1
|
Ruggiero RN, Marques DB, Rossignoli MT, De Ross JB, Prizon T, Beraldo IJS, Bueno-Junior LS, Kandratavicius L, Peixoto-Santos JE, Lopes-Aguiar C, Leite JP. Dysfunctional hippocampal-prefrontal network underlies a multidimensional neuropsychiatric phenotype following early-life seizure. eLife 2024; 12:RP90997. [PMID: 38593008 PMCID: PMC11003745 DOI: 10.7554/elife.90997] [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] [Indexed: 04/11/2024] Open
Abstract
Brain disturbances during development can have a lasting impact on neural function and behavior. Seizures during this critical period are linked to significant long-term consequences such as neurodevelopmental disorders, cognitive impairments, and psychiatric symptoms, resulting in a complex spectrum of multimorbidity. The hippocampus-prefrontal cortex (HPC-PFC) circuit emerges as a potential common link between such disorders. However, the mechanisms underlying these outcomes and how they relate to specific behavioral alterations are unclear. We hypothesized that specific dysfunctions of hippocampal-cortical communication due to early-life seizure would be associated with distinct behavioral alterations observed in adulthood. Here, we performed a multilevel study to investigate behavioral, electrophysiological, histopathological, and neurochemical long-term consequences of early-life Status epilepticus in male rats. We show that adult animals submitted to early-life seizure (ELS) present working memory impairments and sensorimotor disturbances, such as hyperlocomotion, poor sensorimotor gating, and sensitivity to psychostimulants despite not exhibiting neuronal loss. Surprisingly, cognitive deficits were linked to an aberrant increase in the HPC-PFC long-term potentiation (LTP) in a U-shaped manner, while sensorimotor alterations were associated with heightened neuroinflammation, as verified by glial fibrillary acidic protein (GFAP) expression, and altered dopamine neurotransmission. Furthermore, ELS rats displayed impaired HPC-PFC theta-gamma coordination and an abnormal brain state during active behavior resembling rapid eye movement (REM) sleep oscillatory dynamics. Our results point to impaired HPC-PFC functional connectivity as a possible pathophysiological mechanism by which ELS can cause cognitive deficits and psychiatric-like manifestations even without neuronal loss, bearing translational implications for understanding the spectrum of multidimensional developmental disorders linked to early-life seizures.
Collapse
Affiliation(s)
- Rafael Naime Ruggiero
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São PauloRibeirão PretoBrazil
| | - Danilo Benette Marques
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São PauloRibeirão PretoBrazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São PauloRibeirão PretoBrazil
| | - Jana Batista De Ross
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São PauloRibeirão PretoBrazil
| | - Tamiris Prizon
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São PauloRibeirão PretoBrazil
| | - Ikaro Jesus Silva Beraldo
- Department of Physiology and Biophysics Federal University of Minas GeraisBelo HorizonteBrazil
- Laboratory of Molecular and Behavioral Neuroscience (LANEC), Federal University of Minas GeraisBelo HorizonteBrazil
| | | | | | - Jose Eduardo Peixoto-Santos
- Neuroscience Discipline, Department of Neurology and Neurosurgery,Universidade Federal de São PauloSão PauloBrazil
| | - Cleiton Lopes-Aguiar
- Department of Physiology and Biophysics Federal University of Minas GeraisBelo HorizonteBrazil
- Laboratory of Molecular and Behavioral Neuroscience (LANEC), Federal University of Minas GeraisBelo HorizonteBrazil
| | - Joao Pereira Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São PauloRibeirão PretoBrazil
| |
Collapse
|
2
|
Pan K, Jinnah HA, Hess EJ, Smith Y, Villalba RM. Ultrastructural analysis of nigrostriatal dopaminergic terminals in a knockin mouse model of DYT1 dystonia. Eur J Neurosci 2024; 59:1407-1427. [PMID: 38123503 DOI: 10.1111/ejn.16197] [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: 03/08/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 12/23/2023]
Abstract
DYT1 dystonia is associated with decreased striatal dopamine release. In this study, we examined the possibility that ultrastructural changes of nigrostriatal dopamine terminals could contribute to this neurochemical imbalance using a serial block face/scanning electron microscope (SBF/SEM) and three-dimensional reconstruction to analyse striatal tyrosine hydroxylase-immunoreactive (TH-IR) terminals and their synapses in a DYT1(ΔE) knockin (DYT1-KI) mouse model of DYT1 dystonia. Furthermore, to study possible changes in vesicle packaging capacity of dopamine, we used transmission electron microscopy to assess the synaptic vesicle size in striatal dopamine terminals. Quantitative comparative analysis of 80 fully reconstructed TH-IR terminals in the WT and DYT1-KI mice indicate (1) no significant difference in the volume of TH-IR terminals; (2) no major change in the proportion of axo-spinous versus axo-dendritic synapses; (3) no significant change in the post-synaptic density (PSD) area of axo-dendritic synapses, while the PSDs of axo-spinous synapses were significantly smaller in DYT1-KI mice; (4) no significant change in the contact area between TH-IR terminals and dendritic shafts or spines, while the ratio of PSD area/contact area decreased significantly for both axo-dendritic and axo-spinous synapses in DYT1-KI mice; (5) no significant difference in the mitochondria volume; and (6) no significant difference in the synaptic vesicle area between the two groups. Altogether, these findings suggest that abnormal morphometric changes of nigrostriatal dopamine terminals and their post-synaptic targets are unlikely to be a major source of reduced striatal dopamine release in DYT1 dystonia.
Collapse
Affiliation(s)
- Ke Pan
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Physical Therapy & Human Movement Sciences, Northwestern University, Chicago, Illinois, USA
| | - Hyder A Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Human Genetics and Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Ellen J Hess
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, Georgia, USA
| | - Yoland Smith
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Rosa M Villalba
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Lyu S, Xing H, Liu Y, Girdhar P, Yokoi F, Li Y. Further Studies on the Role of BTBD9 in the Cerebellum, Sleep-like Behaviors and the Restless Legs Syndrome. Neuroscience 2022; 505:78-90. [PMID: 36244636 PMCID: PMC10367443 DOI: 10.1016/j.neuroscience.2022.10.008] [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: 08/05/2022] [Revised: 09/15/2022] [Accepted: 10/09/2022] [Indexed: 11/26/2022]
Abstract
Genetic analyses have linked BTBD9 to restless legs syndrome (RLS) and sleep regulation. Btbd9 knockout mice show RLS-like motor restlessness. Previously, we found hyperactivity of cerebellar Purkinje cells (PCs) in Btbd9 knockout mice, which may contribute to the motor restlessness observed. However, underlying mechanisms for PC hyperactivity in Btbd9 knockout mice are unknown. Here, we used dissociated PC recording, brain slice recording and western blot to address this question. Our dissociated recording shows that knockout PCs had increased TEA-sensitive, Ca2+-dependent K+ currents. Applying antagonist to large conductance Ca2+-activated K+ (BK) channels further isolated the increased current as BK current. Consistently, we found increased amplitude of afterhyperpolarization and elevated BK protein levels in the knockout mice. Dissociated recording also shows a decrease in TEA-insensitive, Ca2+-dependent K+ currents. The result is consistent with reduced amplitude of tail currents, mainly composed of small conductance Ca2+-activated K+ (SK) currents, in slice recording. Our results suggest that BK and SK channels may be responsible for the hyperactivity of knockout PCs. Recently, BTBD9 protein was shown to associate with SYNGAP1 protein. We found a decreased cerebellar level of SYNGAP1 in Btbd9 knockout mice. However, Syngap1 heterozygous knockout mice showed nocturnal, instead of diurnal, motor restlessness. Our results suggest that SYNGAP1 deficiency may not contribute directly to the RLS-like motor restlessness observed in Btbd9 knockout mice. Finally, we found that PC-specific Btbd9 knockout mice exhibited deficits in motor coordination and balance similar to Btbd9 knockout mice, suggesting that the motor effect of BTBD9 in PCs is cell-autonomous.
Collapse
Affiliation(s)
- Shangru Lyu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuning Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pallavi Girdhar
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
4
|
Xing H, Yokoi F, Walker AL, Torres-Medina R, Liu Y, Li Y. Electrophysiological characterization of the striatal cholinergic interneurons in Dyt1 ΔGAG knock-in mice. DYSTONIA 2022; 1:10557. [PMID: 36329866 PMCID: PMC9629210 DOI: 10.3389/dyst.2022.10557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
DYT1 dystonia is an inherited early-onset movement disorder characterized by sustained muscle contractions causing twisting, repetitive movements, and abnormal postures. Most DYT1 patients have a heterozygous trinucleotide GAG deletion mutation (ΔGAG) in DYT1/TOR1A, coding for torsinA. Dyt1 heterozygous ΔGAG knock-in (KI) mice show motor deficits and reduced striatal dopamine receptor 2 (D2R). Striatal cholinergic interneurons (ChIs) are essential in regulating striatal motor circuits. Multiple dystonia rodent models, including KI mice, show altered ChI firing and modulation. However, due to the errors in assigning KI mice, it is essential to replicate these findings in genetically confirmed KI mice. Here, we found irregular and decreased spontaneous firing frequency in the acute brain slices from Dyt1 KI mice. Quinpirole, a D2R agonist, showed less inhibitory effect on the spontaneous ChI firing in Dyt1 KI mice, suggesting decreased D2R function on the striatal ChIs. On the other hand, a muscarinic receptor agonist, muscarine, inhibited the ChI firing in both wild-type (WT) and Dyt1 KI mice. Trihexyphenidyl, a muscarinic acetylcholine receptor M1 antagonist, had no significant effect on the firing. Moreover, the resting membrane property and functions of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, μ-opioid receptors, and large-conductance calcium-activated potassium (BK) channels were unaffected in Dyt1 KI mice. The results suggest that the irregular and low-frequency firing and decreased D2R function are the main alterations of striatal ChIs in Dyt1 KI mice. These results appear consistent with the reduced dopamine release and high striatal acetylcholine tone in the previous reports.
Collapse
Affiliation(s)
- Hong Xing
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Fumiaki Yokoi
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Ariel Luz Walker
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Rosemarie Torres-Medina
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Yuning Liu
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Yuqing Li
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| |
Collapse
|
5
|
Yokoi F, Chen HX, Oleas J, Dang MT, Xing H, Dexter KM, Li Y. Characterization of the direct pathway in Dyt1 ΔGAG heterozygous knock-in mice and dopamine receptor 1-expressing-cell-specific Dyt1 conditional knockout mice. Behav Brain Res 2021; 411:113381. [PMID: 34038798 PMCID: PMC8323984 DOI: 10.1016/j.bbr.2021.113381] [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: 07/17/2020] [Revised: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 10/21/2022]
Abstract
DYT1 dystonia is a movement disorder mainly caused by a trinucleotide deletion (ΔGAG) in DYT1 (TOR1A), coding for torsinA. DYT1 dystonia patients show trends of decreased striatal ligand-binding activities to dopamine receptors 1 (D1R) and 2 (D2R). Dyt1 ΔGAG knock-in (KI) mice, which have the corresponding ΔGAG deletion, similarly exhibit reduced striatal D1R and D2R-binding activities and their expression levels. While the consequences of D2R reduction have been well characterized, relatively little is known about the effect of D1R reduction. Here, locomotor responses to D1R and D2R antagonists were examined in Dyt1 KI mice. Dyt1 KI mice showed significantly less responsiveness to both D1R antagonist SCH 23390 and D2R antagonist raclopride. The electrophysiological recording indicated that Dyt1 KI mice showed a significantly increased paired-pulse ratio of the striatal D1R-expressing medium spiny neurons and altered miniature excitatory postsynaptic currents. To analyze the in vivo torsinA function in the D1R-expressing neurons further, Dyt1 conditional knockout (Dyt1 d1KO) mice in these neurons were generated. Dyt1 d1KO mice had decreased spontaneous locomotor activity and reduced numbers of slips in the beam-walking test. Dyt1 d1KO male mice showed abnormal gait. Dyt1 d1KO mice showed defective striatal D1R maturation. Moreover, the mutant striatal D1R-expressing medium spiny neurons had increased capacitance, decreased sEPSC frequency, and reduced intrinsic excitability. The results suggest that torsinA in the D1R-expressing cells plays an important role in the electrophysiological function and motor performance. Medical interventions to the direct pathway may affect the onset and symptoms of this disorder.
Collapse
Affiliation(s)
- Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610-0236, USA.
| | - Huan-Xin Chen
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610-0236, USA
| | - Janneth Oleas
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610-0236, USA
| | - Mai Tu Dang
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610-0236, USA
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610-0236, USA
| | - Kelly M Dexter
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610-0236, USA
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610-0236, USA.
| |
Collapse
|
6
|
McClelland VM, Lin JP. Sensorimotor Integration in Childhood Dystonia and Dystonic Cerebral Palsy-A Developmental Perspective. Front Neurol 2021; 12:668081. [PMID: 34367047 PMCID: PMC8343097 DOI: 10.3389/fneur.2021.668081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/07/2021] [Indexed: 11/15/2022] Open
Abstract
Dystonia is a disorder of sensorimotor integration, involving dysfunction within the basal ganglia, cortex, cerebellum, or their inter-connections as part of the sensorimotor network. Some forms of dystonia are also characterized by maladaptive or exaggerated plasticity. Development of the neuronal processes underlying sensorimotor integration is incompletely understood but involves activity-dependent modeling and refining of sensorimotor circuits through processes that are already taking place in utero and which continue through infancy, childhood, and into adolescence. Several genetic dystonias have clinical onset in early childhood, but there is evidence that sensorimotor circuit development may already be disrupted prenatally in these conditions. Dystonic cerebral palsy (DCP) is a form of acquired dystonia with perinatal onset during a period of rapid neurodevelopment and activity-dependent refinement of sensorimotor networks. However, physiological studies of children with dystonia are sparse. This discussion paper addresses the role of neuroplasticity in the development of sensorimotor integration with particular focus on the relevance of these mechanisms for understanding childhood dystonia, DCP, and implications for therapy selection, including neuromodulation and timing of intervention.
Collapse
Affiliation(s)
- Verity M McClelland
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Children's Neurosciences Department, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Jean-Pierre Lin
- Children's Neurosciences Department, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
7
|
Yokoi F, Dang MT, Zhang L, Dexter KM, Efimenko I, Krishnaswamy S, Villanueva M, Misztal CI, Gerard M, Lynch P, Li Y. Reversal of motor-skill transfer impairment by trihexyphenidyl and reduction of dorsolateral striatal cholinergic interneurons in Dyt1 ΔGAG knock-in mice. IBRO Neurosci Rep 2021; 11:1-7. [PMID: 34189496 PMCID: PMC8215213 DOI: 10.1016/j.ibneur.2021.05.003] [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: 01/27/2021] [Revised: 03/06/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
DYT-TOR1A or DYT1 early-onset generalized dystonia is an inherited movement disorder characterized by sustained muscle contractions causing twisting, repetitive movements, or abnormal postures. The majority of the DYT1 dystonia patients have a trinucleotide GAG deletion in DYT1/TOR1A. Trihexyphenidyl (THP), an antagonist for excitatory muscarinic acetylcholine receptor M1, is commonly used to treat dystonia. Dyt1 heterozygous ΔGAG knock-in (KI) mice, which have the corresponding mutation, exhibit impaired motor-skill transfer. Here, the effect of THP injection during the treadmill training period on the motor-skill transfer to the accelerated rotarod performance was examined. THP treatment reversed the motor-skill transfer impairment in Dyt1 KI mice. Immunohistochemistry showed that Dyt1 KI mice had a significant reduction of the dorsolateral striatal cholinergic interneurons. In contrast, Western blot analysis showed no significant alteration in the expression levels of the striatal enzymes and transporters involved in the acetylcholine metabolism. The results suggest a functional alteration of the cholinergic system underlying the impairment of motor-skill transfer and the pathogenesis of DYT1 dystonia. Training with THP in a motor task may improve another motor skill performance in DYT1 dystonia.
Collapse
Key Words
- ACh, acetylcholine
- AChE, acetylcholinesterase
- BSA, bovine serum albumin
- CI, confidence interval
- ChAT, choline acetyltransferase
- ChI, cholinergic interneuron
- ChT, choline transporter
- Cholinergic interneuron
- DAB, 3,3′-diaminobenzidine
- DF, degrees of freedom
- Dystonia
- Dyt1 KI mice, Dyt1 ΔGAG heterozygous knock-in mice
- GAPDH, Glyceraldehyde-3-phosphate dehydrogenase
- KO, knockout
- LTD, long-term depression
- Motor learning
- PB, phosphate buffer
- PBS, phosphate-buffered saline
- PET, positron emission tomography
- Rotarod
- THP, trihexyphenidyl
- TOR1A
- TorsinA
- TrkA, tropomyosin receptor kinase A
- VAChT, vesicular acetylcholine transporter
- WT, wild-type
- n.s., not significant
Collapse
Affiliation(s)
- Fumiaki Yokoi
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Mai Tu Dang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA.,Department of Biomedical Sciences, Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Kelly M Dexter
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Iakov Efimenko
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Shiv Krishnaswamy
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Matthew Villanueva
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Carly I Misztal
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Malinda Gerard
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Patrick Lynch
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Yuqing Li
- Norman Fixel Institute of Neurological Diseases, McKnight Brain Institute, and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| |
Collapse
|
8
|
Sciamanna G, Ponterio G, Vanni V, Laricchiuta D, Martella G, Bonsi P, Meringolo M, Tassone A, Mercuri NB, Pisani A. Optogenetic Activation of Striatopallidal Neurons Reveals Altered HCN Gating in DYT1 Dystonia. Cell Rep 2021; 31:107644. [PMID: 32433955 DOI: 10.1016/j.celrep.2020.107644] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 12/10/2019] [Accepted: 04/20/2020] [Indexed: 12/30/2022] Open
Abstract
Firing activity of external globus pallidus (GPe) is crucial for motor control and is severely perturbed in dystonia, a movement disorder characterized by involuntary, repetitive muscle contractions. Here, we show that GPe projection neurons exhibit a reduction of firing frequency and an irregular pattern in a DYT1 dystonia model. Optogenetic activation of the striatopallidal pathway fails to reset pacemaking activity of GPe neurons in mutant mice. Abnormal firing is paralleled by alterations in motor learning. We find that loss of dopamine D2 receptor-dependent inhibition causes increased GABA input at striatopallidal synapses, with subsequent downregulation of hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channels. Accordingly, enhancing in vivo HCN channel activity or blocking GABA release restores both the ability of striatopallidal inputs to pause ongoing GPe activity and motor coordination deficits. Our findings demonstrate an impaired striatopallidal connectivity, supporting the central role of GPe in motor control and, more importantly, identifying potential pharmacological targets for dystonia.
Collapse
Affiliation(s)
- Giuseppe Sciamanna
- Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy; Lab of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giulia Ponterio
- Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy
| | - Valentina Vanni
- Lab of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Daniela Laricchiuta
- Department of Psychology, Faculty of Medicine and Psychology, University of Rome Sapienza, Rome, Italy; Lab of Behavioural and Experimental Neurophysiology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Lab of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Bonsi
- Lab of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Meringolo
- Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy
| | - Annalisa Tassone
- Lab of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy; Lab of Experimental Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Pisani
- Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy; Lab of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
9
|
Alteration of the cholinergic system and motor deficits in cholinergic neuron-specific Dyt1 knockout mice. Neurobiol Dis 2021; 154:105342. [PMID: 33757902 DOI: 10.1016/j.nbd.2021.105342] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Dystonia is a neurological movement disorder characterized by sustained or intermittent muscle contractions, repetitive movement, and sometimes abnormal postures. DYT1 dystonia is one of the most common genetic dystonias, and most patients carry heterozygous DYT1 ∆GAG mutations causing a loss of a glutamic acid of the protein torsinA. Patients can be treated with anticholinergics, such as trihexyphenidyl, suggesting an abnormal cholinergic state. Early work on the cell-autonomous effects of Dyt1 deletion with ChI-specific Dyt1 conditional knockout mice (Dyt1 Ch1KO) revealed abnormal electrophysiological responses of striatal ChIs to muscarine and quinpirole, motor deficits, and no changes in the number or size of the ChIs. However, the Chat-cre line that was used to derive Dyt1 Ch1KO mice contained a neomycin cassette and was reported to have ectopic cre-mediated recombination. In this study, we generated a Dyt1 Ch2KO mouse line by removing the neomycin cassette in Dyt1 Ch1KO mice. The Dyt1 Ch2KO mice showed abnormal paw clenching behavior, motor coordination and balance deficits, impaired motor learning, reduced striatal choline acetyltransferase protein level, and a reduced number of striatal ChIs. Furthermore, the mutant striatal ChIs had a normal muscarinic inhibitory function, impaired quinpirole-mediated inhibition, and altered current density. Our findings demonstrate a cell-autonomous effect of Dyt1 deletion on the striatal ChIs and a critical role for the striatal ChIs and corticostriatal pathway in the pathogenesis of DYT1 dystonia.
Collapse
|
10
|
Lo YC, Chen KY, Chou HC, Lin IH, Chen CM. Neonatal hyperoxia induces gut dysbiosis and behavioral changes in adolescent mice. J Chin Med Assoc 2021; 84:290-298. [PMID: 33496514 DOI: 10.1097/jcma.0000000000000488] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Supplemental oxygen is often required to treat preterm infants with respiratory disorders. Experimental studies have demonstrated that hyperoxia results in the disruption of intestinal and neuronal plasticity and myelination of the brain. The association between the neonatal hyperoxia and changes of phenotypes in gut microbiota and in behaviors is not clear to date. METHODS We designed an animal experiment that C57BL/6 mice pups were reared in either room air (RA) or hyperoxia (85% O2) from postnatal days 1 to 7. From postnatal days 8 to 42, the mice were reared in RA. Intestinal microbiota was sampled from the lower gastrointestinal tract on postnatal days 7 and 42, and behavioral tests were performed and brain tissues were collected on postnatal day 42. RESULTS Neonatal hyperoxia decreased intestinal tight junction protein expression and altered intestinal bacterial composition and diversity on postnatal day 7. Among the concrete discriminative features, Proteobacteria and Epsilonbacteraeota were significantly elevated in hyperoxia-reared mice on postnatal days 7 and 42, respectively. Hyperoxia-reared mice exhibited significantly reduced sociability and interest in social novelty and impaired motor coordination compared with RA-reared mice on postnatal day 42. Hyperoxia-reared mice also exhibited significantly reduced myelination and a significantly higher number of apoptotic cells in the brain compared with RA-reared mice on postnatal day 42. CONCLUSION Neonatal hyperoxia during the first week of life altered gut microbiota and reduced brain myelination that might associate with the deficits of social interaction and motor coordination in adolescent mice.
Collapse
Affiliation(s)
- Yu-Chun Lo
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Kai-Yun Chen
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - I-Hsuan Lin
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan, ROC
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| |
Collapse
|
11
|
Yokoi F, Oleas J, Xing H, Liu Y, Dexter KM, Misztal C, Gerard M, Efimenko I, Lynch P, Villanueva M, Alsina R, Krishnaswamy S, Vaillancourt DE, Li Y. Decreased number of striatal cholinergic interneurons and motor deficits in dopamine receptor 2-expressing-cell-specific Dyt1 conditional knockout mice. Neurobiol Dis 2020; 134:104638. [PMID: 31618684 PMCID: PMC7323754 DOI: 10.1016/j.nbd.2019.104638] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 10/07/2019] [Accepted: 10/11/2019] [Indexed: 12/28/2022] Open
Abstract
DYT1 early-onset generalized torsion dystonia is a hereditary movement disorder characterized by abnormal postures and repeated movements. It is caused mainly by a heterozygous trinucleotide deletion in DYT1/TOR1A, coding for torsinA. The mutation may lead to a partial loss of torsinA function. Functional alterations of the basal ganglia circuits have been implicated in this disease. Striatal dopamine receptor 2 (D2R) levels are significantly decreased in DYT1 dystonia patients and in the animal models of DYT1 dystonia. D2R-expressing cells, such as the medium spiny neurons in the indirect pathway, striatal cholinergic interneurons, and dopaminergic neurons in the basal ganglia circuits, contribute to motor performance. However, the function of torsinA in these neurons and its contribution to the motor symptoms is not clear. Here, D2R-expressing-cell-specific Dyt1 conditional knockout (d2KO) mice were generated and in vivo effects of torsinA loss in the corresponding cells were examined. The Dyt1 d2KO mice showed significant reductions of striatal torsinA, acetylcholine metabolic enzymes, Tropomyosin receptor kinase A (TrkA), and cholinergic interneurons. The Dyt1 d2KO mice also showed significant reductions of striatal D2R dimers and tyrosine hydroxylase without significant alteration in striatal monoamine contents or the number of dopaminergic neurons in the substantia nigra. The Dyt1 d2KO male mice showed motor deficits in the accelerated rotarod and beam-walking tests without overt dystonic symptoms. Moreover, the Dyt1 d2KO male mice showed significant correlations between striatal monoamines and locomotion. The results suggest that torsinA in the D2R-expressing cells play a critical role in the development or survival of the striatal cholinergic interneurons, expression of striatal D2R mature form, and motor performance. Medical interventions to compensate for the loss of torsinA function in these neurons may affect the onset and symptoms of this disease.
Collapse
Affiliation(s)
- Fumiaki Yokoi
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States.
| | - Janneth Oleas
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Hong Xing
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Yuning Liu
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Kelly M Dexter
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Carly Misztal
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Melinda Gerard
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Iakov Efimenko
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Patrick Lynch
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Matthew Villanueva
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Raul Alsina
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - Shiv Krishnaswamy
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States
| | - David E Vaillancourt
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611-8205, United States; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-8205, United States; Department of Neurology and Center for Movement Disorders and Neurorestoration, College of Medicine, University of Florida, Gainesville, FL 32611-8205, United States
| | - Yuqing Li
- Norman Fixel Institue for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, United States.
| |
Collapse
|
12
|
Yokoi F, Jiang F, Dexter K, Salvato B, Li Y. Improved survival and overt "dystonic" symptoms in a torsinA hypofunction mouse model. Behav Brain Res 2019; 381:112451. [PMID: 31891745 DOI: 10.1016/j.bbr.2019.112451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022]
Abstract
DYT1 dystonia is an inherited movement disorder without obvious neurodegeneration. Multiple mutant mouse models exhibit motor deficits without overt "dystonic" symptoms and neurodegeneration. However, some mouse models do. Among the later models, the N-CKO mouse model, which has a heterozygous Tor1a/Dyt1 knockout (KO) in one allele and Nestin-cre-mediated conditional KO in the other, exhibits a severe lack of weight gain, neurodegeneration, overt "dystonic" symptoms, such as overt leg extension, weak walking, twisted hindpaw and stiff hindlimb, and complete infantile lethality. However, it is not clear if the overt dystonic symptoms were caused by the neurodegeneration in the dying N-CKO mice. Here, the effects of improved maternal care and nutrition during early life on the symptoms in N-CKO mice were analyzed by culling the litter and providing wet food to examine whether the overt dystonic symptoms and severe lack of weight gain are caused by malnutrition-related neurodegeneration. Although the N-CKO mice in this study replicated the severe lack of weight gain and overt "dystonic" symptoms during the lactation period regardless of culling at postnatal day zero or later, there was no significant difference in the brain astrocytes and apoptosis between the N-CKO and control mice. Moreover, more than half of the N-CKO mice with culling survived past the lactation period. The surviving adult N-CKO mice did not display overt "dystonic" symptoms, and in addition they still exhibited small body weight. The results suggest that the overt "dystonic" symptoms in the N-CKO mice were independent of prominent neurodegeneration, which negates the role of neurodegeneration in the pathogenesis of DYT1 dystonia.
Collapse
Affiliation(s)
- Fumiaki Yokoi
- Department of Neurology and Norman Fixel Institute of Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fangfang Jiang
- Department of Neurology and Norman Fixel Institute of Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL, USA; Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu, PR China
| | - Kelly Dexter
- Department of Neurology and Norman Fixel Institute of Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Bryan Salvato
- Department of Neurology and Norman Fixel Institute of Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuqing Li
- Department of Neurology and Norman Fixel Institute of Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
13
|
Perturbed Ca2+-dependent signaling of DYT2 hippocalcin mutant as mechanism of autosomal recessive dystonia. Neurobiol Dis 2019; 132:104529. [DOI: 10.1016/j.nbd.2019.104529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/22/2019] [Accepted: 07/09/2019] [Indexed: 11/23/2022] Open
|
14
|
Moehle MS, Conn PJ. Roles of the M 4 acetylcholine receptor in the basal ganglia and the treatment of movement disorders. Mov Disord 2019; 34:1089-1099. [PMID: 31211471 DOI: 10.1002/mds.27740] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 05/10/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022] Open
Abstract
Acetylcholine (ACh) released from cholinergic interneurons acting through nicotinic and muscarinic acetylcholine receptors (mAChRs) in the striatum have been thought to be central for the potent cholinergic regulation of basal ganglia activity and motor behaviors. ACh activation of mAChRs has multiple actions to oppose dopamine (DA) release, signaling, and related motor behaviors and has led to the idea that a delicate balance of DA and mAChR signaling in the striatum is critical for maintaining normal motor function. Consistent with this, mAChR antagonists have efficacy in reducing motor symptoms in diseases where DA release or signaling is diminished, such as in Parkinson's disease and dystonia, but are limited in their utility because of severe adverse effects. Recent breakthroughs in understanding both the anatomical sites of action of ACh and the mAChR subtypes involved in regulating basal ganglia function reveal that the M4 subtype plays a central role in regulating DA signaling and release in the basal ganglia. These findings have raised the possibility that sources of ACh outside of the striatum can regulate motor activity and that M4 activity is a potent regulator of motor dysfunction. We discuss how M4 activity regulates DA release and signaling, the potential sources of ACh that can regulate M4 activity, and the implications of targeting M4 activity for the treatment of the motor symptoms in movement disorders. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Mark S Moehle
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Campbell SL, van Groen T, Kadish I, Smoot LHM, Bolger GB. Altered phosphorylation, electrophysiology, and behavior on attenuation of PDE4B action in hippocampus. BMC Neurosci 2017; 18:77. [PMID: 29197324 PMCID: PMC5712142 DOI: 10.1186/s12868-017-0396-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 11/28/2017] [Indexed: 01/19/2023] Open
Abstract
Background PDE4 cyclic nucleotide phosphodiesterases regulate 3′, 5′ cAMP abundance in the CNS and thereby regulate PKA activity and phosphorylation of CREB, which has been implicated in learning and memory, depression and other functions. The PDE4 isoform PDE4B1 also interacts with the DISC1 protein, implicated in neural development and behavioral disorders. The cellular functions of PDE4B1 have been investigated extensively, but its function(s) in the intact organism remained unexplored. Results To specifically disrupt PDE4B1, we developed mice that express a PDE4B1-D564A transgene in the hippocampus and forebrain. The transgenic mice showed enhanced phosphorylation of CREB and ERK1/2 in hippocampus. Hippocampal neurogenesis was increased in the transgenic mice. Hippocampal electrophysiological studies showed increased baseline synaptic transmission and enhanced LTP in male transgenic mice. Behaviorally, male transgenic mice showed increased activity in prolonged open field testing, but neither male nor female transgenic mice showed detectable anxiety-like behavior or antidepressant effects in the elevated plus-maze, tail-suspension or forced-swim tests. Neither sex showed any significant differences in associative fear conditioning or showed any demonstrable abnormalities in pre-pulse inhibition. Conclusions These data support the use of an isoform-selective approach to the study of PDE4B1 function in the CNS and suggest a probable role of PDE4B1 in synaptic plasticity and behavior. They also provide additional rationale and a refined approach to the development of small-molecule PDE4B1-selective inhibitors, which have potential functions in disorders of cognition, memory, mood and affect.
Collapse
Affiliation(s)
- Susan L Campbell
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.,Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA
| | - Thomas van Groen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Inga Kadish
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Lisa High Mitchell Smoot
- Department of Medicine, University of Alabama at Birmingham, NP 2501, 1720 2nd Ave S, Birmingham, AL, 35294-3300, USA
| | - Graeme B Bolger
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA. .,Department of Medicine, University of Alabama at Birmingham, NP 2501, 1720 2nd Ave S, Birmingham, AL, 35294-3300, USA. .,Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA.
| |
Collapse
|
16
|
Xiao J, Vemula SR, Xue Y, Khan MM, Carlisle FA, Waite AJ, Blake DJ, Dragatsis I, Zhao Y, LeDoux MS. Role of major and brain-specific Sgce isoforms in the pathogenesis of myoclonus-dystonia syndrome. Neurobiol Dis 2016; 98:52-65. [PMID: 27890709 DOI: 10.1016/j.nbd.2016.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/06/2016] [Accepted: 11/17/2016] [Indexed: 01/09/2023] Open
Abstract
Loss-of-function mutations in SGCE, which encodes ε-sarcoglycan (ε-SG), cause myoclonus-dystonia syndrome (OMIM159900, DYT11). A "major" ε-SG protein derived from CCDS5637.1 (NM_003919.2) and a "brain-specific" protein, that includes sequence derived from alternative exon 11b (CCDS47642.1, NM_001099400.1), are reportedly localized in post- and pre-synaptic membrane fractions, respectively. Moreover, deficiency of the "brain-specific" isoform and other isoforms derived from exon 11b may be central to the pathogenesis of DYT11. However, no animal model supports this hypothesis. Gene-trapped ES cells (CMHD-GT_148G1-3, intron 9 of NM_011360) were used to generate a novel Sgce mouse model (C57BL/6J background) with markedly reduced expression of isoforms derived from exons 3' to exon 9 of NM_011360. Among those brain regions analyzed in adult (2month-old) wild-type (WT) mice, cerebellum showed the highest relative expression of isoforms incorporating exon 11b. Homozygotes (SgceGt(148G1)Cmhd/Gt(148G1)Cmhd or SgceGt/Gt) and paternal heterozygotes (Sgcem+/pGt, m-maternal, p-paternal) showed 60 to 70% reductions in expression of total Sgce. Although expression of the major (NM_011360) and brain-specific (NM_001130189) isoforms was markedly reduced, expression of short isoforms was preserved and relatively small amounts of chimeric ε-SG/β-galactosidase fusion protein was produced by the Sgce gene-trap locus. Immunoaffinity purification followed by mass spectrometry assessments of Sgcem+/pGt mouse brain using pan- or brain-specific ε-SG antibodies revealed significant reductions of ε-SG and other interacting sarcoglycans. Genome-wide gene-expression data using RNA derived from adult Sgcem+/pGt mouse cerebellum showed that the top up-regulated genes were involved in cell cycle, cellular development, cell death and survival, while the top down-regulated genes were associated with protein synthesis, cellular development, and cell death and survival. In comparison to WT littermates, Sgcem+/pGt mice exhibited "tiptoe" gait and stimulus-induced appendicular posturing between Postnatal Days 14 to 16. Abnormalities noted in older Sgcem+/pGt mice included reduced body weight, altered gait dynamics, and reduced open-field activity. Overt spontaneous or stimulus-sensitive myoclonus was not apparent on the C57BL/6J background or mixed C57BL/6J-BALB/c and C57BL/6J-129S2 backgrounds. Our data confirm that mouse Sgce is a maternally imprinted gene and suggests that short Sgce isoforms may compensate, in part, for deficiency of major and brain-specific Sgce isoforms.
Collapse
Affiliation(s)
- Jianfeng Xiao
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Satya R Vemula
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yi Xue
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mohammad M Khan
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Francesca A Carlisle
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cathays, Cardiff, CF24 4HQ, Great Britain, United Kingdom
| | - Adrian J Waite
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cathays, Cardiff, CF24 4HQ, Great Britain, United Kingdom
| | - Derek J Blake
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cathays, Cardiff, CF24 4HQ, Great Britain, United Kingdom
| | - Ioannis Dragatsis
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yu Zhao
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mark S LeDoux
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
17
|
DeAndrade MP, Trongnetrpunya A, Yokoi F, Cheetham CC, Peng N, Wyss JM, Ding M, Li Y. Electromyographic evidence in support of a knock-in mouse model of DYT1 Dystonia. Mov Disord 2016; 31:1633-1639. [PMID: 27241685 DOI: 10.1002/mds.26677] [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: 02/02/2016] [Revised: 03/29/2016] [Accepted: 04/18/2016] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION DYT1 dystonia is an autosomal-dominant movement disorder characterized by abnormal, often repetitive, movements and postures. Its hallmark feature is sustained or intermittent contractions of muscles involving co-contractions of antagonist muscle pairs. The symptoms are relieved with the anticholinergic drug trihexyphenidyl. The primary mutation is a trinucleotide deletion (ΔGAG) in DYT1/TOR1A, which codes for torsinA. Previous studies showed that (1) heterozygous Dyt1 ΔGAG knock-in mice, which have an analogous mutation in the endogenous gene, exhibit motor deficits and altered corticostriatal synaptic plasticity in the brain and (2) these deficits can be rescued by trihexyphenidyl. However, brain imaging studies suggest that the Dyt1 knock-in mouse models nonmanifesting mutation carriers of DYT1 dystonia. The aim of this work was to examine the hallmark features of DYT1 dystonia in the Dyt1 knock-in mice by analyzing muscular activities. METHODS Wireless telemetry devices with biopotential channels were implanted to the bicep and the rectus femori muscles in Dyt1 knock-in mice, and muscular activities were recorded before and after trihexyphenidyl administration. RESULTS (1) Consistent with DYT1 dystonia patients, Dyt1 knock-in mice showed sustained contractions and co-contractions of the antagonistic bicep femoris and rectus femoris. (2) The abnormal muscle contractions were normalized by trihexyphenidyl. CONCLUSION The results suggest that the motor deficits in Dyt1 knock-in mice are likely produced by abnormal muscle contractions, and Dyt1 knock-in mice can potentially be used as a manifesting disease model to study pathophysiology and develop novel therapeutics. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Mark P DeAndrade
- Department of Neurology, College of Medicine,, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Amy Trongnetrpunya
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Fumiaki Yokoi
- Department of Neurology, College of Medicine,, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Chad C Cheetham
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ning Peng
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - J Michael Wyss
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mingzhou Ding
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Yuqing Li
- Department of Neurology, College of Medicine,, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
18
|
O'Leary H, Bernard PB, Castano AM, Benke TA. Enhanced long term potentiation and decreased AMPA receptor desensitization in the acute period following a single kainate induced early life seizure. Neurobiol Dis 2015; 87:134-44. [PMID: 26706598 DOI: 10.1016/j.nbd.2015.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/17/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023] Open
Abstract
Neonatal seizures are associated with long term disabilities including epilepsy and cognitive deficits. Using a neonatal seizure rat model that does not develop epilepsy, but develops a phenotype consistent with other models of intellectual disability (ID) and autism spectrum disorders (ASD), we sought to isolate the acute effects of a single episode of early life seizure on hippocampal CA1 synaptic development and plasticity. We have previously shown chronic changes in glutamatergic synapses, loss of long term potentiation (LTP) and enhanced long term depression (LTD), in the adult male rat ~50days following kainic acid (KA) induced early life seizure (KA-ELS) in post-natal (P) 7day old male Sprague-Dawley rats. In the present work, we examined the electrophysiological properties and expression levels of glutamate receptors in the acute period, 2 and 7days, post KA-ELS. Our results show for the first time enhanced LTP 7days after KA-ELS, but no change 2days post KA-ELS. Additionally, we report that ionotropic α-amino-3-hydroxy-5-methyl-isoxazole-propionic acid type glutamate receptor (AMPAR) desensitization is decreased in the same time frame, with no changes in AMPAR expression, phosphorylation, or membrane insertion. Inappropriate enhancement of the synaptic connections in the acute period after the seizure could alter the normal patterning of synaptic development in the hippocampus during this critical period and contribute to learning deficits. Thus, this study demonstrates a novel mechanism by which KA-ELS alters early network properties that potentially lead to adverse outcomes.
Collapse
Affiliation(s)
- Heather O'Leary
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Paul B Bernard
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Anna M Castano
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA
| | - Tim A Benke
- Department of Pediatrics, University of Colorado, School of Medicine, 80045, USA; Department of Neurology, University of Colorado, School of Medicine, 80045, USA; Department of Pharmacology, University of Colorado, School of Medicine, 80045, USA; Department of Otolaryngology, University of Colorado, School of Medicine, 80045, USA; Neuroscience Graduate Program, University of Colorado, School of Medicine, 80045, USA.
| |
Collapse
|
19
|
Correction: Behavioral and electrophysiological characterization of Dyt1 heterozygous knockout mice. PLoS One 2015; 10:e0126539. [PMID: 25874703 PMCID: PMC4398377 DOI: 10.1371/journal.pone.0126539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|