: 11/25/2014] [Indexed: 11/24/2022]
Abstract
Memantine (MN), a NMDA blocker is well known for its protective effect against various neurodegenerative diseases. However, its role in improving motor function and regulation of neurotrophic factors in Huntington's disease (HD) has not been studied yet. In the present study, we have investigated the effect of MN against 3-nitropropionic acid (3NP), induced motor impairment, and alterations in the expression of brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF) in mice brain. Further, its role in mitochondrial function was assessed by measuring succinate dehydrogenase (SDH) activity. Glial fibrillary acidic protein (GFAP) and neuronal nuclei (NeuN) immunoreactivity were studied to evaluate the role of MN on glial and neuronal function. Its effect on apoptosis was adjudged by studying the expression of apoptotic markers. MN restored motor functions with an associated up-regulation in neurotrophin expression. MN also enhanced brain SDH activity and decreased glutamate content. MN ameliorated striatal neuronal loss, reduced GFAP immunoreactivity, and exhibited protective effect against neuronal apoptosis. Data from the current study demonstrated that MN exerted neuroprotective effect against 3NP induced neuropathology. Restoration of motor function by MN might be through regulation of neurotrophin expression. MN can therefore be a useful therapeutic choice in the symptomatic management of HD.
Collapse
Affiliation(s)
- Vijayan Ranju
- Centre for Toxicology and Developmental Research, Sri Ramachandra University, Chennai 600116, India; Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Sekar Sathiya
- Centre for Toxicology and Developmental Research, Sri Ramachandra University, Chennai 600116, India; Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Periyathambi Kalaivani
- Centre for Toxicology and Developmental Research, Sri Ramachandra University, Chennai 600116, India
| | - Raju Jyothi Priya
- Centre for Toxicology and Developmental Research, Sri Ramachandra University, Chennai 600116, India
| | - Chidambaram Saravana Babu
- Centre for Toxicology and Developmental Research, Sri Ramachandra University, Chennai 600116, India.
| |
Collapse
|
38
|
Munsie LN, Milnerwood AJ, Seibler P, Beccano-Kelly DA, Tatarnikov I, Khinda J, Volta M, Kadgien C, Cao LP, Tapia L, Klein C, Farrer MJ. Retromer-dependent neurotransmitter receptor trafficking to synapses is altered by the Parkinson's disease VPS35 mutation p.D620N. Hum Mol Genet 2014; 24:1691-703. [PMID: 25416282 DOI: 10.1093/hmg/ddu582] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Vacuolar protein sorting 35 (VPS35) is a core component of the retromer complex, crucial to endosomal protein sorting and intracellular trafficking. We recently linked a mutation in VPS35 (p.D620N) to familial parkinsonism. Here, we characterize human VPS35 and retromer function in mature murine neuronal cultures and investigate neuron-specific consequences of the p.D620N mutation. We find VPS35 localizes to dendritic spines and is involved in the trafficking of excitatory AMPA-type glutamate receptors (AMPARs). Fundamental neuronal processes, including excitatory synaptic transmission, AMPAR surface expression and synaptic recycling are altered by VPS35 overexpression. VPS35 p.D620N acts as a loss-of-function mutation with respect to VPS35 activity regulating synaptic transmission and AMPAR recycling in mouse cortical neurons and dopamine neuron-like cells produced from induced pluripotent stem cells of human p.D620N carriers. Such perturbations to synaptic function likely produce chronic pathophysiological stress upon neuronal circuits that may contribute to neurodegeneration in this, and other, forms of parkinsonism.
Collapse
Affiliation(s)
- L N Munsie
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - A J Milnerwood
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada, Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada V6T 2B5
| | - P Seibler
- Division of Neurogenetics, Department of Neurology, University of Lübeck, Lübeck, Germany
| | - D A Beccano-Kelly
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - I Tatarnikov
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - J Khinda
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - M Volta
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - C Kadgien
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - L P Cao
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - L Tapia
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| | - C Klein
- Division of Neurogenetics, Department of Neurology, University of Lübeck, Lübeck, Germany
| | - M J Farrer
- Department Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafagian Centre for Brain Health, Vancouver, Canada
| |
Collapse
|
39
|
Beccano-Kelly DA, Kuhlmann N, Tatarnikov I, Volta M, Munsie LN, Chou P, Cao LP, Han H, Tapia L, Farrer MJ, Milnerwood AJ. Synaptic function is modulated by LRRK2 and glutamate release is increased in cortical neurons of G2019S LRRK2 knock-in mice. Front Cell Neurosci 2014; 8:301. [PMID: 25309331 PMCID: PMC4176085 DOI: 10.3389/fncel.2014.00301] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 09/08/2014] [Indexed: 01/04/2023] Open
Abstract
Mutations in Leucine-Rich Repeat Kinase-2 (LRRK2) result in familial Parkinson's disease and the G2019S mutation alone accounts for up to 30% in some ethnicities. Despite this, the function of LRRK2 is largely undetermined although evidence suggests roles in phosphorylation, protein interactions, autophagy and endocytosis. Emerging reports link loss of LRRK2 to altered synaptic transmission, but the effects of the G2019S mutation upon synaptic release in mammalian neurons are unknown. To assess wild type and mutant LRRK2 in established neuronal networks, we conducted immunocytochemical, electrophysiological and biochemical characterization of >3 week old cortical cultures of LRRK2 knock-out, wild-type overexpressing and G2019S knock-in mice. Synaptic release and synapse numbers were grossly normal in LRRK2 knock-out cells, but discretely reduced glutamatergic activity and reduced synaptic protein levels were observed. Conversely, synapse density was modestly but significantly increased in wild-type LRRK2 overexpressing cultures although event frequency was not. In knock-in cultures, glutamate release was markedly elevated, in the absence of any change to synapse density, indicating that physiological levels of G2019S LRRK2 elevate probability of release. Several pre-synaptic regulatory proteins shown by others to interact with LRRK2 were expressed at normal levels in knock-in cultures; however, synapsin 1 phosphorylation was significantly reduced. Thus, perturbations to the pre-synaptic release machinery and elevated synaptic transmission are early neuronal effects of LRRK2 G2019S. Furthermore, the comparison of knock-in and overexpressing cultures suggests that one copy of the G2019S mutation has a more pronounced effect than an ~3-fold increase in LRRK2 protein. Mutant-induced increases in transmission may convey additional stressors to neuronal physiology that may eventually contribute to the pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Dayne A Beccano-Kelly
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada
| | - Naila Kuhlmann
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada ; Graduate Program in Neuroscience, University of British Columbia Vancouver, BC, Canada
| | - Igor Tatarnikov
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada
| | - Mattia Volta
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada
| | - Lise N Munsie
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada
| | - Patrick Chou
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada
| | - Li-Ping Cao
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada
| | - Heather Han
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada
| | - Lucia Tapia
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada
| | - Matthew J Farrer
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada ; Department of Medical Genetics, University of British Columbia Vancouver, BC, Canada
| | - Austen J Milnerwood
- Centre for Applied Neurogenetics, Medical Genetics, University of British Columbia Vancouver, BC, Canada ; Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia Vancouver, BC, Canada ; Division of Neurology, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
40
|
Sepers MD, Raymond LA. Mechanisms of synaptic dysfunction and excitotoxicity in Huntington's disease. Drug Discov Today 2014; 19:990-6. [DOI: 10.1016/j.drudis.2014.02.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 12/19/2022]
|
41
|
Differential Synaptic and Extrasynaptic Glutamate-Receptor Alterations in Striatal Medium-Sized Spiny Neurons of Aged YAC128 Huntington's Disease Mice. PLOS CURRENTS 2014; 6. [PMID: 24894506 PMCID: PMC4032384 DOI: 10.1371/currents.hd.34957c4f8bd7cb1f5ec47381dfc811c3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Huntington’s disease (HD) is a late-onset, slowly progressing neurodegenerative disorder caused by an expansion of glutamine repeats. The YAC128 mouse model has been widely used to study the progression of HD symptoms, but little is known about synaptic alterations in very old animals. The present experiments examined synaptic properties of striatal medium-sized spiny neurons (MSNs) in 16 month-old YAC128 mice. These mice were crossed with mice expressing enhanced green fluorescent protein (EGFP) under the control of either D1 or D2 dopamine receptor promoters to identify MSNs originating the direct and indirect pathways, respectively. The input-output curves of evoked excitatory postsynaptic currents mediated by activation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) or N-methyl-D-aspartate (NMDA) receptors were reduced in MSNs in both pathways. In the presence of DL-threo-β-Benzyloxyaspartic acid (DL-TBOA), a glutamate transporter blocker used to increase activation of extrasynaptic receptors, NMDA receptor-mediated currents displayed altered amplitudes, longer decay times, and greater charge (response areas) in both direct and indirect pathway MSNs in YAC128 mice compared to wildtype controls. Amplitudes were significantly increased, primarily in direct pathway MSNs while normalized areas were significantly increased only in indirect pathway MSNs, suggesting that the two types of MSNs are affected in different ways. It may be that indirect pathway neurons are more susceptible to changes in glutamate transport. Taken together, the present findings demonstrate differential alterations in synaptic versus extrasynaptic NMDA receptors in both direct and indirect pathway MSNs in late HD, which may contribute to the dysfunction and degeneration in both pathways.
Collapse
|
42
|
Coquinco A, Kojic L, Wen W, Wang YT, Jeon NL, Milnerwood AJ, Cynader M. A microfluidic based in vitro model of synaptic competition. Mol Cell Neurosci 2014; 60:43-52. [DOI: 10.1016/j.mcn.2014.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 03/11/2014] [Accepted: 03/15/2014] [Indexed: 10/25/2022] Open
|
43
|
Parsons M, Raymond L. Extrasynaptic NMDA Receptor Involvement in Central Nervous System Disorders. Neuron 2014; 82:279-93. [DOI: 10.1016/j.neuron.2014.03.030] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2014] [Indexed: 12/21/2022]
|
44
|
Neck muscle afferents influence oromotor and cardiorespiratory brainstem neural circuits. Brain Struct Funct 2014; 220:1421-36. [PMID: 24595534 PMCID: PMC4409642 DOI: 10.1007/s00429-014-0734-8] [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: 08/09/2013] [Accepted: 02/11/2014] [Indexed: 12/17/2022]
Abstract
Sensory information arising from the upper neck is important in the reflex control of posture and eye position. It has also been linked to the autonomic control of the cardiovascular and respiratory systems. Whiplash associated disorders (WAD) and cervical dystonia, which involve disturbance to the neck region, can often present with abnormalities to the oromotor, respiratory and cardiovascular systems. We investigated the potential neural pathways underlying such symptoms. Simulating neck afferent activity by electrical stimulation of the second cervical nerve in a working heart brainstem preparation (WHBP) altered the pattern of central respiratory drive and increased perfusion pressure. Tracing central targets of these sensory afferents revealed projections to the intermedius nucleus of the medulla (InM). These anterogradely labelled afferents co-localised with parvalbumin and vesicular glutamate transporter 1 indicating that they are proprioceptive. Anterograde tracing from the InM identified projections to brain regions involved in respiratory, cardiovascular, postural and oro-facial behaviours—the neighbouring hypoglossal nucleus, facial and motor trigeminal nuclei, parabrachial nuclei, rostral and caudal ventrolateral medulla and nucleus ambiguus. In brain slices, electrical stimulation of afferent fibre tracts lateral to the cuneate nucleus monosynaptically excited InM neurones. Direct stimulation of the InM in the WHBP mimicked the response of second cervical nerve stimulation. These results provide evidence of pathways linking upper cervical sensory afferents with CNS areas involved in autonomic and oromotor control, via the InM. Disruption of these neuronal pathways could, therefore, explain the dysphagic and cardiorespiratory abnormalities which may accompany cervical dystonia and WAD.
Collapse
|
45
|
Dau A, Gladding CM, Sepers MD, Raymond LA. Chronic blockade of extrasynaptic NMDA receptors ameliorates synaptic dysfunction and pro-death signaling in Huntington disease transgenic mice. Neurobiol Dis 2014; 62:533-42. [DOI: 10.1016/j.nbd.2013.11.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 10/17/2013] [Accepted: 11/12/2013] [Indexed: 12/16/2022] Open
|
46
|
Parsons MP, Kang R, Buren C, Dau A, Southwell AL, Doty CN, Sanders SS, Hayden MR, Raymond LA. Bidirectional control of postsynaptic density-95 (PSD-95) clustering by Huntingtin. J Biol Chem 2013; 289:3518-28. [PMID: 24347167 DOI: 10.1074/jbc.m113.513945] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Huntington disease is associated with early alterations in corticostriatal synaptic function that precede cell death, and it is postulated that ameliorating such changes may delay clinical onset and/or prevent neurodegeneration. Although many of these synaptic alterations are thought to be attributable to a toxic gain of function of the mutant huntingtin protein, the role that nonpathogenic huntingtin (HTT) plays in synaptic function is relatively unexplored. Here, we compare the immunocytochemical localization of a major postsynaptic scaffolding protein, PSD-95, in striatal neurons from WT mice and mice overexpressing HTT with 18 glutamine repeats (YAC18, nonpathogenic). We found that HTT overexpression resulted in a palmitoylation- and BDNF-dependent increase in PSD-95 clustering at synaptic sites in striatal spiny projection neurons (SPNs) co-cultured with cortical neurons. Surprisingly, the latter effect was mediated presynaptically, as HTT overexpression in cortical neurons alone was sufficient to increase PSD-95 clustering in the postsynaptic SPNs. In contrast, antisense oligonucleotide knockdown of HTT in WT co-cultures resulted in a significant reduction of PSD-95 clustering in SPNs. Notably, despite these bidirectional changes in PSD-95 clustering, we did not observe an alteration in basal electrophysiological measures of AMPA and NMDA receptors. Thus, unlike in previous studies in the hippocampus, enhanced or decreased PSD-95 clustering alone was insufficient to drive AMPA or NMDA receptors into or out of SPN synapses. In all, our results demonstrate that nonpathogenic HTT can indeed influence synaptic protein localization and uncover a novel role of HTT in PSD-95 distribution.
Collapse
Affiliation(s)
- Matthew P Parsons
- From the Department of Psychiatry and Brain Research Centre, University of British Columbia, Vancouver, British Columbia V6T 1Z3 and
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Laird MD, Shields JS, Sukumari-Ramesh S, Kimbler DE, Fessler RD, Shakir B, Youssef P, Yanasak N, Vender JR, Dhandapani KM. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia 2013; 62:26-38. [PMID: 24166800 DOI: 10.1002/glia.22581] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 09/06/2013] [Accepted: 09/09/2013] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Cerebral edema, a life-threatening medical complication, contributes to elevated intracranial pressure (ICP) and a poor clinical prognosis after TBI. Unfortunately, treatment options to reduce post-traumatic edema remain suboptimal, due in part, to a dearth of viable therapeutic targets. Herein, we tested the hypothesis that cerebral innate immune responses contribute to edema development after TBI. Our results demonstrate that high-mobility group box protein 1 (HMGB1) was released from necrotic neurons via a NR2B-mediated mechanism. HMGB1 was clinically associated with elevated ICP in patients and functionally promoted cerebral edema after TBI in mice. The detrimental effects of HMGB1 were mediated, at least in part, via activation of microglial toll-like receptor 4 (TLR4) and the subsequent expression of the astrocytic water channel, aquaporin-4 (AQP4). Genetic or pharmacological (VGX-1027) TLR4 inhibition attenuated the neuroinflammatory response and limited post-traumatic edema with a delayed, clinically implementable therapeutic window. Human and rodent tissue culture studies further defined the cellular mechanisms demonstrating neuronal HMGB1 initiates the microglial release of interleukin-6 (IL-6) in a TLR4 dependent mechanism. In turn, microglial IL-6 increased the astrocytic expression of AQP4. Taken together, these data implicate microglia as key mediators of post-traumatic brain edema and suggest HMGB1-TLR4 signaling promotes neurovascular dysfunction after TBI.
Collapse
Affiliation(s)
- Melissa D Laird
- Department of Neurosurgery, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gillingwater TH, Wishart TM. Mechanisms underlying synaptic vulnerability and degeneration in neurodegenerative disease. Neuropathol Appl Neurobiol 2013; 39:320-34. [PMID: 23289367 DOI: 10.1111/nan.12014] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 12/21/2012] [Indexed: 02/06/2023]
Abstract
Recent developments in our understanding of events underlying neurodegeneration across the central and peripheral nervous systems have highlighted the critical role that synapses play in the initiation and progression of neuronal loss. With the development of increasingly accurate and versatile animal models of neurodegenerative disease it has become apparent that disruption of synaptic form and function occurs comparatively early, preceding the onset of degenerative changes in the neuronal cell body. Yet, despite our increasing awareness of the importance of synapses in neurodegeneration, the mechanisms governing the particular susceptibility of distal neuronal processes are only now becoming clear. In this review we bring together recent developments in our understanding of cellular and molecular mechanisms regulating synaptic vulnerability. We have placed a particular focus on three major areas of research that have gained significant interest over the last few years: (i) the contribution of synaptic mitochondria to neurodegeneration; (ii) the contribution of pathways that modulate synaptic function; and (iii) regulation of synaptic degeneration by local posttranslational modifications such as ubiquitination. We suggest that targeting these organelles and pathways may be a productive way to develop synaptoprotective strategies applicable to a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- T H Gillingwater
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
49
|
Baucum AJ, Brown AM, Colbran RJ. Differential association of postsynaptic signaling protein complexes in striatum and hippocampus. J Neurochem 2013; 124:490-501. [PMID: 23173822 PMCID: PMC3557548 DOI: 10.1111/jnc.12101] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 11/19/2012] [Accepted: 11/20/2012] [Indexed: 02/03/2023]
Abstract
Distinct physiological stimuli are required for bidirectional synaptic plasticity in striatum and hippocampus, but differences in the underlying signaling mechanisms are poorly understood. We have begun to compare levels and interactions of key excitatory synaptic proteins in whole extracts and subcellular fractions isolated from micro-dissected striatum and hippocampus. Levels of multiple glutamate receptor subunits, calcium/calmodulin-dependent protein kinase II (CaMKII), a highly abundant serine/threonine kinase, and spinophilin, a F-actin and protein phosphatase 1 (PP1) binding protein, were significantly lower in striatal extracts, as well as in synaptic and/or extrasynaptic fractions, compared with similar hippocampal extracts/fractions. However, CaMKII interactions with spinophilin were more robust in striatum compared with hippocampus, and this enhanced association was restricted to the extrasynaptic fraction. NMDAR GluN2B subunits associate with both spinophilin and CaMKII, but spinophilin-GluN2B complexes were enriched in extrasynaptic fractions whereas CaMKII-GluN2B complexes were enriched in synaptic fractions. Notably, the association of GluN2B with both CaMKII and spinophilin was more robust in striatal extrasynaptic fractions compared with hippocampal extrasynaptic fractions. Selective differences in the assembly of synaptic and extrasynaptic signaling complexes may contribute to differential physiological regulation of excitatory transmission in striatum and hippocampus.
Collapse
Affiliation(s)
- Anthony J Baucum
- Department of Molecular Physiology and Biophysics, Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| | | | | |
Collapse
|
50
|
Mrzljak L, Munoz-Sanjuan I. Therapeutic Strategies for Huntington's Disease. Curr Top Behav Neurosci 2013; 22:161-201. [PMID: 24277342 DOI: 10.1007/7854_2013_250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Huntington's disease (HD) is a devastating autosomal dominant neurodegenerative disease, caused by expansion of the CAG repeat in the huntingtin (HTT) gene and characterized pathologically by the loss of pyramidal neurons in several cortical areas, of striatal medium spiny neurons, and of hypothalamic neurons. Clinically, a distinguishing feature of the disease is uncontrolled involuntary movements (chorea, dyskensias) accompanied by progressive cognitive, motor, and psychiatric impairment. This review focuses on the current state of therapeutic development for the treatment of HD, including the preclinical and clinical development of small molecules and molecular therapies.
Collapse
|