1
|
Azman KF, Zakaria R. Brain-Derived Neurotrophic Factor (BDNF) in Huntington's Disease: Neurobiology and Therapeutic Potential. Curr Neuropharmacol 2025; 23:384-403. [PMID: 40123457 DOI: 10.2174/1570159x22666240530105516] [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: 12/04/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2025] Open
Abstract
Huntington's disease is a hereditary neurodegenerative disorder marked by severe neurodegeneration in the striatum and cortex. Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family of growth factors. It plays a crucial role in maintaining the survival and proper function of striatal neurons. Depletion of BDNF has been linked to impairment and death of striatal neurons, leading to the manifestation of motor, cognitive, and behavioral dysfunctions characteristic of Huntington's disease. This review highlights the current update on the neurobiology of BDNF in the pathogenesis of Huntington's disease. The molecular evidence and the affected signaling pathways are also discussed. In addition, the impact of experimental manipulation of BDNF levels and its pharmaceutical potential for Huntington's disease treatment are explicitly reviewed.
Collapse
Affiliation(s)
- Khairunnuur Fairuz Azman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kota Bharu, Kelantan, Malaysia
| | - Rahimah Zakaria
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
2
|
Han JY, Seo J, Choi Y, Im W, Ban JJ, Sung JJ. CRISPR-Cas9 mediated genome editing of Huntington's disease neurospheres. Mol Biol Rep 2023; 50:2127-2136. [PMID: 36550260 DOI: 10.1007/s11033-022-08175-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Huntington's disease (HD) is a fatal genetic disease caused by polyglutamine aggregation encoded by an expanded CAG repeat in the huntingtin gene (HTT). In this study, we cultured neurospheres derived from R6/2 mice, a representative animal model of HD, as an in vitro model. GuideRNAs were designed to induce large deletion or frameshift indel mutation of CAG expansion. These gRNAs and Cas9 were delivered to the R6/2 neurospheres and disease-related phenotypes were observed. METHODS AND RESULTS Deletion or indel mutation of the CAG repeat was confirmed by PCR, T7E1 assay and sequencing of the edited neurospheres. Edited neurospheres showed decreased polyglutamine aggregation compared with control HD neurospheres. In the edited neurosphere, we confirmed the upregulation of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and brain-derived neurotrophic factor (BDNF), whose reduced expressions are closely involved in the disease progression. In addition, flow cytometry result showed an increase in cell viability with an overall decrease in necrotic and apoptotic populations among edited R6/2 neurospheres. Additional siRNA experiments confirmed that the increased viability was decreased through inhibition of PGC-1α or BDNF. CONCLUSION Our study confirmed that CAG repeat of R6/2 mouse-derived neurospheres can be edited through CRISPR-Cas9. Editing of CAG repeat sequence decreases polyglutamine aggregation and cellular apoptosis of HD neurospheres, which may be related to the increased expressions of PGC-1α and BDNF. Our data provide the evidence that CRISPR-Cas9 mediated genome editing has therapeutic potential on HD neuronal cells.
Collapse
Affiliation(s)
- Ji Yun Han
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 110-744, South Korea
| | - Jaewoo Seo
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 110-744, South Korea
| | - Yoori Choi
- Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea.,Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Wooseok Im
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae-Jun Ban
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 110-744, South Korea. .,Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea.
| | - Jung-Joon Sung
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 110-744, South Korea. .,Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
3
|
Li SH, Abd-Elrahman KS, Ferguson SS. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacol Ther 2022; 239:108275. [DOI: 10.1016/j.pharmthera.2022.108275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
|
4
|
Group II Metabotropic Glutamate Receptors Reduce Apoptosis and Regulate BDNF and GDNF Levels in Hypoxic-Ischemic Injury in Neonatal Rats. Int J Mol Sci 2022; 23:ijms23137000. [PMID: 35806000 PMCID: PMC9266366 DOI: 10.3390/ijms23137000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/13/2022] Open
Abstract
Birth asphyxia causes brain injury in neonates, but a fully successful treatment has yet to be developed. This study aimed to investigate the effect of group II mGlu receptors activation after experimental birth asphyxia (hypoxia-ischemia) on the expression of factors involved in apoptosis and neuroprotective neurotrophins. Hypoxia-ischemia (HI) on 7-day-old rats was used as an experimental model. The effects of intraperitoneal application of mGluR2 agonist LY379268 (5 mg/kg) and the specific mGluR3 agonist NAAG (5 mg/kg) (1 h or 6 h after HI) on apoptotic processes and initiation of the neuroprotective mechanism were investigated. LY379268 and NAAG applied shortly after HI prevented brain damage and significantly decreased pro-apoptotic Bax and HtrA2/Omi expression, increasing expression of anti-apoptotic Bcl-2. NAAG or LY379268 applied at both times also decreased HIF-1α formation. HI caused a significant decrease in BDNF concentration, which was restored after LY379268 or NAAG administration. HI-induced increase in GDNF concentration was decreased after administration of LY379268 or NAAG. Our results show that activation of mGluR2/3 receptors shortly after HI prevents brain damage by the inhibition of excessive glutamate release and apoptotic damage decrease. mGluR2 and mGluR3 agonists produced comparable results, indicating that both receptors may be a potential target for early treatment in neonatal HI.
Collapse
|
5
|
Wang H, Del Mar N, Deng Y, Reiner A. Rescue of BDNF expression by the thalamic parafascicular nucleus with chronic treatment with the mGluR2/3 agonist LY379268 may contribute to the LY379268 rescue of enkephalinergic striatal projection neurons in R6/2 Huntington's disease mice. Neurosci Lett 2021; 763:136180. [PMID: 34416343 DOI: 10.1016/j.neulet.2021.136180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022]
Abstract
We have found that daily subcutaneous injection with a maximum tolerated dose of the mGluR2/3 agonist LY379268 (20 mg/kg) beginning at 4 weeks of age dramatically improves the motor, neuronal and neurochemical phenotype in R6/2 mice, a rapidly progressing transgenic model of Huntington's disease (HD). We also previously showed that the benefit of daily LY379268 in R6/2 mice was associated with increases in corticostriatal brain-derived neurotrophic factor (BDNF), and in particular was associated with a reduction in enkephalinergic striatal projection neuron loss. In the present study, we show that daily LY379268 also rescues expression of BDNF by neurons of the thalamic parafascicular nucleus in R6/2 mice, which projects prominently to the striatum, and this increase too is linked to the rescue of enkephalinergic striatal neurons. Thus, LY379268 may protect enkephalinergic striatal projection neurons from loss by boosting BDNF production and delivery via both the corticostriatal and thalamostriatal projection systems. These results suggest that chronic treatment with mGluR2/3 agonists may represent an approach for slowing enkephalinergic neuron loss in HD, and perhaps progression in general.
Collapse
Affiliation(s)
- H Wang
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| | - N Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| | - Y Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| | - A Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
6
|
Deng Y, Wang H, Joni M, Sekhri R, Reiner A. Progression of basal ganglia pathology in heterozygous Q175 knock-in Huntington's disease mice. J Comp Neurol 2021; 529:1327-1371. [PMID: 32869871 PMCID: PMC8049038 DOI: 10.1002/cne.25023] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/07/2020] [Accepted: 08/26/2020] [Indexed: 12/29/2022]
Abstract
We used behavioral testing and morphological methods to detail the progression of basal ganglia neuron type-specific pathology and the deficits stemming from them in male heterozygous Q175 mice, compared to age-matched WT males. A rotarod deficit was not present in Q175 mice until 18 months, but increased open field turn rate (reflecting hyperkinesia) and open field anxiety were evident at 6 months. No loss of striatal neurons was seen out to 18 months, but ENK+ and DARPP32+ striatal perikarya were fewer by 6 months, due to diminished expression, with further decline by 18 months. No reduction in SP+ striatal perikarya or striatal interneurons was seen in Q175 mice at 18 months, but cholinergic interneurons showed dendrite attenuation by 6 months. Despite reduced ENK expression in indirect pathway striatal perikarya, ENK-immunostained terminals in globus pallidus externus (GPe) were more abundant at 6 months and remained so out to 18 months. Similarly, SP-immunostained terminals from striatal direct pathway neurons were more abundant in globus pallidus internus and substantia nigra at 6 months and remained so at 18 months. FoxP2+ arkypallidal GPe neurons and subthalamic nucleus neurons were lost by 18 months but not prototypical PARV+ GPe neurons or dopaminergic nigral neurons. Our results show that striatal projection neuron abnormalities and behavioral abnormalities reflecting them develop between 2 and 6 months of age in Q175 male heterozygotes, indicating early effects of the HD mutation. The striatal pathologies resemble those in human HD, but are less severe at 18 months than even in premanifest HD.
Collapse
Affiliation(s)
- Yunping Deng
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Hongbing Wang
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Marion Joni
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Radhika Sekhri
- Department of PathologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Anton Reiner
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Department of OphthalmologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| |
Collapse
|
7
|
Tang X, Ross CA, Johnson H, Paulsen JS, Younes L, Albin RL, Ratnanather JT, Miller MI. Regional subcortical shape analysis in premanifest Huntington's disease. Hum Brain Mapp 2018; 40:1419-1433. [PMID: 30376191 DOI: 10.1002/hbm.24456] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/18/2018] [Accepted: 10/23/2018] [Indexed: 11/11/2022] Open
Abstract
Huntington's disease (HD) involves preferential and progressive degeneration of striatum and other subcortical regions as well as regional cortical atrophy. It is caused by a CAG repeat expansion in the Huntingtin gene, and the longer the expansion the earlier the age of onset. Atrophy begins prior to manifest clinical signs and symptoms, and brain atrophy in premanifest expansion carriers can be studied. We employed a diffeomorphometric pipeline to contrast subcortical structures' morphological properties in a control group with three disease groups representing different phases of premanifest HD (far, intermediate, and near to onset) as defined by the length of the CAG expansion and the participant's age (CAG-Age-Product). A total of 1,428 magnetic resonance image scans from 694 participants from the PREDICT-HD cohort were used. We found significant region-specific atrophies in all subcortical structures studied, with the estimated abnormality onset time varying from structure to structure. Heterogeneous shape abnormalities of caudate nuclei were present in premanifest HD participants estimated furthest from onset and putaminal shape abnormalities were present in participants intermediate to onset. Thalamic, hippocampal, and amygdalar shape abnormalities were present in participants nearest to onset. We assessed whether the estimated progression of subcortical pathology in premanifest HD tracked specific pathways. This is plausible for changes in basal ganglia circuits but probably not for changes in hippocampus and amygdala. The regional shape analyses conducted in this study provide useful insights into the effects of HD pathology in subcortical structures.
Collapse
Affiliation(s)
- Xiaoying Tang
- Department of Electrical and Electronic Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Christopher A Ross
- Division of Neurobiology, Departments of Psychiatry, Neurology, Neuroscience and Pharmacology, and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hans Johnson
- Departments of Neurology and Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jane S Paulsen
- Departments of Neurology and Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Laurent Younes
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland.,Center for Imaging Science, Johns Hopkins University, Baltimore, Maryland.,Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Roger L Albin
- Neurology Service and GRECC, VAAAHS, Ann Arbor, Michigan.,Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan
| | - J Tilak Ratnanather
- Center for Imaging Science, Johns Hopkins University, Baltimore, Maryland.,Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Michael I Miller
- Center for Imaging Science, Johns Hopkins University, Baltimore, Maryland.,Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
8
|
Simmons DA. Modulating Neurotrophin Receptor Signaling as a Therapeutic Strategy for Huntington's Disease. J Huntingtons Dis 2018; 6:303-325. [PMID: 29254102 PMCID: PMC5757655 DOI: 10.3233/jhd-170275] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by CAG repeat expansions in the IT15 gene which encodes the huntingtin (HTT) protein. Currently, no treatments capable of preventing or slowing disease progression exist. Disease modifying therapeutics for HD would be expected to target a comprehensive set of degenerative processes given the diverse mechanisms contributing to HD pathogenesis including neuroinflammation, excitotoxicity, and transcription dysregulation. A major contributor to HD-related degeneration is mutant HTT-induced loss of neurotrophic support. Thus, neurotrophin (NT) receptors have emerged as therapeutic targets in HD. The considerable overlap between NT signaling networks and those dysregulated by mutant HTT provides strong theoretical support for this approach. This review will focus on the contributions of disrupted NT signaling in HD-related neurodegeneration and how targeting NT receptors to augment pro-survival signaling and/or to inhibit degenerative signaling may combat HD pathologies. Therapeutic strategies involving NT delivery, peptidomimetics, and the targeting of specific NT receptors (e.g., Trks or p75NTR), particularly with small molecule ligands, are discussed.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
9
|
Dong BE, Xue Y, Sakata K. The effect of enriched environment across ages: A study of anhedonia and BDNF gene induction. GENES BRAIN AND BEHAVIOR 2018; 17:e12485. [PMID: 29717802 DOI: 10.1111/gbb.12485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/26/2018] [Accepted: 04/26/2018] [Indexed: 12/19/2022]
Abstract
Enriched environment treatment (EET) is a potential intervention for depression by inducing brain-derived neurotrophic factor (BDNF). However, its age dependency remains unclear. We recently found that EET during early-life development (ED) was effective in increasing exploratory activity and anti-despair behavior, particularly in promoter IV-driven BDNF deficient mice (KIV), with the largest BDNF protein induction in the hippocampus and frontal cortex. Here, we further determined age dependency of EET effects on anhedonia and promoter-specific BDNF transcription, by using the sucrose preference test and qRT-PCR. Wild-type (WT) and KIV mice received 2 months of EET during ED, young-adulthood and old-adulthood (0-2, 2-4 and 12-14 months, respectively). All KIV groups showed reduced sucrose preference, which EET equally reversed regardless of age. EET increased hippocampal BDNF mRNA levels for all ages and genotypes, but increased frontal cortex BDNF mRNA levels only in ED KIV and old WT mice. Transcription by promoters I and IV was age-dependent in the hippocampus of WT mice: more effective induction of exon IV or I during ED or old-adulthood, respectively. Transcription by almost all 9 promoters was age-specific in the frontal cortex, mostly observed in ED KIV mice. After discontinuance of EET, the EET effects on anti-anhedonia and BDNF transcription in both regions persisted only in ED KIV mice. These results suggested that EET was equally effective in reversing anhedonia and inducing hippocampal BDNF transcription, but was more effective during ED in inducing frontal cortex BDNF transcription and for lasting anti-anhedonic and BDNF effects particularly in promoter IV-BDNF deficiency.
Collapse
Affiliation(s)
- B E Dong
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Y Xue
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - K Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
10
|
Reiner A, Deng Y. Disrupted striatal neuron inputs and outputs in Huntington's disease. CNS Neurosci Ther 2018; 24:250-280. [PMID: 29582587 PMCID: PMC5875736 DOI: 10.1111/cns.12844] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/22/2022] Open
Abstract
Huntington's disease (HD) is a hereditary progressive neurodegenerative disorder caused by a CAG repeat expansion in the gene coding for the protein huntingtin, resulting in a pathogenic expansion of the polyglutamine tract in the N-terminus of this protein. The HD pathology resulting from the mutation is most prominent in the striatal part of the basal ganglia, and progressive differential dysfunction and loss of striatal projection neurons and interneurons account for the progression of motor deficits seen in this disease. The present review summarizes current understanding regarding the progression in striatal neuron dysfunction and loss, based on studies both in human HD victims and in genetic mouse models of HD. We review evidence on early loss of inputs to striatum from cortex and thalamus, which may be the basis of the mild premanifest bradykinesia in HD, as well as on the subsequent loss of indirect pathway striatal projection neurons and their outputs to the external pallidal segment, which appears to be the basis of the chorea seen in early symptomatic HD. Later loss of direct pathway striatal projection neurons and their output to the internal pallidal segment account for the severe akinesia seen late in HD. Loss of parvalbuminergic striatal interneurons may contribute to the late dystonia and rigidity.
Collapse
Affiliation(s)
- Anton Reiner
- Department of Anatomy & NeurobiologyThe University of Tennessee Health Science CenterMemphisTNUSA
- Department of OphthalmologyThe University of Tennessee Health Science CenterMemphisTNUSA
| | - Yun‐Ping Deng
- Department of Anatomy & NeurobiologyThe University of Tennessee Health Science CenterMemphisTNUSA
| |
Collapse
|
11
|
Dragatsis I, Dietrich P, Ren H, Deng YP, Del Mar N, Wang HB, Johnson IM, Jones KR, Reiner A. Effect of early embryonic deletion of huntingtin from pyramidal neurons on the development and long-term survival of neurons in cerebral cortex and striatum. Neurobiol Dis 2017; 111:102-117. [PMID: 29274742 PMCID: PMC5821111 DOI: 10.1016/j.nbd.2017.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 11/07/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022] Open
Abstract
We evaluated the impact of early embryonic deletion of huntingtin (htt) from pyramidal neurons on cortical development, cortical neuron survival and motor behavior, using a cre-loxP strategy to inactivate the mouse htt gene (Hdh) in emx1-expressing cell lineages. Western blot confirmed substantial htt reduction in cerebral cortex of these Emx-httKO mice, with residual cortical htt in all likelihood restricted to cortical interneurons of the subpallial lineage and/or vascular endothelial cells. Despite the loss of htt early in development, cortical lamination was normal, as revealed by layer-specific markers. Cortical volume and neuron abundance were, however, significantly less than normal, and cortical neurons showed reduced brain-derived neurotrophic factor (BDNF) expression and reduced activation of BDNF signaling pathways. Nonetheless, cortical volume and neuron abundance did not show progressive age-related decline in Emx-httKO mice out to 24 months. Although striatal neurochemistry was normal, reductions in striatal volume and neuron abundance were seen in Emx-httKO mice, which were again not progressive. Weight maintenance was normal in Emx-httKO mice, but a slight rotarod deficit and persistent hyperactivity were observed throughout the lifespan. Our results show that embryonic deletion of htt from developing pallium does not substantially alter migration of cortical neurons to their correct laminar destinations, but does yield reduced cortical and striatal size and neuron numbers. The Emx-httKO mice were persistently hyperactive, possibly due to defects in corticostriatal development. Importantly, deletion of htt from cortical pyramidal neurons did not yield age-related progressive cortical or striatal pathology.
Collapse
Affiliation(s)
- I Dragatsis
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - P Dietrich
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - H Ren
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Y P Deng
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - N Del Mar
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - H B Wang
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - I M Johnson
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - K R Jones
- Department of Molecular, Cellular, & Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309, United States
| | - A Reiner
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
12
|
Sakata K, Overacre AE. Promoter IV-BDNF deficiency disturbs cholinergic gene expression of CHRNA5, CHRM2, and CHRM5: effects of drug and environmental treatments. J Neurochem 2017; 143:49-64. [PMID: 28722769 DOI: 10.1111/jnc.14129] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 11/29/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) promotes maturation of cholinergic neurons. However, how activity-dependent BDNF expression affects specific cholinergic gene expression remains unclear. This study addressed this question by determining mRNA levels of 22 acetylcholine receptor subunits, the choline transporter (CHT), and the choline acetyltransferase (ChAT) in mice deficient in activity-dependent BDNF via promoter IV (KIV) and control wild-type mice. Quantitative RT-PCR revealed significant reductions in nicotinic acetylcholine receptor alpha 5 (CHRNA5) in the frontal cortex and hippocampus and M5 muscarinic acetylcholine receptor (CHRM5) in the hippocampus, but significant increases in M2 muscarinic acetylcholine receptor (CHRM2) in the frontal cortex of KIV mice compared to wild-type mice. Three-week treatments with fluoxetine, phenelzine, duloxetine, imipramine, or an enriched environment treatment (EET) did not affect the altered expression of these genes except that EET increased CHRNA5 levels only in KIV frontal cortex. EET also increased levels of CHRNA7, CHT, and ChAT, again only in the KIV frontal cortex. The imipramine treatment was most prominent among the four antidepressants; it up-regulated hippocampal CHRM2 and frontal cortex CHRM5 in both genotypes, and frontal cortex CHRNA7 only in KIV mice. To the best of our knowledge, this is the first evidence that BDNF deficiency disturbs expression of CHRNA5, CHRM2, and CHRM5. Our results suggest that promoter IV-BDNF deficiency - which occurs under chronic stress - causes cholinergic dysfunctions via these receptors. EET is effective on CHRNA5, while its compensatory induction of other cholinergic genes or drugs targeting CHRNA5, CHRM2, and CHRM5 may become an alternative strategy to reverse these BDNF-linked cholinergic dysfunctions.
Collapse
Affiliation(s)
- Kazuko Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Abigail E Overacre
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
13
|
Wright DJ, Renoir T, Gray LJ, Hannan AJ. Huntington’s Disease: Pathogenic Mechanisms and Therapeutic Targets. ADVANCES IN NEUROBIOLOGY 2017; 15:93-128. [DOI: 10.1007/978-3-319-57193-5_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
14
|
Metabotropic glutamate receptors and neurodegenerative diseases. Pharmacol Res 2017; 115:179-191. [DOI: 10.1016/j.phrs.2016.11.013] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
|
15
|
Pollock K, Dahlenburg H, Nelson H, Fink KD, Cary W, Hendrix K, Annett G, Torrest A, Deng P, Gutierrez J, Nacey C, Pepper K, Kalomoiris S, D Anderson J, McGee J, Gruenloh W, Fury B, Bauer G, Duffy A, Tempkin T, Wheelock V, Nolta JA. Human Mesenchymal Stem Cells Genetically Engineered to Overexpress Brain-derived Neurotrophic Factor Improve Outcomes in Huntington's Disease Mouse Models. Mol Ther 2016; 24:965-77. [PMID: 26765769 PMCID: PMC4881765 DOI: 10.1038/mt.2016.12] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 12/05/2015] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a fatal degenerative autosomal dominant neuropsychiatric disease that causes neuronal death and is characterized by progressive striatal and then widespread brain atrophy. Brain-derived neurotrophic factor (BDNF) is a lead candidate for the treatment of HD, as it has been shown to prevent cell death and to stimulate the growth and migration of new neurons in the brain in transgenic mouse models. BDNF levels are reduced in HD postmortem human brain. Previous studies have shown efficacy of mesenchymal stem/stromal cells (MSC)/BDNF using murine MSCs, and the present study used human MSCs to advance the therapeutic potential of the MSC/BDNF platform for clinical application. Double-blinded studies were performed to examine the effects of intrastriatally transplanted human MSC/BDNF on disease progression in two strains of immune-suppressed HD transgenic mice: YAC128 and R6/2. MSC/BDNF treatment decreased striatal atrophy in YAC128 mice. MSC/BDNF treatment also significantly reduced anxiety as measured in the open-field assay. Both MSC and MSC/BDNF treatments induced a significant increase in neurogenesis-like activity in R6/2 mice. MSC/BDNF treatment also increased the mean lifespan of the R6/2 mice. Our genetically modified MSC/BDNF cells set a precedent for stem cell-based neurotherapeutics and could potentially be modified for other neurodegenerative disorders such as amyotrophic lateral sclerosis, Alzheimer's disease, and some forms of Parkinson's disease. These cells provide a platform delivery system for future studies involving corrective gene-editing strategies.
Collapse
Affiliation(s)
- Kari Pollock
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Heather Dahlenburg
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Haley Nelson
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Kyle D Fink
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Whitney Cary
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Kyle Hendrix
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Geralyn Annett
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Audrey Torrest
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Peter Deng
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Joshua Gutierrez
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Catherine Nacey
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Karen Pepper
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Stefanos Kalomoiris
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Johnathon D Anderson
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Jeannine McGee
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - William Gruenloh
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Brian Fury
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Gerhard Bauer
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Alexandria Duffy
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Theresa Tempkin
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Vicki Wheelock
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Jan A Nolta
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| |
Collapse
|
16
|
Abstract
In this review, we explore the similarities and differences in the behavioural neurobiology found in the mouse models of Huntington's disease (HD) and the human disease state. The review is organised with a comparative focus on the functional domains of motor control, cognition and behavioural disturbance (akin to psychiatric disturbance in people) and how our knowledge of the underlying physiological changes that are manifest in the HD mouse lines correspond to those seen in the HD clinical population. The review is framed in terms of functional circuitry and neurotransmitter systems and how abnormalities in these systems impact on the behavioural readouts across the mouse lines and how these may correspond to the deficits observed in people. In addition, interpretational issues associated with the data from animal studies are discussed.
Collapse
Affiliation(s)
- Simon P Brooks
- Brain Repair Group, Division of Neuroscience, Cardiff University School of Bioscience, Museum Avenue, Cardiff, Wales, UK,
| | | |
Collapse
|
17
|
Fink KD, Deng P, Torrest A, Stewart H, Pollock K, Gruenloh W, Annett G, Tempkin T, Wheelock V, Nolta JA. Developing stem cell therapies for juvenile and adult-onset Huntington's disease. Regen Med 2015; 10:623-46. [PMID: 26237705 PMCID: PMC6785015 DOI: 10.2217/rme.15.25] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Stem cell therapies have been explored as a new avenue for the treatment of neurologic disease and damage within the CNS in part due to their native ability to mimic repair mechanisms in the brain. Mesenchymal stem cells have been of particular clinical interest due to their ability to release beneficial neurotrophic factors and their ability to foster a neuroprotective microenviroment. While early stem cell transplantation therapies have been fraught with technical and political concerns as well as limited clinical benefits, mesenchymal stem cell therapies have been shown to be clinically beneficial and derivable from nonembryonic, adult sources. The focus of this review will be on emerging and extant stem cell therapies for juvenile and adult-onset Huntington's disease.
Collapse
Affiliation(s)
- Kyle D Fink
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Peter Deng
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
- GenomeCenter, Biochemistry & Molecular Medicine, University of California, 451 Health Sciences Dr. Davis, CA 95616, USA
| | - Audrey Torrest
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Heather Stewart
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Kari Pollock
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - William Gruenloh
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Geralyn Annett
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Teresa Tempkin
- Department of Neurology, University of California Davis Health Systems, 4860 Y Street Sacramento, CA 95817, USA
| | - Vicki Wheelock
- Department of Neurology, University of California Davis Health Systems, 4860 Y Street Sacramento, CA 95817, USA
| | - Jan A Nolta
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| |
Collapse
|
18
|
Mu S, Lin E, Liu B, Ma Y, OuYang L, Li Y, Chen S, Zhang J, Lei W. Melatonin reduces projection neuronal injury induced by 3-nitropropionic acid in the rat striatum. NEURODEGENER DIS 2014; 14:139-50. [PMID: 25342207 DOI: 10.1159/000365891] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 07/10/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Melatonin has shown a protective effect against various oxidative damages in the nervous system. Our previous studies have also confirmed its effect on behavioral dysfunction of experimental rats and injury of striatal interneurons induced by 3-nitropropionic acid. The present study aimed to further determine the effect of melatonin on the injury of striatal projection neurons induced by 3-nitropropionic acid. METHODS Classic histology, immunohistochemistry, Western blotting and immunoelectron microscopy were applied in this study. RESULTS The results were as follows: (1) in the striatum, 3-nitropropionic acid induced a clear lesion area with a transition zone around it, in which both D1+ and D2+ fibers were decreased significantly. However, in the group with melatonin treatment, the striatal lesion area was smaller than in the 3-nitropropionic acid group and the loss of D1+ and D2+ fibers was less pronounced than in the 3-nitropropionic acid group. (2) Histochemical results showed that the dendritic spine density of striatal projection neurons was decreased more seriously after 3-nitropropionic acid treatment, whereas the loss of dendritic spines was less marked in the melatonin-treated group than in the 3- nitropropionic acid group. Immunoelectron microscopy showed that the density of D1+ and D2+ dendrites and spines was significantly decreased in the 3-nitropropionic acid group, and the loss of D1+ and D2+ spines as well as D2+ dendrites was significantly reversed by melatonin administration. (3) Western blotting showed that the expression level of projection neuron protein markers decreased more significantly in the 3-nitropropionic acid group than in the control group and increased significantly in the melatonin-treated group. CONCLUSIONS The present results suggest that 3-nitropropionic acid induces serious injury of striatal projection neurons and that melatonin effectively protects against this pathological damage.
Collapse
Affiliation(s)
- Shuhua Mu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Deng YP, Wong T, Wan JY, Reiner A. Differential loss of thalamostriatal and corticostriatal input to striatal projection neuron types prior to overt motor symptoms in the Q140 knock-in mouse model of Huntington's disease. Front Syst Neurosci 2014; 8:198. [PMID: 25360089 PMCID: PMC4197654 DOI: 10.3389/fnsys.2014.00198] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/23/2014] [Indexed: 11/13/2022] Open
Abstract
Motor slowing and forebrain white matter loss have been reported in premanifest Huntington's disease (HD) prior to substantial striatal neuron loss. These findings raise the possibility that early motor defects in HD may be related to loss of excitatory input to striatum. In a prior study, we showed that in the heterozygous Q140 knock-in mouse model of HD that loss of thalamostriatal axospinous terminals is evident by 4 months, and loss of corticostriatal axospinous terminals is evident at 12 months, before striatal projection neuron pathology. In the present study, we specifically characterized the loss of thalamostriatal and corticostriatal terminals on direct (dSPN) and indirect (iSPN) pathway striatal projection neurons, using immunolabeling to identify thalamostriatal (VGLUT2+) and corticostriatal (VGLUT1+) axospinous terminals, and D1 receptor immunolabeling to distinguish dSPN (D1+) and iSPN (D1-) synaptic targets. We found that the loss of corticostriatal terminals at 12 months of age was preferential for D1+ spines, and especially involved smaller terminals, presumptively of the intratelencephalically projecting (IT) type. By contrast, indirect pathway D1- spines showed little loss of axospinous terminals at the same age. Thalamostriatal terminal loss was comparable for D1+ and D1- spines at both 4 and 12 months. Regression analysis showed that the loss of VGLUT1+ terminals on D1+ spines was correlated with a slight decline in open field motor parameters at 12 months. Our overall results raise the possibility that differential thalamic and cortical input loss to SPNs is an early event in human HD, with cortical loss to dSPNs in particular contributing to premanifest motor slowing.
Collapse
Affiliation(s)
- Yun-Ping Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center Memphis, TN, USA
| | - Ting Wong
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center Memphis, TN, USA
| | - Jim Y Wan
- Department of Preventive Medicine, The University of Tennessee Health Science Center Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center Memphis, TN, USA
| |
Collapse
|
20
|
Dhanya RP, Sheffler DJ, Dahl R, Davis M, Lee PS, Yang L, Nickols HH, Cho HP, Smith LH, D'Souza MS, Conn PJ, Der-Avakian A, Markou A, Cosford NDP. Design and synthesis of systemically active metabotropic glutamate subtype-2 and -3 (mGlu2/3) receptor positive allosteric modulators (PAMs): pharmacological characterization and assessment in a rat model of cocaine dependence. J Med Chem 2014; 57:4154-72. [PMID: 24735492 PMCID: PMC4033659 DOI: 10.1021/jm5000563] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
![]()
As
part of our ongoing small-molecule metabotropic glutamate (mGlu) receptor
positive allosteric modulator (PAM) research, we performed structure–activity
relationship (SAR) studies around a series of group II mGlu PAMs.
Initial analogues exhibited weak activity as mGlu2 receptor
PAMs and no activity at mGlu3. Compound optimization led
to the identification of potent mGlu2/3 selective PAMs
with no in vitro activity at mGlu1,4–8 or 45 other
CNS receptors. In vitro pharmacological characterization of representative
compound 44 indicated agonist-PAM activity toward mGlu2 and PAM activity at mGlu3. The most potent mGlu2/3 PAMs were characterized in assays predictive of ADME/T
and pharmacokinetic (PK) properties, allowing the discovery of systemically
active mGlu2/3 PAMs. On the basis of its overall profile,
compound 74 was selected for behavioral studies and was
shown to dose-dependently decrease cocaine self-administration in
rats after intraperitoneal administration. These mGlu2/3 receptor PAMs have significant potential as small molecule tools
for investigating group II mGlu pharmacology.
Collapse
Affiliation(s)
- Raveendra-Panickar Dhanya
- Cell Death and Survival Networks Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute , 10901 N. Torrey Pines Road, La Jolla, California 92037, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Mishra J, Chaudhary T, Kumar A. Rosiglitazone synergizes the neuroprotective effects of valproic acid against quinolinic acid-induced neurotoxicity in rats: targeting PPARγ and HDAC pathways. Neurotox Res 2014; 26:130-51. [PMID: 24566814 DOI: 10.1007/s12640-014-9458-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 02/01/2014] [Accepted: 02/04/2014] [Indexed: 12/15/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant progressive neurodegenerative disorder which affects medium spiny GABAergic neurons mainly in the striatum. Oxidative damage, neuro-inflammation, apoptosis, protein aggregation, and signaling of neurotrophic factors are some of the common cellular pathways involved in HD. Quinolinic acid (QA) causes excitotoxicity by stimulating N-methyl-D-aspartate receptors via calcium overload leading to neurodegeneration. Neuroprotective potential of peroxisome proliferator activated receptor-γ (PPARγ) agonists and histone deacetylase (HDAC) inhibitors have been well documented in experimental models of neurodegenerative disorders; however, their exact mechanisms are not clear. Therefore, present study has been designed to explore possible neuroprotective mechanism of valproic acid (VPA) and its interaction with rosiglitazone against QA induced HD-like symptoms in rats. Single bilateral intrastriatal QA (200 nmol/2 μl saline) administration significantly caused motor incoordination, memory impairment, oxidative damage, mitochondrial dysfunction (complex I, II, II and IV), cellular alterations [tumor necrosis factor-alpha (TNF-α), caspase-3, brain derived neurotrophic factor, acetylcholinesterase], and striatal neurodegeneration as compared to sham group. Treatment with rosiglitazone (5, 10 mg/kg) and VPA (100, 200 mg/kg) for 21 days significantly attenuated these behavioral, biochemical, and cellular alterations as compared to control (QA 200 nmol) group. However, VPA (100 mg/kg) treatment in combination with rosiglitazone (5 mg/kg) for 21 days synergized their neuroprotective effect, which was significant as compared to their effects per se in QA-treated animals. The present study provides an evidence of possible interplay of PPARγ agonists and HDAC inhibitors as a novel therapeutic strategy in the management of HD.
Collapse
Affiliation(s)
- Jitendriya Mishra
- Pharmacology Division, UGC Centre of Advanced Study (UGC-CAS), University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | | | | |
Collapse
|
22
|
Heldt SA, Elberger AJ, Deng Y, Guley NH, Del Mar N, Rogers J, Choi GW, Ferrell J, Rex TS, Honig MG, Reiner A. A novel closed-head model of mild traumatic brain injury caused by primary overpressure blast to the cranium produces sustained emotional deficits in mice. Front Neurol 2014; 5:2. [PMID: 24478749 PMCID: PMC3898331 DOI: 10.3389/fneur.2014.00002] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/06/2014] [Indexed: 12/14/2022] Open
Abstract
Emotional disorders are a common outcome from mild traumatic brain injury (TBI) in humans, but their pathophysiological basis is poorly understood. We have developed a mouse model of closed-head blast injury using an air pressure wave delivered to a small area on one side of the cranium, to create mild TBI. We found that 20-psi blasts in 3-month-old C57BL/6 male mice yielded no obvious behavioral or histological evidence of brain injury, while 25-40 psi blasts produced transient anxiety in an open field arena but little histological evidence of brain damage. By contrast, 50-60 psi blasts resulted in anxiety-like behavior in an open field arena that became more evident with time after blast. In additional behavioral tests conducted 2-8 weeks after blast, 50-60 psi mice also demonstrated increased acoustic startle, perseverance of learned fear, and enhanced contextual fear, as well as depression-like behavior and diminished prepulse inhibition. We found no evident cerebral pathology, but did observe scattered axonal degeneration in brain sections from 50 to 60 psi mice 3-8 weeks after blast. Thus, the TBI caused by single 50-60 psi blasts in mice exhibits the minimal neuronal loss coupled to "diffuse" axonal injury characteristic of human mild TBI. A reduction in the abundance of a subpopulation of excitatory projection neurons in basolateral amygdala enriched in Thy1 was, however, observed. The reported link of this neuronal population to fear suppression suggests their damage by mild TBI may contribute to the heightened anxiety and fearfulness observed after blast in our mice. Our overpressure air blast model of concussion in mice will enable further studies of the mechanisms underlying the diverse emotional deficits seen after mild TBI.
Collapse
Affiliation(s)
- Scott A. Heldt
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andrea J. Elberger
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yunping Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Natalie H. Guley
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nobel Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joshua Rogers
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gy Won Choi
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jessica Ferrell
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tonia S. Rex
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, Nashville, TN, USA
| | - Marcia G. Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
23
|
Sakata K, Duke SM. Lack of BDNF expression through promoter IV disturbs expression of monoamine genes in the frontal cortex and hippocampus. Neuroscience 2013; 260:265-75. [PMID: 24345476 DOI: 10.1016/j.neuroscience.2013.12.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/29/2013] [Accepted: 12/06/2013] [Indexed: 02/07/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is implicated in the pathophysiology of psychiatric conditions including major depression and schizophrenia. Mice lacking activity-driven BDNF expression through promoter IV (knock-in promoter IV: KIV) exhibit depression-like behavior, inflexible learning, and impaired response inhibition. Monoamine systems (serotonin, dopamine, and noradrenaline) are suggested to be involved in depression and schizophrenia since many of the current antidepressants and antipsychotics increase the brain levels of monoamines and/or act on monoamine receptors. To elucidate the impact of activity-driven BDNF on the monoamine systems, we examined mRNA levels for 30 monoamine-related genes, including receptors, transporters, and synthesizing enzymes, in KIV and control wild-type mice by using quantitative reverse-transcription polymerase chain reaction (qRT-PCR). mRNA levels were measured in the frontal cortex and hippocampus, which are regions related to depression and schizophrenia and where promoter IV is active. The frontal cortex of KIV mice showed reduced levels of mRNA expression for serotonin receptors 1b, 2a, and 5b (5HTR1b, 5HTR2a, 5HTR5b), dopamine D2 receptors (DRD2), and adrenergic receptors alpha 1a and 1d (AdRα1a and AdRα1b), but increased levels for serotonin synthesizing enzyme, tryptophan hydroxylase (TPH), and dopamine D4 receptor (DRD4) when compared to control wild-type mice. The hippocampus of KIV mice showed decreased levels of 5HTR5b. Our results provide causal evidence that lack of promoter IV-driven BDNF disturbs expression of monoaminergic genes in the frontal cortex and hippocampus. These disturbed expression changes in the monoamine systems may mediate the depression- and schizophrenia-like behavior of KIV mice. Our results also suggest that antidepressant and antipsychotic treatments may actually interfere with and normalize the disturbed monoamine systems caused by reduced activity-dependent BDNF, while the treatment responses to these drugs may differ in the subject with reduced BDNF levels caused by stress and lack of neuronal activity.
Collapse
Affiliation(s)
- K Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - S M Duke
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
24
|
Kumar A, Chaudhary T, Mishra J. Minocycline modulates neuroprotective effect of hesperidin against quinolinic acid induced Huntington's disease like symptoms in rats: behavioral, biochemical, cellular and histological evidences. Eur J Pharmacol 2013; 720:16-28. [PMID: 24211676 DOI: 10.1016/j.ejphar.2013.10.057] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/25/2013] [Accepted: 10/29/2013] [Indexed: 12/18/2022]
Abstract
Emerging evidences indicate hesperidin, a citrus flavanone, attenuates neurodegenerative processes and related complications. Besides its anti-oxidant properties, the other probable mechanisms which underpin its neuroprotective potential are still not clear. In light of emerging role of flavonoids in modulating oxidative stress and neuro-inflammation, the study has been designed to explore the possible neuroprotective effect of hesperidin and its combination with minocycline (microglial inhibitor), against quinolinic acid (QA) induced Huntington's disease (HD) like symptoms in rats. Unilateral intrastriatal administration of QA (300 nmol/4 µl) significantly reduced body weight, impaired behavior (locomotor activity, beam balance and memory performance), caused oxidative damage (increased lipid peroxidation, nitrite concentration, depleted super oxide dismutase and reduced glutathione), demonstrated mitochondrial dysfunction (decreased Complex-I, II, III, and IV activities), increased striatal lesion volume and altered the levels of TNF-α, caspase-3 as well as BDNF expression, as compared to sham group. Meanwhile, chronic hesperidin (100mg/kg, p.o.) and minocycline (25mg/kg, p.o.) treatment for 21 days significantly attenuated the behavioral, biochemical and cellular alterations as compared to QA treated (control) animals, whereas hesperidin (50mg/kg, p.o.) treatment was found to be non-significant. However, treatment of hesperidin (50mg/kg) in combination with minocycline (25mg/kg) potentiated their neuroprotective effect, which was significant as compared to their effects per se in QA treated animals. Taken altogether, the results of the present study suggest a possible interplay of microglial modulation and anti-oxidant effect in neuroprotective potential of hesperidin against QA induced HD like symptoms in rats.
Collapse
Affiliation(s)
- Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh 160014, India.
| | | | | |
Collapse
|
25
|
Loss of corticostriatal and thalamostriatal synaptic terminals precedes striatal projection neuron pathology in heterozygous Q140 Huntington's disease mice. Neurobiol Dis 2013; 60:89-107. [PMID: 23969239 DOI: 10.1016/j.nbd.2013.08.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 07/22/2013] [Accepted: 08/07/2013] [Indexed: 01/18/2023] Open
Abstract
Motor slowing, forebrain white matter loss, and striatal shrinkage have been reported in premanifest Huntington's disease (HD) prior to overt striatal neuron loss. We carried out detailed LM and EM studies in a genetically precise HD mimic, heterozygous Q140 HD knock-in mice, to examine the possibility that loss of corticostriatal and thalamostriatal terminals prior to striatal neuron loss underlies these premanifest HD abnormalities. In our studies, we used VGLUT1 and VGLUT2 immunolabeling to detect corticostriatal and thalamostriatal (respectively) terminals in dorsolateral (motor) striatum over the first year of life, prior to striatal projection neuron pathology. VGLUT1+ axospinous corticostriatal terminals represented about 55% of all excitatory terminals in striatum, and VGLUT2+ axospinous thalamostriatal terminals represented about 35%, with VGLUT1+ and VGLUT2+ axodendritic terminals accounting for the remainder. In Q140 mice, a significant 40% shortfall in VGLUT2+ axodendritic thalamostriatal terminals and a 20% shortfall in axospinous thalamostriatal terminals were already observed at 1 month of age, but VGLUT1+ terminals were normal in abundance. The 20% deficiency in VGLUT2+ thalamostriatal axospinous terminals persisted at 4 and 12 months in Q140 mice, and an additional 30% loss of VGLUT1+ corticostriatal terminals was observed at 12 months. The early and persistent deficiency in thalamostriatal axospinous terminals in Q140 mice may reflect a development defect, and the impoverishment of this excitatory drive to striatum may help explain early motor defects in Q140 mice and in premanifest HD. The loss of corticostriatal terminals at 1 year in Q140 mice is consistent with prior evidence from other mouse models of corticostriatal disconnection early during progression, and can explain both the measurable bradykinesia and striatal white matter loss in late premanifest HD.
Collapse
|
26
|
Role of BDNF in Central Motor Structures and Motor Diseases. Mol Neurobiol 2013; 48:783-93. [DOI: 10.1007/s12035-013-8466-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 04/22/2013] [Indexed: 12/29/2022]
|
27
|
Sakata K, Mastin JR, Duke SM, Vail MG, Overacre AE, Dong BE, Jha S. Effects of antidepressant treatment on mice lacking brain-derived neurotrophic factor expression through promoter IV. Eur J Neurosci 2013; 37:1863-74. [DOI: 10.1111/ejn.12148] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 12/01/2022]
Affiliation(s)
- Kazuko Sakata
- Department of Pharmacology/Psychiatry; College of Medicine; University of Tennessee Health Science Center; 874 Union Avenue, Room 430; Memphis; TN; 38163; USA
| | - Joshua R. Mastin
- Department of Pharmacology/Psychiatry; College of Medicine; University of Tennessee Health Science Center; 874 Union Avenue, Room 430; Memphis; TN; 38163; USA
| | - Sean M. Duke
- Department of Pharmacology/Psychiatry; College of Medicine; University of Tennessee Health Science Center; 874 Union Avenue, Room 430; Memphis; TN; 38163; USA
| | - Meghan G. Vail
- Department of Pharmacology/Psychiatry; College of Medicine; University of Tennessee Health Science Center; 874 Union Avenue, Room 430; Memphis; TN; 38163; USA
| | - Abigail E. Overacre
- Department of Pharmacology/Psychiatry; College of Medicine; University of Tennessee Health Science Center; 874 Union Avenue, Room 430; Memphis; TN; 38163; USA
| | - Brittany E. Dong
- Department of Pharmacology/Psychiatry; College of Medicine; University of Tennessee Health Science Center; 874 Union Avenue, Room 430; Memphis; TN; 38163; USA
| | - Shanker Jha
- Department of Pharmacology/Psychiatry; College of Medicine; University of Tennessee Health Science Center; 874 Union Avenue, Room 430; Memphis; TN; 38163; USA
| |
Collapse
|