1
|
Xiong H, Tang F, Guo Y, Xu R, Lei P. Neural Circuit Changes in Neurological Disorders: Evidence from in vivo Two-photon Imaging. Ageing Res Rev 2023; 87:101933. [PMID: 37061201 DOI: 10.1016/j.arr.2023.101933] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
Neural circuits, such as synaptic plasticity and neural activity, are critical components of healthy brain function. The consequent dynamic remodeling of neural circuits is an ongoing procedure affecting neuronal activities. Disruption of this essential process results in diseases. Advanced microscopic applications such as two-photon laser scanning microscopy have recently been applied to understand neural circuit changes during disease since it can visualize fine structural and functional cellular activation in living animals. In this review, we have summarized the latest work assessing the dynamic rewiring of postsynaptic dendritic spines and modulation of calcium transients in neurons of the intact living brain, focusing on their potential roles in neurological disorders (e.g. Alzheimer's disease, stroke, and epilepsy). Understanding the fine changes that occurred in the brain during disease is crucial for future clinical intervention developments.
Collapse
Affiliation(s)
- Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China; Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Fei Tang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Yujie Guo
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China.
| |
Collapse
|
2
|
Martin-Lopez E, Vidyadhara DJ, Liberia T, Meller SJ, Harmon LE, Hsu RM, Spence N, Brennan B, Han K, Yücel B, Chandra SS, Greer CA. α-Synuclein Pathology and Reduced Neurogenesis in the Olfactory System Affect Olfaction in a Mouse Model of Parkinson's Disease. J Neurosci 2023; 43:1051-1071. [PMID: 36596700 PMCID: PMC9908323 DOI: 10.1523/jneurosci.1526-22.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023] Open
Abstract
Parkinson's disease (PD) is characterized by multiple symptoms including olfactory dysfunction, whose underlying mechanisms remain unclear. Here, we explored pathologic changes in the olfactory pathway of transgenic (Tg) mice of both sexes expressing the human A30P mutant α-synuclein (α-syn; α-syn-Tg mice) at 6-7 and 12-14 months of age, representing early and late-stages of motor progression, respectively. α-Syn-Tg mice at late stages exhibited olfactory behavioral deficits, which correlated with severe α-syn pathology in projection neurons (PNs) of the olfactory pathway. In parallel, olfactory bulb (OB) neurogenesis in α-syn-Tg mice was reduced in the OB granule cells at six to seven months and OB periglomerular cells at 12-14 months, respectively, both of which could contribute to olfactory dysfunction. Proteomic analyses showed a disruption in endocytic and exocytic pathways in the OB during the early stages which appeared exacerbated at the synaptic terminals when the mice developed olfactory deficits at 12-14 months. Our data suggest that (1) the α-syn-Tg mice recapitulate the olfactory functional deficits seen in PD; (2) olfactory structures exhibit spatiotemporal disparities for vulnerability to α-syn pathology; (3) α-syn pathology is restricted to projection neurons in the olfactory pathway; (4) neurogenesis in adult α-syn-Tg mice is reduced in the OB; and (5) synaptic endocytosis and exocytosis defects in the OB may further explain olfactory deficits.SIGNIFICANCE STATEMENT Olfactory dysfunction is a characteristic symptom of Parkinson's disease (PD). Using the human A30P mutant α-synuclein (α-syn)-expressing mouse model, we demonstrated the appearance of olfactory deficits at late stages of the disease, which was accompanied by the accumulation of α-syn pathology in projection neurons (PNs) of the olfactory system. This dysfunction included a reduction in olfactory bulb (OB) neurogenesis as well as changes in synaptic vesicular transport affecting synaptic function, both of which are likely contributing to olfactory behavioral deficits.
Collapse
Affiliation(s)
- Eduardo Martin-Lopez
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - D J Vidyadhara
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Teresa Liberia
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Sarah J Meller
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Leah E Harmon
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Ryan M Hsu
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Natalie Spence
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Bowen Brennan
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Kimberly Han
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Betül Yücel
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Sreeganga S Chandra
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Charles A Greer
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| |
Collapse
|
3
|
How Well Do Rodent Models of Parkinson's Disease Recapitulate Early Non-Motor Phenotypes? A Systematic Review. Biomedicines 2022; 10:biomedicines10123026. [PMID: 36551782 PMCID: PMC9775565 DOI: 10.3390/biomedicines10123026] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
The prodromal phase of Parkinson's disease (PD) is characterised by many non-motor symptoms, and these have recently been posited to be predictive of later diagnosis. Genetic rodent models can develop non-motor phenotypes, providing tools to identify mechanisms underlying the early development of PD. However, it is not yet clear how reproducible non-motor phenotypes are amongst genetic PD rodent models, whether phenotypes are age-dependent, and the translatability of these phenotypes has yet to be explored. A systematic literature search was conducted on studies using genetic PD rodent models to investigate non-motor phenotypes; cognition, anxiety/depressive-like behaviour, gastrointestinal (GI) function, olfaction, circadian rhythm, cardiovascular and urinary function. In total, 51 genetic models of PD across 150 studies were identified. We found outcomes of most phenotypes were inconclusive due to inadequate studies, assessment at different ages, or variation in experimental and environmental factors. GI dysfunction was the most reproducible phenotype across all genetic rodent models. The mouse model harbouring mutant A53T, and the wild-type hα-syn overexpression (OE) model recapitulated the majority of phenotypes, albeit did not reliably produce concurrent motor deficits and nigral cell loss. Furthermore, animal models displayed different phenotypic profiles, reflecting the distinct genetic risk factors and heterogeneity of disease mechanisms. Currently, the inconsistent phenotypes within rodent models pose a challenge in the translatability and usefulness for further biomechanistic investigations. This review highlights opportunities to improve phenotype reproducibility with an emphasis on phenotypic assay choice and robust experimental design.
Collapse
|
4
|
Aniszewska A, Bergström J, Ingelsson M, Ekmark-Lewén S. Modeling Parkinson's disease-related symptoms in alpha-synuclein overexpressing mice. Brain Behav 2022; 12:e2628. [PMID: 35652155 PMCID: PMC9304846 DOI: 10.1002/brb3.2628] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Intracellular deposition of alpha-synuclein (α-syn) as Lewy bodies and Lewy neurites is a central event in the pathogenesis of Parkinson's disease (PD) and other α-synucleinopathies. Transgenic mouse models overexpressing human α-syn, are useful research tools in preclinical studies of pathogenetic mechanisms. Such mice develop α-syn inclusions as well as neurodegeneration with a topographical distribution that varies depending on the choice of promoter and which form of α-syn that is overexpressed. Moreover, they display motor symptoms and cognitive disturbances that to some extent resemble the human conditions. PURPOSE One of the main motives for assessing behavior in these mouse models is to evaluate the potential of new treatment strategies, including their impact on motor and cognitive symptoms. However, due to a high within-group variability with respect to such features, the behavioral studies need to be applied with caution. In this review, we discuss how to make appropriate choices in the experimental design and which tests that are most suitable for the evaluation of PD-related symptoms in such studies. METHODS We have evaluated published results on two selected transgenic mouse models overexpressing wild type (L61) and mutated (A30P) α-syn in the context of their validity and utility for different types of behavioral studies. CONCLUSIONS By applying appropriate behavioral tests, α-syn transgenic mouse models provide an appropriate experimental platform for studies of symptoms related to PD and other α-synucleinopathies.
Collapse
Affiliation(s)
- Agata Aniszewska
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Joakim Bergström
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden.,Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada.,Department of Medicine and Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Ontario, Canada
| | - Sara Ekmark-Lewén
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
5
|
Chen F, Liu W, Liu P, Wang Z, Zhou Y, Liu X, Li A. α-Synuclein aggregation in the olfactory bulb induces olfactory deficits by perturbing granule cells and granular-mitral synaptic transmission. NPJ Parkinsons Dis 2021; 7:114. [PMID: 34903719 PMCID: PMC8668919 DOI: 10.1038/s41531-021-00259-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 11/21/2021] [Indexed: 11/10/2022] Open
Abstract
Olfactory dysfunction is an early pre-motor symptom of Parkinson's disease (PD) but the neural mechanisms underlying this dysfunction remain largely unknown. Aggregation of α-synuclein is observed in the olfactory bulb (OB) during the early stages of PD, indicating a relationship between α-synuclein pathology and hyposmia. Here we investigate whether and how α-synuclein aggregates modulate neural activity in the OB at the single-cell and synaptic levels. We induced α-synuclein aggregation specifically in the OB via overexpression of double-mutant human α-synuclein by an adeno-associated viral (AAV) vector. We found that α-synuclein aggregation in the OB decreased the ability of mice to detect odors and to perceive attractive odors. The spontaneous activity and odor-evoked firing rates of single mitral/tufted cells (M/Ts) were increased by α-synuclein aggregates with the amplitude of odor-evoked high-gamma oscillations increased. Furthermore, the decreased activity in granule cells (GCs) and impaired inhibitory synaptic function were responsible for the observed hyperactivity of M/Ts induced by α-synuclein aggregates. These results provide direct evidences of the role of α-synuclein aggregates on PD-related olfactory dysfunction and reveal the neural circuit mechanisms by which olfaction is modulated by α-synuclein pathology.
Collapse
Affiliation(s)
- Fengjiao Chen
- grid.417303.20000 0000 9927 0537Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Wei Liu
- grid.417303.20000 0000 9927 0537Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Penglai Liu
- grid.417303.20000 0000 9927 0537Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Zhen Wang
- grid.417303.20000 0000 9927 0537School of Life Science, Xuzhou Medical University, Xuzhou, China
| | - You Zhou
- grid.417303.20000 0000 9927 0537School of Life Science, Xuzhou Medical University, Xuzhou, China
| | - Xingyu Liu
- grid.417303.20000 0000 9927 0537School of Life Science, Xuzhou Medical University, Xuzhou, China
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
6
|
Behere A, Thörnqvist PO, Winberg S, Ingelsson M, Bergström J, Ekmark-Lewén S. Visualization of early oligomeric α-synuclein pathology and its impact on the dopaminergic system in the (Thy-1)-h[A30P]α-syn transgenic mouse model. J Neurosci Res 2021; 99:2525-2539. [PMID: 34292621 DOI: 10.1002/jnr.24927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/02/2021] [Accepted: 07/02/2021] [Indexed: 12/17/2022]
Abstract
Aggregation of alpha-synuclein (α-syn) into Lewy bodies and Lewy neurites is a pathological hallmark in the Parkinson´s disease (PD) brain. The formation of α-syn oligomers is believed to be an early pathogenic event and the A30P mutation in the gene encoding α-syn, causing familial PD, has been shown to cause an accelerated oligomerization. Due to the problem of preserving antigen conformation on tissue surfaces, α-syn oligomers are difficult to detect ex vivo using conventional immunohistochemistry with oligomer-selective antibodies. Herein, we have instead employed the previously reported α-syn oligomer proximity ligation assay (ASO-PLA), along with a wide variety of biochemical assays, to discern the pathological progression of α-syn oligomers and their impact on the dopaminergic system in male and female (Thy-1)-h[A30P]α-syn transgenic (A30P-tg) mice. Our results reveal a previously undetected abundance of α-syn oligomers in midbrain of young mice, whereas phosphorylated (pS129) and proteinase k-resistant α-syn species were observed to a larger extent in aged mice. Although we did not detect loss of dopaminergic neurons in A30P-tg mice, a dysregulation in the monoaminergic system was recorded in older mice. Taken together, ASO-PLA should be a useful method for the detection of early changes in α-syn aggregation on brain tissue, from experimental mouse models in addition to post mortem PD cases.
Collapse
Affiliation(s)
- Anish Behere
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Per-Ove Thörnqvist
- Department of Neuroscience, Physiology Unit, Uppsala University, Uppsala, Sweden
| | - Svante Winberg
- Department of Neuroscience, Physiology Unit, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Joakim Bergström
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Sara Ekmark-Lewén
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
7
|
Zhang XM, Anwar S, Kim Y, Brown J, Comte I, Cai H, Cai NN, Wade-Martins R, Szele FG. The A30P α-synuclein mutation decreases subventricular zone proliferation. Hum Mol Genet 2019; 28:2283-2294. [PMID: 31267130 PMCID: PMC6606853 DOI: 10.1093/hmg/ddz057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/05/2019] [Accepted: 03/14/2019] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease (PD) is associated with olfactory defects in addition to dopaminergic degeneration. Dopaminergic signalling is necessary for subventricular zone (SVZ) proliferation and olfactory bulb (OB) neurogenesis. Alpha-synuclein (α-syn or Snca) modulates dopaminergic neurotransmission, and SNCA mutations cause familial PD, but how α-syn and its mutations affect adult neurogenesis is unclear. To address this, we studied a bacterial artificial chromosome transgenic mouse expressing the A30P SNCA familial PD point mutation on an Snca-/- background. We confirmed that the SNCA-A30P transgene recapitulates endogenous α-syn expression patterns and levels by immunohistochemical detection of endogenous α-syn in a wild-type mouse and transgenic SNCA-A30P α-syn protein in the forebrain. The number of SVZ stem cells (BrdU+GFAP+) was decreased in SNCA-A30P mice, whereas proliferating (phospho-histone 3+) cells were decreased in Snca-/- and even more so in SNCA-A30P mice. Similarly, SNCA-A30P mice had fewer Mash1+ transit-amplifying SVZ progenitor cells but Snca-/- mice did not. These data suggest the A30P mutation aggravates the effect of Snca loss in the SVZ. Interestingly, calbindin+ and calretinin (CalR)+ periglomerular neurons were decreased in both Snca-/-, and SNCA-A30P mice but tyrosine hydroxylase+ periglomerular OB neurons were only decreased in Snca-/- mice. Cell death decreased in the OB granule layer of Snca-/- and SNCA-A30P mice. In the same region, CalR+ numbers increased in Snca-/- and SNCA-A30P mice. Thus, α-syn loss and human A30P SNCA decrease SVZ proliferation, cell death in the OB and differentially alter interneuron numbers. Similar disruptions in human neurogenesis may contribute to the olfactory deficits, which are observed in PD.
Collapse
Affiliation(s)
- Xue-Ming Zhang
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford, UK
- College of Veterinary Medicine, Jilin University, Xi-an Road, Changchun, China
| | - Sabina Anwar
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford, UK
| | - Yongsoo Kim
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford, UK
| | - Jennifer Brown
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford, UK
| | - Isabelle Comte
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford, UK
| | - Huan Cai
- College of Veterinary Medicine, Jilin University, Xi-an Road, Changchun, China
| | - Ning-Ning Cai
- College of Veterinary Medicine, Jilin University, Xi-an Road, Changchun, China
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford, UK
| |
Collapse
|
8
|
Liang H, Ding B, Liang J, Shi X, Jiang X, Gao Y. MicroRNA-10a inhibits A30P α-synuclein aggregation and toxicity by targeting proapoptotic protein BCL2L11. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:624-633. [PMID: 31938148 PMCID: PMC6958036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 12/22/2017] [Indexed: 06/10/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder around the world, and is characterized by progressive loss of nigrostriatal dopaminergic neurons. Certain microRNAs (miRNAs) are aberrantly expressed in the post-mortem brain tissues of patients with PD and in vivo PD model mice. However, the role of brain-enriched miRNA (miR)-10a in PD has not been studied. To investigate the regulatory role of miR-10a on α-synuclein (α-syn) in the pathology of PD, the present study aimed to examine whether upregulation of miR-10a attenuated A30P α-syn mutant aggregation and cellular toxicity. miRNA expression analysis by reverse transcription-quantitative polymerase chain reaction demonstrated that miR-10a expression was decreased in the midbrain of A30P α-syn transgenic mice and in SH-SY5Y human neuroblastoma cells transfected with A30P α-syn. In addition, miR-10a mimics were used to upregulate miR-10a expression. It was revealed that the upregulation of miR-10a suppressed α-syn intracellular accumulation and toxicity in α-syn-overexpressing SH-SY5Y cells. In addition, miR-10a overexpression resulted in a reversal of the A30P α-syn-induced upregulation of proapoptotic protein Bcl-2-associated X protein and cleaved caspase-3 expression and downregulation of antiapoptotic protein B-cell lymphoma-2 (BCL2) expression. A luciferase reporter assay demonstrated that BCL2-like 11 (BCL2L11), an apoptosis inducer, was a novel target gene of miR-10a. A30P α-syn aggregation and toxicity were alleviated by knocking down endogenous BCL2L11 in SH-SY5Y cells using a small interfering RNA specific for BCL2L11. In conclusion, these results demonstrate that miR-10a may serve a functional role in α-syn-induced neuronal pathology by inhibiting expression of BCL2L11 and that upregulation of miR-10a expression may be a useful therapeutic strategy for the treatment of PD.
Collapse
Affiliation(s)
- Huimin Liang
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| | - Bingqian Ding
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| | - Junhui Liang
- Department of Laboratory Medicine, Wugang People’s HospitalPingdingshan, Henan, P. R. China
| | - Xiaoyan Shi
- College of Pharmacy, Henan UniversityKaifeng, Henan, P. R. China
| | - Xin Jiang
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| | - Yang Gao
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| |
Collapse
|
9
|
Abstract
Neural stem cells (NSCs) give rise to the entire nervous system. Animal models suggest that defects in NSC proliferation and differentiation contribute to several brain disorders (e.g., microcephaly, macrocephaly, autism, schizophrenia, and Huntington's disease). However, animal models of such diseases do not fully recapitulate all disease-related phenotypes because of substantial differences in brain development between rodents and humans. Therefore, additional human-based evidence is required to understand the mechanisms that are involved in the development of neurological diseases that result from human NSC (hNSC) dysfunction. Human-induced pluripotent stem cells provide a new model to investigate the contribution of hNSCs to various neurological pathologies. In this chapter, we review the role of hNSCs in both neurodevelopment- and neurodegeneration-related human brain pathologies, with an emphasis on recent evidence that has been obtained using embryonic stem cell- or induced pluripotent stem cell-derived hNSCs and progenitors.
Collapse
Affiliation(s)
- Ewa Liszewska
- International Institute of Molecular and Cell Biology, Warsaw, Poland.
| | - Jacek Jaworski
- International Institute of Molecular and Cell Biology, Warsaw, Poland.
| |
Collapse
|
10
|
Langley MR, Ghaisas S, Ay M, Luo J, Palanisamy BN, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Manganese exposure exacerbates progressive motor deficits and neurodegeneration in the MitoPark mouse model of Parkinson's disease: Relevance to gene and environment interactions in metal neurotoxicity. Neurotoxicology 2018; 64:240-255. [PMID: 28595911 PMCID: PMC5736468 DOI: 10.1016/j.neuro.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is now recognized as a neurodegenerative condition caused by a complex interplay of genetic and environmental influences. Chronic manganese (Mn) exposure has been implicated in the development of PD. Since mitochondrial dysfunction is associated with PD pathology as well as Mn neurotoxicity, we investigated whether Mn exposure augments mitochondrial dysfunction and neurodegeneration in the nigrostriatal dopaminergic system using a newly available mitochondrially defective transgenic mouse model of PD, the MitoPark mouse. This unique PD model recapitulates key features of the disease including progressive neurobehavioral changes and neuronal degeneration. We exposed MitoPark mice to a low dose of Mn (10mg/kg, p.o.) daily for 4 weeks starting at age 8 wks and then determined the behavioral, neurochemical and histological changes. Mn exposure accelerated the rate of progression of motor deficits in MitoPark mice when compared to the untreated MitoPark group. Mn also worsened olfactory function in this model. Most importantly, Mn exposure intensified the depletion of striatal dopamine and nigral TH neuronal loss in MitoPark mice. The neurodegenerative changes were accompanied by enhanced oxidative damage in the striatum and substantia nigra (SN) of MitoPark mice treated with Mn. Furthermore, Mn-treated MitoPark mice had significantly more oligomeric protein and IBA-1-immunoreactive microglia cells, suggesting Mn augments neuroinflammatory processes in the nigrostriatal pathway. To further confirm the direct effect of Mn on impaired mitochondrial function, we also generated a mitochondrially defective dopaminergic cell model by knocking out the TFAM transcription factor by using a CRISPR-Cas9 gene-editing method. Seahorse mitochondrial bioenergetic analysis revealed that Mn decreases mitochondrial basal and ATP-linked respiration in the TFAM KO cells. Collectively, our results reveal that Mn can augment mitochondrial dysfunction to exacerbate nigrostriatal neurodegeneration and PD-related behavioral symptoms. Our study also demonstrates that the MitoPark mouse is an excellent model to study the gene-environment interactions associated with mitochondrial defects in the nigral dopaminergic system as well as to evaluate the contribution of potential environmental toxicant interactions in a slowly progressive model of Parkinsonism.
Collapse
Affiliation(s)
- Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Jie Luo
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
11
|
Regensburger M, Schreglmann SR, Stoll S, Rockenstein E, Loskarn S, Xiang W, Masliah E, Winner B. Oligomer-prone E57K-mutant alpha-synuclein exacerbates integration deficit of adult hippocampal newborn neurons in transgenic mice. Brain Struct Funct 2017; 223:1357-1368. [PMID: 29124353 PMCID: PMC5869938 DOI: 10.1007/s00429-017-1561-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 11/02/2017] [Indexed: 11/26/2022]
Abstract
In the adult mammalian hippocampus, new neurons are constantly added to the dentate gyrus. Adult neurogenesis is impaired in several neurodegenerative mouse models including α-synuclein (a-syn) transgenic mice. Among different a-syn species, a-syn oligomers were reported to be the most toxic species for neurons. Here, we studied the impact of wild-type vs. oligomer-prone a-syn on neurogenesis. We compared the wild-type a-syn transgenic mouse model (Thy1-WTS) to its equivalent transgenic for oligomer-prone E57K-mutant a-syn (Thy1-E57K). Transgenic a-syn was highly expressed within the hippocampus of both models, but was not present within adult neural stem cells and neuroblasts. Proliferation and survival of newly generated neurons were unchanged in both transgenic models. Thy1-WTS showed a minor integration deficit regarding mushroom spine density of newborn neurons, whereas Thy1-E57K exhibited a severe reduction of all spines. We conclude that cell-extrinsic a-syn impairs mushroom spine formation of adult newborn neurons and that oligomer-prone a-syn exacerbates this integration deficit. Moreover, our data suggest that a-syn reduces the survival of newborn neurons by a cell-intrinsic mechanism during the early neuroblast development. The finding of increased spine pathology in Thy1-E57K is a new pathogenic function of oligomeric a-syn and precedes overt neurodegeneration. Thus, it may constitute a readout for therapeutic approaches.
Collapse
Affiliation(s)
- Martin Regensburger
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- IZKF Junior Research Group III, and BMBF Research Group Neuroscience, FAU, Erlangen, Germany
- Department of Neurology, FAU, Erlangen, Germany
| | - Sebastian R Schreglmann
- School of Medicine, University of Regensburg, Regensburg, Germany
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, UCL, London, UK
| | - Svenja Stoll
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- IZKF Junior Research Group III, and BMBF Research Group Neuroscience, FAU, Erlangen, Germany
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Sandra Loskarn
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- IZKF Junior Research Group III, and BMBF Research Group Neuroscience, FAU, Erlangen, Germany
- Department of Neurology, FAU, Erlangen, Germany
| | - Wei Xiang
- Institute of Biochemistry, FAU, Erlangen, Germany
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- IZKF Junior Research Group III, and BMBF Research Group Neuroscience, FAU, Erlangen, Germany.
| |
Collapse
|
12
|
Neurogenesis from Sox2 expressing cells in the adult cerebellar cortex. Sci Rep 2017; 7:6137. [PMID: 28733588 PMCID: PMC5522437 DOI: 10.1038/s41598-017-06150-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 06/09/2017] [Indexed: 02/06/2023] Open
Abstract
We identified a rare undifferentiated cell population that is intermingled with the Bergmann glia of the adult murine cerebellar cortex, expresses the stem cell markers Sox2 and Nestin, and lacks markers of glial or neuronal differentiation. Interestingly, such Sox2+ S100- cells of the adult cerebellum expanded after adequate physiological stimuli in mice (exercise), and Sox2+ precursors acquired positivity for the neuronal marker NeuN over time and integrated into cellular networks. In human patients, SOX2+ S100- cells similarly increased in number after relevant pathological insults (infarcts), suggesting a similar expansion of cells that lack terminal glial differentiation.
Collapse
|
13
|
Fortuna JTS, Gralle M, Beckman D, Neves FS, Diniz LP, Frost PS, Barros-Aragão F, Santos LE, Gonçalves RA, Romão L, Zamberlan DC, Soares FAA, Braga C, Foguel D, Gomes FCA, De Felice FG, Ferreira ST, Clarke JR, Figueiredo CP. Brain infusion of α-synuclein oligomers induces motor and non-motor Parkinson's disease-like symptoms in mice. Behav Brain Res 2017; 333:150-160. [PMID: 28668282 DOI: 10.1016/j.bbr.2017.06.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/16/2017] [Accepted: 06/27/2017] [Indexed: 01/05/2023]
Abstract
Parkinson's disease (PD) is characterized by motor dysfunction, which is preceded by a number of non-motor symptoms including olfactory deficits. Aggregation of α-synuclein (α-syn) gives rise to Lewy bodies in dopaminergic neurons and is thought to play a central role in PD pathology. However, whether amyloid fibrils or soluble oligomers of α-syn are the main neurotoxic species in PD remains controversial. Here, we performed a single intracerebroventricular (i.c.v.) infusion of α-syn oligomers (α-SYOs) in mice and evaluated motor and non-motor symptoms. Familiar bedding and vanillin essence discrimination tasks showed that α-SYOs impaired olfactory performance of mice, and decreased TH and dopamine levels in the olfactory bulb early after infusion. The olfactory deficit persisted until 45days post-infusion (dpi). α- SYO-infused mice behaved normally in the object recognition and forced swim tests, but showed increased anxiety-like behavior in the open field and elevated plus maze tests 20 dpi. Finally, administration of α-SYOs induced late motor impairment in the pole test and rotarod paradigms, along with reduced TH and dopamine content in the caudate putamen, 45 dpi. Reduced number of TH-positive cells was also seen in the substantia nigra of α-SYO-injected mice compared to control. In conclusion, i.c.v. infusion of α-SYOs recapitulated some of PD-associated non-motor symptoms, such as increased anxiety and olfactory dysfunction, but failed to recapitulate memory impairment and depressive-like behavior typical of the disease. Moreover, α-SYOs i.c.v. administration induced motor deficits and loss of TH and dopamine levels, key features of PD. Results point to α-syn oligomers as the proximal neurotoxins responsible for early non-motor and motor deficits in PD and suggest that the i.c.v. infusion model characterized here may comprise a useful tool for identification of PD novel therapeutic targets and drug screening.
Collapse
Affiliation(s)
- Juliana T S Fortuna
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Matthias Gralle
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Danielle Beckman
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Fernanda S Neves
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Luan P Diniz
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Paula S Frost
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Fernanda Barros-Aragão
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Luís E Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Rafaella A Gonçalves
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Luciana Romão
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Campus Xerém, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Daniele C Zamberlan
- Department of Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Felix A A Soares
- Department of Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Carolina Braga
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Campus Xerém, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Debora Foguel
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Flávia C A Gomes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| | - Cláudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| |
Collapse
|
14
|
Molecular, Cellular and Functional Effects of Radiation-Induced Brain Injury: A Review. Int J Mol Sci 2015; 16:27796-815. [PMID: 26610477 PMCID: PMC4661926 DOI: 10.3390/ijms161126068] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/10/2015] [Accepted: 10/23/2015] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy is the most effective non-surgical treatment of primary brain tumors and metastases. Preclinical studies have provided valuable insights into pathogenesis of radiation-induced injury to the central nervous system. Radiation-induced brain injury can damage neuronal, glial and vascular compartments of the brain and may lead to molecular, cellular and functional changes. Given its central role in memory and adult neurogenesis, the majority of studies have focused on the hippocampus. These findings suggested that hippocampal avoidance in cranial radiotherapy prevents radiation-induced cognitive impairment of patients. However, multiple rodent studies have shown that this problem is more complex. As the radiation-induced cognitive impairment reflects hippocampal and non-hippocampal compartments, it is of critical importance to investigate molecular, cellular and functional modifications in various brain regions as well as their integration at clinically relevant doses and schedules. We here provide a literature overview, including our previously published results, in order to support the translation of preclinical findings to clinical practice, and improve the physical and mental status of patients with brain tumors.
Collapse
|
15
|
Le Grand JN, Gonzalez-Cano L, Pavlou MA, Schwamborn JC. Neural stem cells in Parkinson's disease: a role for neurogenesis defects in onset and progression. Cell Mol Life Sci 2015; 72:773-97. [PMID: 25403878 PMCID: PMC11113294 DOI: 10.1007/s00018-014-1774-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 10/09/2014] [Accepted: 11/03/2014] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, leading to a variety of motor and non-motor symptoms. Interestingly, non-motor symptoms often appear a decade or more before the first signs of motor symptoms. Some of these non-motor symptoms are remarkably similar to those observed in cases of impaired neurogenesis and several PD-related genes have been shown to play a role in embryonic or adult neurogenesis. Indeed, animal models deficient in Nurr1, Pitx3, SNCA and PINK1 display deregulated embryonic neurogenesis and LRRK2 and VPS35 have been implicated in neuronal development-related processes such as Wnt/β-catenin signaling and neurite outgrowth. Moreover, adult neurogenesis is affected in both PD patients and PD animal models and is regulated by dopamine and dopaminergic (DA) receptors, by chronic neuroinflammation, such as that observed in PD, and by differential expression of wild-type or mutant forms of PD-related genes. Indeed, an increasing number of in vivo studies demonstrate a role for SNCA and LRRK2 in adult neurogenesis and in the generation and maintenance of DA neurons. Finally, the roles of PD-related genes, SNCA, LRRK2, VPS35, Parkin, PINK1 and DJ-1 have been studied in NSCs, progenitor cells and induced pluripotent stem cells, demonstrating a role for some of these genes in stem/progenitor cell proliferation and maintenance. Together, these studies strongly suggest a link between deregulated neurogenesis and the onset and progression of PD and present strong evidence that, in addition to a neurodegenerative disorder, PD can also be regarded as a developmental disorder.
Collapse
Affiliation(s)
- Jaclyn Nicole Le Grand
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Laura Gonzalez-Cano
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Maria Angeliki Pavlou
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
16
|
Neuner J, Ovsepian SV, Dorostkar M, Filser S, Gupta A, Michalakis S, Biel M, Herms J. Pathological α-synuclein impairs adult-born granule cell development and functional integration in the olfactory bulb. Nat Commun 2014; 5:3915. [PMID: 24867427 PMCID: PMC4050256 DOI: 10.1038/ncomms4915] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 04/18/2014] [Indexed: 12/21/2022] Open
Abstract
Although the role of noxious α-synuclein (α-SYN) in the degeneration of midbrain dopaminergic
neurons and associated motor deficits of Parkinson’s disease is
recognized, its impact on non-motor brain circuits and related symptoms remains
elusive. Through combining in vivo two-photon imaging with time-coded
labelling of neurons in the olfactory bulb of A30P α-SYN transgenic mice, we show impaired growth and
branching of dendrites of adult-born granule cells (GCs), with reduced gain and
plasticity of dendritic spines. The spine impairments are especially pronounced
during the critical phase of integration of new neurons into existing circuits.
Functionally, retarded dendritic expansion translates into reduced electrical
capacitance with enhanced intrinsic excitability and responsiveness of GCs to
depolarizing inputs, while the spine loss correlates with decreased frequency of
AMPA-mediated miniature EPSCs.
Changes described here are expected to interfere with the functional integration and
survival of new GCs into bulbar networks, contributing towards olfactory deficits
and related behavioural impairments. Aggregation-prone forms of α-synuclein lead to
degeneration of midbrain dopaminergic neurons, as seen in Parkinson’s
disease, but less is known about the effects that the noxious protein has in other brain
regions. Here, the authors investigate the effect of a pathological form of
α-synuclein on the functional integration of new neurons into the olfactory
bulb of adult mice.
Collapse
Affiliation(s)
- Johanna Neuner
- Center for Neuropathology and Prion Research, Department for Translationsal Brain Research, Ludwig Maximilian University, Feodor-Lynen-Strassee 23, Munich 81377, Germany
| | - Saak V Ovsepian
- German Center for Neurodegeneratione Diseases (DZNE), Department for Translational Brain Research, Feodor-Lynen-Strasse 23, Munich 81377, Germany
| | - Mario Dorostkar
- Center for Neuropathology and Prion Research, Department for Translationsal Brain Research, Ludwig Maximilian University, Feodor-Lynen-Strassee 23, Munich 81377, Germany
| | - Severin Filser
- German Center for Neurodegeneratione Diseases (DZNE), Department for Translational Brain Research, Feodor-Lynen-Strasse 23, Munich 81377, Germany
| | - Aayush Gupta
- Center for Neuropathology and Prion Research, Department for Translationsal Brain Research, Ludwig Maximilian University, Feodor-Lynen-Strassee 23, Munich 81377, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich, (CiPSM) and Department of Pharmacy-Center for Drug Research, Ludwig Maximilian University, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich, (CiPSM) and Department of Pharmacy-Center for Drug Research, Ludwig Maximilian University, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Jochen Herms
- 1] German Center for Neurodegeneratione Diseases (DZNE), Department for Translational Brain Research, Feodor-Lynen-Strasse 23, Munich 81377, Germany [2] Munich Cluster of Systems Neurology (SyNergy), Ludwig Maximilian University, Feodor-Lynen-Strasse 23, Munich 81377, Germany
| |
Collapse
|
17
|
Sakamoto M, Kageyama R, Imayoshi I. The functional significance of newly born neurons integrated into olfactory bulb circuits. Front Neurosci 2014; 8:121. [PMID: 24904263 PMCID: PMC4033306 DOI: 10.3389/fnins.2014.00121] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/06/2014] [Indexed: 12/27/2022] Open
Abstract
The olfactory bulb (OB) is the first central processing center for olfactory information connecting with higher areas in the brain, and this neuronal circuitry mediates a variety of odor-evoked behavioral responses. In the adult mammalian brain, continuous neurogenesis occurs in two restricted regions, the subventricular zone (SVZ) of the lateral ventricle and the hippocampal dentate gyrus. New neurons born in the SVZ migrate through the rostral migratory stream and are integrated into the neuronal circuits of the OB throughout life. The significance of this continuous supply of new neurons in the OB has been implicated in plasticity and memory regulation. Two decades of huge investigation in adult neurogenesis revealed the biological importance of integration of new neurons into the olfactory circuits. In this review, we highlight the recent findings about the physiological functions of newly generated neurons in rodent OB circuits and then discuss the contribution of neurogenesis in the brain function. Finally, we introduce cutting edge technologies to monitor and manipulate the activity of new neurons.
Collapse
Affiliation(s)
- Masayuki Sakamoto
- Institute for Virus Research, Kyoto University Kyoto, Japan ; Kyoto University Graduate School of Biostudies Kyoto, Japan
| | - Ryoichiro Kageyama
- Institute for Virus Research, Kyoto University Kyoto, Japan ; World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto University Kyoto, Japan ; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology Kyoto, Japan
| | - Itaru Imayoshi
- Institute for Virus Research, Kyoto University Kyoto, Japan ; World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto University Kyoto, Japan ; The Hakubi Center, Kyoto University Kyoto, Japan ; Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology Kyoto, Japan
| |
Collapse
|