1
|
Jin Y, Zhao L, Zhang Y, Chen T, Shi H, Sun H, Ding S, Chen S, Cao H, Zhang G, Li Q, Gao J, Xiao M, Sheng C. BIN1 deficiency enhances ULK3-dependent autophagic flux and reduces dendritic size in mouse hippocampal neurons. Autophagy 2025; 21:223-242. [PMID: 39171951 DOI: 10.1080/15548627.2024.2393932] [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: 07/14/2023] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024] Open
Abstract
Genome-wide association studies identified variants around the BIN1 (bridging integrator 1) gene locus as prominent risk factors for late-onset Alzheimer disease. In the present study, we decreased the expression of BIN1 in mouse hippocampal neurons to investigate its neuronal function. Bin1 knockdown via RNAi reduced the dendritic arbor size in primary cultured hippocampal neurons as well as in mature Cornu Ammonis 1 excitatory neurons. The AAV-mediated Bin1 RNAi knockdown also generated a significant regional volume loss around the injection sites at the organ level, as revealed by 7-Tesla structural magnetic resonance imaging, and an impaired spatial reference memory performance in the Barnes maze test. Unexpectedly, Bin1 knockdown led to concurrent activation of both macroautophagy/autophagy and MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1). Autophagy inhibition with the lysosome inhibitor chloroquine effectively mitigated the Bin1 knockdown-induced dendritic regression. The subsequent molecular studydemonstrated that increased expression of ULK3 (unc-51 like kinase 3), which is MTOR-insensitive, supported autophagosome formation in BIN1 deficiency. Reducing ULK3 activity with SU6668, a receptor tyrosine kinase inhibitor, or decreasing neuronal ULK3 expression through AAV-mediated RNAi, significantly attenuated Bin1 knockdown-induced hippocampal volume loss and spatial memory decline. In Alzheimer disease patients, the major neuronal isoform of BIN1 is specifically reduced. Our work suggests this reduction is probably an important molecular event that increases the autophagy level, which might subsequently promote brain atrophy and cognitive impairment through reducing dendritic structures, and ULK3 is a potential interventional target for relieving these detrimental effects.Abbreviations: AV: adeno-associated virus; Aβ: amyloid-β; ACTB: actin, beta; AD: Alzheimer disease; Aduk: Another Drosophila Unc-51-like kinase; AKT1: thymoma viral proto-oncogene 1; AMPK: AMP-activated protein kinase; AP: autophagosome; BafA1: bafilomycin A1; BDNF: brain derived neurotrophic factor; BIN1: bridging integrator 1; BIN1-iso1: BIN1, isoform 1; CA1: cornu Ammonis 1; CA3: cornu Ammonis 3; CLAP: clathrin and adapter binding; CQ: chloroquine; DMEM: Dulbecco's modified Eagle medium; EGFP: enhanced green fluorescent protein; GWAS: genome-wide association study; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MRI: magnetic resonance imaging; MTOR; mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; PET: positron emission tomography; qRT-PCR: real-time quantitative reverse transcription PCR; ROS: reactive oxygen species; RPS6KB1: ribosomal protein S6 kinase B1; TFEB: transcription factor EB; ULK1: unc-51 like kinase 1; ULK3: unc-51 like kinase 3.
Collapse
Affiliation(s)
- Yuxi Jin
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lin Zhao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yanli Zhang
- Department of Psychiatry, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Tingzhen Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huili Shi
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Huaiqing Sun
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shixin Ding
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Sijia Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Haifeng Cao
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Guannan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chengyu Sheng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Meftah S, Cavallini A, Murray TK, Jankowski L, Bose S, Ashby MC, Brown JT, Witton J. Synaptic alterations associated with disrupted sensory encoding in a mouse model of tauopathy. Brain Commun 2024; 6:fcae134. [PMID: 38712321 PMCID: PMC11073755 DOI: 10.1093/braincomms/fcae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 02/09/2024] [Accepted: 04/11/2024] [Indexed: 05/08/2024] Open
Abstract
Synapse loss is currently the best biological correlate of cognitive decline in Alzheimer's disease and other tauopathies. Synapses seem to be highly vulnerable to tau-mediated disruption in neurodegenerative tauopathies. However, it is unclear how and when this leads to alterations in function related to the progression of tauopathy and neurodegeneration. We used the well-characterized rTg4510 mouse model of tauopathy at 5-6 months and 7-8 months of age, respectively, to study the functional impact of cortical synapse loss. The earlier age was used as a model of prodromal tauopathy, with the later age corresponding to more advanced tau pathology and presumed progression of neurodegeneration. Analysis of synaptic protein expression in the somatosensory cortex showed significant reductions in synaptic proteins and NMDA and AMPA receptor subunit expression in rTg4510 mice. Surprisingly, in vitro whole-cell patch clamp electrophysiology from putative pyramidal neurons in layer 2/3 of the somatosensory cortex suggested no functional alterations in layer 4 to layer 2/3 synaptic transmission at 5-6 months. From these same neurons, however, there were alterations in dendritic structure, with increased branching proximal to the soma in rTg4510 neurons. Therefore, in vivo whole-cell patch clamp recordings were utilized to investigate synaptic function and integration in putative pyramidal neurons in layer 2/3 of the somatosensory cortex. These recordings revealed a significant increase in the peak response to synaptically driven sensory stimulation-evoked activity and a loss of temporal fidelity of the evoked signal to the input stimulus in rTg4510 neurons. Together, these data suggest that loss of synapses, changes in receptor expression and dendritic restructuring may lead to alterations in synaptic integration at a network level. Understanding these compensatory processes could identify targets to help delay symptomatic onset of dementia.
Collapse
Affiliation(s)
- Soraya Meftah
- Faculty of Health and Life Sciences, Department of Clinical and Biomedical Science, University of Exeter, Exeter, EX1 2LU, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Annalisa Cavallini
- Erl Wood Manor, Eli Lilly Pharmaceuticals, Windlesham, Surrey, GU20 6PH, UK
| | - Tracey K Murray
- Erl Wood Manor, Eli Lilly Pharmaceuticals, Windlesham, Surrey, GU20 6PH, UK
| | - Lukasz Jankowski
- Erl Wood Manor, Eli Lilly Pharmaceuticals, Windlesham, Surrey, GU20 6PH, UK
| | - Suchira Bose
- Erl Wood Manor, Eli Lilly Pharmaceuticals, Windlesham, Surrey, GU20 6PH, UK
| | - Michael C Ashby
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Jonathan T Brown
- Faculty of Health and Life Sciences, Department of Clinical and Biomedical Science, University of Exeter, Exeter, EX1 2LU, UK
| | - Jonathan Witton
- Faculty of Health and Life Sciences, Department of Clinical and Biomedical Science, University of Exeter, Exeter, EX1 2LU, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
3
|
Moustafa M, Mousa MH, Saad MS, Basha T, Elbasiouny SM. Bifurcation analysis of motoneuronal excitability mechanisms under normal and ALS conditions. Front Cell Neurosci 2023; 17:1093199. [PMID: 36874210 PMCID: PMC9978418 DOI: 10.3389/fncel.2023.1093199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/25/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Bifurcation analysis allows the examination of steady-state, non-linear dynamics of neurons and their effects on cell firing, yet its usage in neuroscience is limited to single-compartment models of highly reduced states. This is primarily due to the difficulty in developing high-fidelity neuronal models with 3D anatomy and multiple ion channels in XPPAUT, the primary bifurcation analysis software in neuroscience. Methods To facilitate bifurcation analysis of high-fidelity neuronal models under normal and disease conditions, we developed a multi-compartment model of a spinal motoneuron (MN) in XPPAUT and verified its firing accuracy against its original experimental data and against an anatomically detailed cell model that incorporates known MN non-linear firing mechanisms. We used the new model in XPPAUT to study the effects of somatic and dendritic ion channels on the MN bifurcation diagram under normal conditions and after amyotrophic lateral sclerosis (ALS) cellular changes. Results Our results show that somatic small-conductance Ca2+-activated K (SK) channels and dendritic L-type Ca2+ channels have the strongest effects on the bifurcation diagram of MNs under normal conditions. Specifically, somatic SK channels extend the limit cycles and generate a subcritical Hopf bifurcation node in the V-I bifurcation diagram of the MN to replace a supercritical node Hopf node, whereas L-type Ca2+ channels shift the limit cycles to negative currents. In ALS, our results show that dendritic enlargement has opposing effects on MN excitability, has a greater overall impact than somatic enlargement, and dendritic overbranching offsets the dendritic enlargement hyperexcitability effects. Discussion Together, the new multi-compartment model developed in XPPAUT facilitates studying neuronal excitability in health and disease using bifurcation analysis.
Collapse
Affiliation(s)
- Muhammad Moustafa
- Department of Systems and Biomedical Engineering, Faculty of Engineering, Cairo University, Giza, Egypt
| | - Mohamed H. Mousa
- Department of Biomedical, Industrial, and Human Factors Engineering, College of Engineering and Computer Science, Wright State University, Dayton, OH, United States
| | - Mohamed S. Saad
- Department of Electrical Power Engineering, Faculty of Engineering, Cairo University, Giza, Egypt
| | - Tamer Basha
- Department of Systems and Biomedical Engineering, Faculty of Engineering, Cairo University, Giza, Egypt
| | - Sherif M. Elbasiouny
- Department of Biomedical, Industrial, and Human Factors Engineering, College of Engineering and Computer Science, Wright State University, Dayton, OH, United States
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, United States
| |
Collapse
|
4
|
Weerasinghe-Mudiyanselage PDE, Ang MJ, Kang S, Kim JS, Moon C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:3349. [PMID: 35328770 PMCID: PMC8955928 DOI: 10.3390/ijms23063349] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/10/2022] Open
Abstract
Neuroplasticity is the capacity of neural networks in the brain to alter through development and rearrangement. It can be classified as structural and functional plasticity. The hippocampus is more susceptible to neuroplasticity as compared to other brain regions. Structural modifications in the hippocampus underpin several neurodegenerative diseases that exhibit cognitive and emotional dysregulation. This article reviews the findings of several preclinical and clinical studies about the role of structural plasticity in the hippocampus in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In this study, literature was surveyed using Google Scholar, PubMed, Web of Science, and Scopus, to review the mechanisms that underlie the alterations in the structural plasticity of the hippocampus in neurodegenerative diseases. This review summarizes the role of structural plasticity in the hippocampus for the etiopathogenesis of neurodegenerative diseases and identifies the current focus and gaps in knowledge about hippocampal dysfunctions. Ultimately, this information will be useful to propel future mechanistic and therapeutic research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Poornima D. E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
- College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| |
Collapse
|
5
|
Magliaro C, Callara AL, Vanello N, Ahluwalia A. Gotta Trace 'em All: A Mini-Review on Tools and Procedures for Segmenting Single Neurons Toward Deciphering the Structural Connectome. Front Bioeng Biotechnol 2019; 7:202. [PMID: 31555642 PMCID: PMC6727034 DOI: 10.3389/fbioe.2019.00202] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/06/2019] [Indexed: 12/12/2022] Open
Abstract
Decoding the morphology and physical connections of all the neurons populating a brain is necessary for predicting and studying the relationships between its form and function, as well as for documenting structural abnormalities in neuropathies. Digitizing a complete and high-fidelity map of the mammalian brain at the micro-scale will allow neuroscientists to understand disease, consciousness, and ultimately what it is that makes us humans. The critical obstacle for reaching this goal is the lack of robust and accurate tools able to deal with 3D datasets representing dense-packed cells in their native arrangement within the brain. This obliges neuroscientist to manually identify the neurons populating an acquired digital image stack, a notably time-consuming procedure prone to human bias. Here we review the automatic and semi-automatic algorithms and software for neuron segmentation available in the literature, as well as the metrics purposely designed for their validation, highlighting their strengths and limitations. In this direction, we also briefly introduce the recent advances in tissue clarification that enable significant improvements in both optical access of neural tissue and image stack quality, and which could enable more efficient segmentation approaches. Finally, we discuss new methods and tools for processing tissues and acquiring images at sub-cellular scales, which will require new robust algorithms for identifying neurons and their sub-structures (e.g., spines, thin neurites). This will lead to a more detailed structural map of the brain, taking twenty-first century cellular neuroscience to the next level, i.e., the Structural Connectome.
Collapse
Affiliation(s)
- Chiara Magliaro
- Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | | | - Nicola Vanello
- Research Center "E. Piaggio", University of Pisa, Pisa, Italy.,Dipartimento di Ingegneria dell'Informazione, University of Pisa, Pisa, Italy
| | - Arti Ahluwalia
- Research Center "E. Piaggio", University of Pisa, Pisa, Italy.,Dipartimento di Ingegneria dell'Informazione, University of Pisa, Pisa, Italy
| |
Collapse
|
6
|
Jin G, Zhu L, Liu P, Xu Q, Qi Y, Zhou X, Xu J, Ji X, Chi T, Zou L. Xanthoceraside prevented synaptic loss and reversed learning-memory deficits in APP/PS1 transgenic mice. J Physiol Sci 2019; 69:477-488. [PMID: 30767122 PMCID: PMC10718037 DOI: 10.1007/s12576-019-00664-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/22/2019] [Indexed: 01/02/2023]
Abstract
Xanthoceraside, a novel triterpenoid saponin, has been found to attenuate learning and memory impairments in AD animal models. However, whether xanthoceraside has a positive effect on synaptic morphology remains unclear. Herein, we evaluated the effects of xanthoceraside on learning and memory impairments and the abnormalities of synaptic structure in APP/PS1 transgenic mice. The behavioral experiments demonstrated that xanthoceraside attenuated the imaginal memory and spatial learning impairments, and improved social interaction. Transmission electron microscopy and Golgi staining showed that xanthoceraside ameliorated synapse morphology abnormalities and dendritic spine density deficits, respectively. Western-blot analysis identified that xanthoceraside increased the expression of SYP and PSD95, activated BDNF/TrkB/MAPK/ERK and PI3K/Akt signaling pathways, meanwhile decreased the expression of RhoA, ROCK and Snk, increased the levels of SPAR, and activated the BDNF/TrkB/cofilin signaling pathway. Taken together, our study indicated that xanthoceraside improved cognitive function and protected both synaptic morphology and dendritic spine in APP/PS1 transgenic mice, which might be related in part to its activation in the BDNF/TrkB pathway.
Collapse
Affiliation(s)
- Ge Jin
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
- Shenyang Medical College, Shenyang, 110034, People's Republic of China
| | - Lin Zhu
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
- Shenyang Medical College, Shenyang, 110034, People's Republic of China
| | - Peng Liu
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Qian Xu
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Yue Qi
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xiaoyu Zhou
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Jikai Xu
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xuefei Ji
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Tianyan Chi
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China
| | - Libo Zou
- Department of Pharmacology, Life Science and Biopharmaceutics School, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
7
|
Van Houcke J, Geeraerts E, Vanhunsel S, Beckers A, Noterdaeme L, Christiaens M, Bollaerts I, De Groef L, Moons L. Extensive growth is followed by neurodegenerative pathology in the continuously expanding adult zebrafish retina. Biogerontology 2019. [PMID: 30382466 DOI: 10.1007/s10522-018-9780-6/figures/10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
The development of effective treatments for age-related neurodegenerative diseases remains one of the biggest medical challenges today, underscoring the high need for suitable animal model systems to improve our understanding of aging and age-associated neuropathology. Zebrafish have become an indispensable complementary model organism in gerontology research, yet their growth-control properties significantly differ from those in mammals. Here, we took advantage of the clearly defined and highly conserved structure of the fish retina to study the relationship between the processes of growth and aging in the adult zebrafish central nervous system (CNS). Detailed morphological measurements reveal an early phase of extensive retinal growth, where both the addition of new cells and stretching of existent tissue drive the increase in retinal surface. Thereafter, and coinciding with a significant decline in retinal growth rate, a neurodegenerative phenotype becomes apparent,-characterized by a loss of synaptic integrity, an age-related decrease in cell density and the onset of cellular senescence. Altogether, these findings support the adult zebrafish retina as a valuable model for gerontology research and CNS disease modeling and will hopefully stimulate further research into the mechanisms of aging and age-related pathology.
Collapse
Affiliation(s)
- Jessie Van Houcke
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Emiel Geeraerts
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Sophie Vanhunsel
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - An Beckers
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Lut Noterdaeme
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Marijke Christiaens
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Ilse Bollaerts
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium.
| |
Collapse
|
8
|
Van Houcke J, Geeraerts E, Vanhunsel S, Beckers A, Noterdaeme L, Christiaens M, Bollaerts I, De Groef L, Moons L. Extensive growth is followed by neurodegenerative pathology in the continuously expanding adult zebrafish retina. Biogerontology 2018; 20:109-125. [PMID: 30382466 DOI: 10.1007/s10522-018-9780-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/26/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Jessie Van Houcke
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Emiel Geeraerts
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Sophie Vanhunsel
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - An Beckers
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Lut Noterdaeme
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Marijke Christiaens
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Ilse Bollaerts
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Naamsestraat 61, Box 2464, 3000, Leuven, Belgium.
| |
Collapse
|
9
|
Butterfield DA. Perspectives on Oxidative Stress in Alzheimer’s Disease and Predictions of Future Research Emphases. J Alzheimers Dis 2018; 64:S469-S479. [DOI: 10.3233/jad-179912] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
10
|
Transgenic autoinhibition of p21-activated kinase exacerbates synaptic impairments and fronto-dependent behavioral deficits in an animal model of Alzheimer's disease. Aging (Albany NY) 2018; 9:1386-1403. [PMID: 28522792 PMCID: PMC5472739 DOI: 10.18632/aging.101239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/11/2017] [Indexed: 11/28/2022]
Abstract
Defects in p21-activated kinase (PAK) lead to dendritic spine abnormalities and are sufficient to cause cognition impairment. The decrease in PAK in the brain of Alzheimer's disease (AD) patients is suspected to underlie synaptic and dendritic disturbances associated with its clinical expression, particularly with symptoms related to frontal cortex dysfunction. To investigate the role of PAK combined with Aβ and tau pathologies (3xTg-AD mice) in the frontal cortex, we generated a transgenic model of AD with a deficit in PAK activity (3xTg-AD-dnPAK mice). PAK inactivation had no effect on Aβ40 and Aβ42 levels, but increased the phosphorylation ratio of tau in detergent-insoluble protein fractions in the frontal cortex of 18-month-old heterozygous 3xTg-AD mice. Morphometric analyses of layer II/III pyramidal neurons in the frontal cortex showed that 3xTg-AD-dnPAK neurons exhibited significant dendritic attrition, lower spine density and longer spines compared to NonTg and 3xTg-AD mice. Finally, behavioral assessments revealed that 3xTg-AD-dnPAK mice exhibited pronounced anxious traits and disturbances in social behaviors, reminiscent of fronto-dependent symptoms observed in AD. Our results substantiate a critical role for PAK in the genesis of neuronal abnormalities in the frontal cortex underlying the emergence of psychiatric-like symptoms in AD.
Collapse
|
11
|
Herzog JJ, Deshpande M, Shapiro L, Rodal AA, Paradis S. TDP-43 misexpression causes defects in dendritic growth. Sci Rep 2017; 7:15656. [PMID: 29142232 PMCID: PMC5688077 DOI: 10.1038/s41598-017-15914-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/03/2017] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) share overlapping genetic causes and disease symptoms, and are linked neuropathologically by the RNA binding protein TDP-43 (TAR DNA binding protein-43 kDa). TDP-43 regulates RNA metabolism, trafficking, and localization of thousands of target genes. However, the cellular and molecular mechanisms by which dysfunction of TDP-43 contributes to disease pathogenesis and progression remain unclear. Severe changes in the structure of neuronal dendritic arbors disrupt proper circuit connectivity, which in turn could contribute to neurodegenerative disease. Although aberrant dendritic morphology has been reported in non-TDP-43 mouse models of ALS and in human ALS patients, this phenotype is largely unexplored with regards to TDP-43. Here we have employed a primary rodent neuronal culture model to study the cellular effects of TDP-43 dysfunction in hippocampal and cortical neurons. We show that manipulation of TDP-43 expression levels causes significant defects in dendritic branching and outgrowth, without an immediate effect on cell viability. The effect on dendritic morphology is dependent on the RNA-binding ability of TDP-43. Thus, this model system will be useful in identifying pathways downstream of TDP-43 that mediate dendritic arborization, which may provide potential new avenues for therapeutic intervention in ALS/FTD.
Collapse
Affiliation(s)
- Josiah J Herzog
- Department of Biology, Volen Center for Complex Systems, and National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts, 02454, USA
| | - Mugdha Deshpande
- Department of Biology, Volen Center for Complex Systems, and National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts, 02454, USA
| | - Leah Shapiro
- Department of Biology, Volen Center for Complex Systems, and National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts, 02454, USA
| | - Avital A Rodal
- Department of Biology, Volen Center for Complex Systems, and National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts, 02454, USA
| | - Suzanne Paradis
- Department of Biology, Volen Center for Complex Systems, and National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts, 02454, USA.
| |
Collapse
|
12
|
Hannan MA, Mohibbullah M, Hong YK, Moon IS. Proteomic Analysis of the Neurotrophic Effect of Gelidium amansii in Primary Cultured Neurons. J Med Food 2017; 20:279-287. [DOI: 10.1089/jmf.2016.3848] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Md. Abdul Hannan
- Department of Biotechnology, Pukyong National University, Busan, Republic of Korea
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Mohibbullah
- Department of Biotechnology, Pukyong National University, Busan, Republic of Korea
| | - Yong-Ki Hong
- Department of Biotechnology, Pukyong National University, Busan, Republic of Korea
| | - Il Soo Moon
- Department of Anatomy, Dongguk University Graduate School of Medicine, Gyeongju, Republic of Korea
| |
Collapse
|
13
|
Wnt5a is essential for hippocampal dendritic maintenance and spatial learning and memory in adult mice. Proc Natl Acad Sci U S A 2017; 114:E619-E628. [PMID: 28069946 PMCID: PMC5278440 DOI: 10.1073/pnas.1615792114] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Stability of neuronal connectivity is critical for brain functions, and morphological perturbations are associated with neurodegenerative disorders. However, how neuronal morphology is maintained in the adult brain remains poorly understood. Here, we identify Wnt5a, a member of the Wnt family of secreted morphogens, as an essential factor in maintaining dendritic architecture in the adult hippocampus and for related cognitive functions in mice. Wnt5a expression in hippocampal neurons begins postnatally, and its deletion attenuated CaMKII and Rac1 activity, reduced GluN1 glutamate receptor expression, and impaired synaptic plasticity and spatial learning and memory in 3-mo-old mice. With increased age, Wnt5a loss caused progressive attrition of dendrite arbors and spines in Cornu Ammonis (CA)1 pyramidal neurons and exacerbated behavioral defects. Wnt5a functions cell-autonomously to maintain CA1 dendrites, and exogenous Wnt5a expression corrected structural anomalies even at late-adult stages. These findings reveal a maintenance factor in the adult brain, and highlight a trophic pathway that can be targeted to ameliorate dendrite loss in pathological conditions.
Collapse
|
14
|
Fuzzy-Logic Based Detection and Characterization of Junctions and Terminations in Fluorescence Microscopy Images of Neurons. Neuroinformatics 2016; 14:201-19. [PMID: 26701809 PMCID: PMC4823367 DOI: 10.1007/s12021-015-9287-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Digital reconstruction of neuronal cell morphology is an important step toward understanding the functionality of neuronal networks. Neurons are tree-like structures whose description depends critically on the junctions and terminations, collectively called critical points, making the correct localization and identification of these points a crucial task in the reconstruction process. Here we present a fully automatic method for the integrated detection and characterization of both types of critical points in fluorescence microscopy images of neurons. In view of the majority of our current studies, which are based on cultured neurons, we describe and evaluate the method for application to two-dimensional (2D) images. The method relies on directional filtering and angular profile analysis to extract essential features about the main streamlines at any location in an image, and employs fuzzy logic with carefully designed rules to reason about the feature values in order to make well-informed decisions about the presence of a critical point and its type. Experiments on simulated as well as real images of neurons demonstrate the detection performance of our method. A comparison with the output of two existing neuron reconstruction methods reveals that our method achieves substantially higher detection rates and could provide beneficial information to the reconstruction process.
Collapse
|
15
|
Raffini F, Fruciano C, Franchini P, Meyer A. Towards understanding the genetic basis of mouth asymmetry in the scale-eating cichlidPerissodus microlepis. Mol Ecol 2016; 26:77-91. [DOI: 10.1111/mec.13699] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Francesca Raffini
- Lehrstuhl für Zoologie und Evolutionsbiologie; Department of Biology; University of Konstanz; Universitätsstrasse 10 78464 Konstanz Germany
- International Max Planck Research School (IMPRS) for Organismal Biology; Max-Planck-Institut für Ornithologie; Am Obstberg 1 78315 Radolfzell Germany
| | - Carmelo Fruciano
- Lehrstuhl für Zoologie und Evolutionsbiologie; Department of Biology; University of Konstanz; Universitätsstrasse 10 78464 Konstanz Germany
| | - Paolo Franchini
- Lehrstuhl für Zoologie und Evolutionsbiologie; Department of Biology; University of Konstanz; Universitätsstrasse 10 78464 Konstanz Germany
| | - Axel Meyer
- Lehrstuhl für Zoologie und Evolutionsbiologie; Department of Biology; University of Konstanz; Universitätsstrasse 10 78464 Konstanz Germany
- International Max Planck Research School (IMPRS) for Organismal Biology; Max-Planck-Institut für Ornithologie; Am Obstberg 1 78315 Radolfzell Germany
| |
Collapse
|
16
|
Jones BW, Pfeiffer RL, Ferrell WD, Watt CB, Marmor M, Marc RE. Retinal remodeling in human retinitis pigmentosa. Exp Eye Res 2016; 150:149-65. [PMID: 27020758 DOI: 10.1016/j.exer.2016.03.018] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/23/2016] [Accepted: 03/18/2016] [Indexed: 12/11/2022]
Abstract
Retinitis Pigmentosa (RP) in the human is a progressive, currently irreversible neural degenerative disease usually caused by gene defects that disrupt the function or architecture of the photoreceptors. While RP can initially be a disease of photoreceptors, there is increasing evidence that the inner retina becomes progressively disorganized as the outer retina degenerates. These alterations have been extensively described in animal models, but remodeling in humans has not been as well characterized. This study, using computational molecular phenotyping (CMP) seeks to advance our understanding of the retinal remodeling process in humans. We describe cone mediated preservation of overall topology, retinal reprogramming in the earliest stages of the disease in retinal bipolar cells, and alterations in both small molecule and protein signatures of neurons and glia. Furthermore, while Müller glia appear to be some of the last cells left in the degenerate retina, they are also one of the first cell classes in the neural retina to respond to stress which may reveal mechanisms related to remodeling and cell death in other retinal cell classes. Also fundamentally important is the finding that retinal network topologies are altered. Our results suggest interventions that presume substantial preservation of the neural retina will likely fail in late stages of the disease. Even early intervention offers no guarantee that the interventions will be immune to progressive remodeling. Fundamental work in the biology and mechanisms of disease progression are needed to support vision rescue strategies.
Collapse
Affiliation(s)
- B W Jones
- Dept. Ophthalmology, Moran Eye Center, University of Utah, USA.
| | - R L Pfeiffer
- Dept. Ophthalmology, Moran Eye Center, University of Utah, USA
| | - W D Ferrell
- Dept. Ophthalmology, Moran Eye Center, University of Utah, USA
| | - C B Watt
- Dept. Ophthalmology, Moran Eye Center, University of Utah, USA
| | - M Marmor
- Dept. Ophthalmology, Stanford University, USA
| | - R E Marc
- Dept. Ophthalmology, Moran Eye Center, University of Utah, USA
| |
Collapse
|
17
|
Golovyashkina N, Penazzi L, Ballatore C, Smith AB, Bakota L, Brandt R. Region-specific dendritic simplification induced by Aβ, mediated by tau via dysregulation of microtubule dynamics: a mechanistic distinct event from other neurodegenerative processes. Mol Neurodegener 2015; 10:60. [PMID: 26541821 PMCID: PMC4634596 DOI: 10.1186/s13024-015-0049-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 09/28/2015] [Indexed: 12/30/2022] Open
Abstract
Background Dendritic simplification, a key feature of the neurodegenerative triad of Alzheimer’s disease (AD) in addition to spine changes and neuron loss, occurs in a region-specific manner. However, it is unknown how changes in dendritic complexity are mediated and how they relate to spine changes and neuron loss. Results To investigate the mechanisms of dendritic simplification in an authentic CNS environment we employed an ex vivo model, based on targeted expression of enhanced green fluorescent protein (EGFP)-tagged constructs in organotypic hippocampal slices of mice. Algorithm-based 3D reconstruction of whole neuron morphology in different hippocampal regions was performed on slices from APPSDL-transgenic and control animals. We demonstrate that induction of dendritic simplification requires the combined action of amyloid beta (Aβ) and human tau. Simplification is restricted to principal neurons of the CA1 region, recapitulating the region specificity in AD patients, and occurs at sites of Schaffer collateral input. We report that γ-secretase inhibition and treatment with the NMDA-receptor antagonist, CPP, counteract dendritic simplification. The microtubule-stabilizing drug epothilone D (EpoD) induces simplification in control cultures per se. Similar morphological changes were induced by a phosphoblocking tau construct, which also increases microtubule stability. In fact, low nanomolar concentrations of naturally secreted Aβ decreased phosphorylation at S262 in a cellular model, a site which is known to directly modulate tau-microtubule interactions. Conclusions The data provide evidence that dendritic simplification is mechanistically distinct from other neurodegenerative events and involves microtubule stabilization by dendritic tau, which becomes dephosphorylated at certain sites. They imply that treatments leading to an overall decrease of tau phosphorylation might have a negative impact on neuronal connectivity.
Collapse
Affiliation(s)
- Nataliya Golovyashkina
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany.
| | - Lorène Penazzi
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany.
| | - Carlo Ballatore
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, 19014, USA. .,Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Amos B Smith
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, 19014, USA.
| | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany.
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Barbarastrasse 11, 49076, Osnabrück, Germany.
| |
Collapse
|
18
|
Braak H, Del Tredici K. The preclinical phase of the pathological process underlying sporadic Alzheimer’s disease. Brain 2015; 138:2814-33. [DOI: 10.1093/brain/awv236] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/21/2015] [Indexed: 12/13/2022] Open
|
19
|
Abstract
Over the past 35 years or so, PET brain imaging has allowed powerful and unique insights into brain function under normal conditions and in disease states. Initially, as PET instrumentation continued to develop, studies were focused on brain perfusion and glucose metabolism. This permitted refinement of brain imaging for important, non-oncologic clinical indications. The ability of PET to not only provide spatial localization of metabolic changes but also to accurately and consistently quantify their distribution proved valuable for applications in the clinical setting. Specifically, glucose metabolism brain imaging using (F-18) fluorodeoxyglucose continues to be invaluable for evaluating patients with intractable seizures for identifying seizure foci and operative planning. Cerebral glucose metabolism also contributes to diagnosis of neurodegenerative diseases that cause dementia. Alzheimer disease, dementia with Lewy bodies, and the several variants of frontotemporal lobar degeneration have differing typical patterns of hypometabolism. In Alzheimer disease, hypometabolism has furthermore been associated with poorer cognitive performance and ensuing cognitive and functional decline. As the field of radiochemistry evolved, novel radioligands including radiolabeled flumazenil, dopamine transporter ligands, nicotine receptor ligands, and others have allowed for further understanding of molecular changes in the brain associated with various diseases. Recently, PET brain imaging reached another milestone with the approval of (F-18) florbetapir imaging by the United States Federal Drug Administration for detection of amyloid plaque accumulation in brain, the major histopathologic hallmark of Alzheimer disease, and efforts have been made to define the clinical role of this imaging agent in the setting of the currently limited treatment options. Hopefully, this represents the first of many new radiopharmaceuticals that would allow improved diagnostic and prognostic information in these and other clinical applications, including Parkinson disease and traumatic brain injury.
Collapse
Affiliation(s)
- Ilya Nasrallah
- Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
20
|
Cochran JN, Hall AM, Roberson ED. The dendritic hypothesis for Alzheimer's disease pathophysiology. Brain Res Bull 2014; 103:18-28. [PMID: 24333192 PMCID: PMC3989444 DOI: 10.1016/j.brainresbull.2013.12.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 11/28/2013] [Accepted: 12/02/2013] [Indexed: 01/02/2023]
Abstract
Converging evidence indicates that processes occurring in and around neuronal dendrites are central to the pathogenesis of Alzheimer's disease. These data support the concept of a "dendritic hypothesis" of AD, closely related to the existing synaptic hypothesis. Here we detail dendritic neuropathology in the disease and examine how Aβ, tau, and AD genetic risk factors affect dendritic structure and function. Finally, we consider potential mechanisms by which these key drivers could affect dendritic integrity and disease progression. These dendritic mechanisms serve as a framework for therapeutic target identification and for efforts to develop disease-modifying therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- J Nicholas Cochran
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Alicia M Hall
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
21
|
Fujiwara H, Kimura J, Sakamoto M, Yokosuka A, Mimaki Y, Murata K, Yamaguchi K, Ohizumi Y. Nobiletin, a flavone from Citrus depressa, induces gene expression and increases the protein level and activity of neprilysin in SK-N-SH cells. Can J Physiol Pharmacol 2014; 92:351-5. [PMID: 24784468 DOI: 10.1139/cjpp-2013-0440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neprilysin (NEP) is one of the candidate amyloid β protein (Aβ) degrading enzymes affecting brain Aβ clearance. This enzyme declines in the brain with age, which leads to the increased Aβ deposition in Alzheimer's disease (AD). Pharmacological activation of NEP during the aging process, therefore, represents a potential strategy to prevent the development of AD. To examine the influence of nobiletin on neprilysin activity, we measured cellular NEP activity in SK-N-SH cells. Moreover, NEP expression was examined by using reverse transcription - polymerase chain reaction and Western blotting. Measurement of cellular NEP activity showed that nobiletin stimulated this in a dose- and time-dependent manner in SK-N-SH cells. Moreover, nobiletin increased the expression of NEP mRNA, and then the levels of NEP protein, also in a dose- and time-dependent manner. Our findings showed that nobiletin promoted NEP gene and protein expression, resulting in enhancement of cellular NEP activity in SK-N-SH cells. This compound could be a novel Aβ-degrading compound for use in the development of disease-modifying drugs to prevent and (or) cure AD.
Collapse
Affiliation(s)
- Hironori Fujiwara
- a Department of Anti-dementia Functional Food Development, Graduate School of Engineering, Tohoku University, Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Crowe SE, Ellis-Davies GCR. In vivo characterization of a bigenic fluorescent mouse model of Alzheimer's disease with neurodegeneration. J Comp Neurol 2013; 521:2181-94. [PMID: 23348594 DOI: 10.1002/cne.23306] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/14/2012] [Accepted: 01/17/2013] [Indexed: 11/11/2022]
Abstract
The loss of cognitive function in Alzheimer's disease (AD) patients is strongly correlated with the loss of neurons in various regions of the brain. We have created a new fluorescent bigenic mouse model of AD by crossing "H-line" yellow fluorescent protein (YFP) mice with the 5xFAD mouse model, which we call the 5XY mouse model. The 5xFAD mouse has been shown to have significant loss of L5 pyramidal neurons by 12 months of age. These neurons are transgenically labeled with YFP in the 5XY mouse, which enable longitudinal imaging of structural changes. In the 5XY mice, we observed an appearance of axonal dystrophies, with two distinct morphologies in the early stages of the disease progression. Simple swelling dystrophies are transient in nature and are not directly associated with amyloid plaques. Rosette dystrophies are more complex structures that remained stable throughout all imaging sessions, and always surrounded an amyloid plaque. Plaque growth was followed over 4 weeks, and significant growth was seen between weekly imaging sessions. In addition to axonal dystrophy appearance and plaque growth, we were able to follow spine stability in 4-month old 5XY mice, which revealed no significant loss of spines. 5XY mice also showed a striking shrinkage of the neocortex at older ages (12-14 months). The 5XY mouse model may be a valuable tool for studying specific events in the degeneration of the neocortex, and may suggest new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Sarah E Crowe
- Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | |
Collapse
|
23
|
Couch BA, Kerrisk ME, Kaufman AC, Nygaard HB, Strittmatter SM, Koleske AJ. Delayed amyloid plaque deposition and behavioral deficits in outcrossed AβPP/PS1 mice. J Comp Neurol 2013; 521:1395-408. [PMID: 23047754 DOI: 10.1002/cne.23239] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 05/22/2012] [Accepted: 10/02/2012] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative dementia characterized by amyloid plaque accumulation, synapse/dendrite loss, and cognitive impairment. Transgenic mice expressing mutant forms of amyloid-β precursor protein (AβPP) and presenilin-1 (PS1) recapitulate several aspects of this disease and provide a useful model system for studying elements of AD progression. AβPP/PS1 mice have been previously shown to exhibit behavioral deficits and amyloid plaque deposition between 4-9 months of age. We crossed AβPP/PS1 animals with mice of a mixed genetic background (C57BL/6 × 129/SvJ) and investigated the development of AD-like features in the resulting outcrossed mice. The onset of memory-based behavioral impairment is delayed considerably in outcrossed AβPP/PS1 mice relative to inbred mice on a C57BL/6 background. While inbred AβPP/PS1 mice develop deficits in radial-arm water maze performance and novel object recognition as early as 8 months, outcrossed AβPP/PS1 mice do not display defects until 18 months. Within the forebrain, we find that inbred AβPP/PS1 mice have significantly higher amyloid plaque burden at 12 months than outcrossed AβPP/PS1 mice of the same age. Surprisingly, inbred AβPP/PS1 mice at 8 months have low plaque burden, suggesting that plaque burden alone cannot explain the accompanying behavioral deficits. Analysis of AβPP processing revealed that elevated levels of soluble Aβ correlate with the degree of behavioral impairment in both strains. Taken together, these findings suggest that animal behavior, amyloid plaque deposition, and AβPP processing are sensitive to genetic differences between mouse strains.
Collapse
Affiliation(s)
- Brian A Couch
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
| | | | | | | | | | | |
Collapse
|
24
|
Shahnawaz M, Sharoar MG, Shin SY, Park IS. Wild-type, Flemish, and Dutch amyloid-β
exhibit different cytotoxicities depending on Aβ
40 to Aβ
42 interaction time and concentration ratio. J Pept Sci 2013; 19:545-53. [DOI: 10.1002/psc.2531] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/03/2013] [Accepted: 05/26/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Md. Shahnawaz
- Department of Bio-materials Engineering; College of Medicine, Chosun University; Gwangju 501-759 Korea
- Medical School; The University of Texas Health Science Center at Houston; 6431 Fannin St Houston TX 77030 USA
| | - Md. Golam Sharoar
- Department of Bio-materials Engineering; College of Medicine, Chosun University; Gwangju 501-759 Korea
- Department of Genetic Engineering and Biotechnology; University of Rajshahi; Rajshahi 6205 Bangladesh
| | - Song Yub Shin
- Department of Bio-materials Engineering; College of Medicine, Chosun University; Gwangju 501-759 Korea
- Department of Cell and Molecular Biology; College of Medicine, Chosun University; Gwangju 501-759 Korea
| | - Il-Seon Park
- Department of Bio-materials Engineering; College of Medicine, Chosun University; Gwangju 501-759 Korea
- Department of Cell and Molecular Biology; College of Medicine, Chosun University; Gwangju 501-759 Korea
| |
Collapse
|
25
|
Abstract
In the developing brain, dendrite branches and dendritic spines form and turn over dynamically. By contrast, most dendrite arbors and dendritic spines in the adult brain are stable for months, years and possibly even decades. Emerging evidence reveals that dendritic spine and dendrite arbor stability have crucial roles in the correct functioning of the adult brain and that loss of stability is associated with psychiatric disorders and neurodegenerative diseases. Recent findings have provided insights into the molecular mechanisms that underlie long-term dendrite stabilization, how these mechanisms differ from those used to mediate structural plasticity and how they are disrupted in disease.
Collapse
|
26
|
Retinal ganglion cell dendritic degeneration in a mouse model of Alzheimer's disease. Neurobiol Aging 2013; 34:1799-806. [DOI: 10.1016/j.neurobiolaging.2013.01.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 01/11/2013] [Accepted: 01/11/2013] [Indexed: 12/30/2022]
|
27
|
Ando K, Brion JP, Stygelbout V, Suain V, Authelet M, Dedecker R, Chanut A, Lacor P, Lavaur J, Sazdovitch V, Rogaeva E, Potier MC, Duyckaerts C. Clathrin adaptor CALM/PICALM is associated with neurofibrillary tangles and is cleaved in Alzheimer's brains. Acta Neuropathol 2013; 125:861-78. [PMID: 23589030 DOI: 10.1007/s00401-013-1111-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 02/20/2013] [Accepted: 03/24/2013] [Indexed: 02/07/2023]
Abstract
PICALM, a clathrin adaptor protein, plays important roles in clathrin-mediated endocytosis in all cell types. Recently, genome-wide association studies identified single nucleotide polymorphisms in PICALM gene as genetic risk factors for late-onset Alzheimer disease (LOAD). We analysed by western blotting with several anti-PICALM antibodies the pattern of expression of PICALM in human brain extracts. We found that PICALM was abnormally cleaved in AD samples and that the level of the uncleaved 65-75 kDa full-length PICALM species was significantly decreased in AD brains. Cleavage of human PICALM after activation of endogenous calpain or caspase was demonstrated in vitro. Immunohistochemistry revealed that PICALM was associated in situ with neurofibrillary tangles, co-localising with conformationally abnormal and hyperphosphorylated tau in LOAD, familial AD and Down syndrome cases. PHF-tau proteins co-immunoprecipitated with PICALM. PICALM was highly expressed in microglia in LOAD. These observations suggest that PICALM is associated with the development of AD tau pathology. PICALM cleavage could contribute to endocytic dysfunction in AD.
Collapse
Affiliation(s)
- Kunie Ando
- Laboratoire de Neuropathologie Escourolle, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Spine pruning in 5xFAD mice starts on basal dendrites of layer 5 pyramidal neurons. Brain Struct Funct 2013; 219:571-80. [PMID: 23417057 DOI: 10.1007/s00429-013-0518-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 02/01/2013] [Indexed: 12/19/2022]
Abstract
Transgenic mice with Alzheimer's disease (AD) mutations have been widely used to model changes in neuronal structure and function. While there are clear gross structural changes in post-mortem brains of AD patients, most mouse models of AD do not recapitulate the considerable loss of neurons. Furthermore, possible connections between early subtle structural changes and the loss of neurons are difficult to study. In an attempt to start unraveling how neurons are affected during the early stages of what becomes full neurodegeneration, we crossed a mouse model of familial AD, which displays massive neocortical neurodegeneration (the 5xFAD mouse), with the fluorescent H-line YFP mouse. This novel bigenic mouse model of AD, which we have named the 5XY mouse, expresses YFP in principal neurons in the cortex such that even fine details of cells are clearly visible. Such bright fluorescence allowed us to use high-resolution confocal microscopy to quantify changes in spine density in the somatosensory cortex, prefrontal cortex, and hippocampus at 2, 4, and 6 months of age. A significant loss of spines on basal dendrites in the somatosensory and prefrontal cortices of 6-month-old 5XY female mice was found. There was no observed spine loss at 6 months of age on the oblique dendrites of the hippocampus in the same mice. These data suggest that spine loss is an early event in the degeneration of the neocortical neurons in 5xFAD mice and a likely contributor to the cognitive impairments reported previously in this AD mouse model.
Collapse
|
29
|
Yadav A, Gao YZ, Rodriguez A, Dickstein DL, Wearne SL, Luebke JI, Hof PR, Weaver CM. Morphologic evidence for spatially clustered spines in apical dendrites of monkey neocortical pyramidal cells. J Comp Neurol 2012; 520:2888-902. [PMID: 22315181 DOI: 10.1002/cne.23070] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The general organization of neocortical connectivity in rhesus monkey is relatively well understood. However, mounting evidence points to an organizing principle that involves clustered synapses at the level of individual dendrites. Several synaptic plasticity studies have reported cooperative interaction between neighboring synapses on a given dendritic branch, which may potentially induce synapse clusters. Additionally, theoretical models have predicted that such cooperativity is advantageous, in that it greatly enhances a neuron's computational repertoire. However, largely because of the lack of sufficient morphologic data, the existence of clustered synapses in neurons on a global scale has never been established. The majority of excitatory synapses are found within dendritic spines. In this study, we demonstrate that spine clusters do exist on pyramidal neurons by analyzing the three-dimensional locations of ∼40,000 spines on 280 apical dendritic branches in layer III of the rhesus monkey prefrontal cortex. By using clustering algorithms and Monte Carlo simulations, we quantify the probability that the observed extent of clustering does not occur randomly. This provides a measure that tests for spine clustering on a global scale, whenever high-resolution morphologic data are available. Here we demonstrate that spine clusters occur significantly more frequently than expected by pure chance and that spine clustering is concentrated in apical terminal branches. These findings indicate that spine clustering is driven by systematic biological processes. We also found that mushroom-shaped and stubby spines are predominant in clusters on dendritic segments that display prolific clustering, independently supporting a causal link between spine morphology and synaptic clustering.
Collapse
Affiliation(s)
- Aniruddha Yadav
- Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Gur RE, Gur RC. Gender differences in aging: cognition, emotions, and neuroimaging studies. DIALOGUES IN CLINICAL NEUROSCIENCE 2012. [PMID: 22033483 PMCID: PMC3181676 DOI: 10.31887/dcns.2002.4.2/rgur] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gender and aging moderate brain-behavior relationships. Advances in neuroscience enable integration of neurobehavioral, neuroanatomic, and neurophysiology measures. Here we present neurobehavioral studies thai examine cognitive and emotion processing in healthy men and women and highlight the effects of sex differences and aqinq. Neuroanatomic studies with maqnetic resonance imaging (MRI) indicate that the progressive decrease in brain volume affects froniotemporal brain regions in men more than in Vi/omen, Functional imaging methods suggest sex differences in rate of blood flow, pattern of glucose metabolism, and receptor activity. The role of ovarian hormones is important in elucidating the observed relationships. A life span perspective on gender differences through the integration of available methodologies will advance understanding healthy people and the effects of brain disorders.
Collapse
Affiliation(s)
- Raquel E Gur
- Neuropsychiatry Section, Department of Psychiatry, University of Pennsylvania Medical Center, Philadelphia, Pa, USA
| | | |
Collapse
|
31
|
YIN DZ, LIU WF. Carbonyl Stress, Aging Mechanisms, and The Up-stream Molecular Etiology of Alzheimer′s Disease*. PROG BIOCHEM BIOPHYS 2012. [DOI: 10.3724/sp.j.1206.2012.00316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Ritz MF, Grond-Ginsbach C, Engelter S, Lyrer P. Gene expression suggests spontaneously hypertensive rats may have altered metabolism and reduced hypoxic tolerance. Curr Neurovasc Res 2012; 9:10-9. [PMID: 22272763 PMCID: PMC3296125 DOI: 10.2174/156720212799297074] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 12/07/2011] [Accepted: 12/15/2011] [Indexed: 12/27/2022]
Abstract
Cerebral small vessel disease (SVD) is an important cause of stroke, cognitive decline and vascular dementia (VaD). It is associated with diffuse white matter abnormalities and small deep cerebral ischemic infarcts. The molecular mechanisms involved in the development and progression of SVD are unclear. As hypertension is a major risk factor for developing SVD, Spontaneously Hypertensive Rats (SHR) are considered an appropriate experimental model for SVD. Prior work suggested an imbalance between the number of blood microvessels and astrocytes at the level of the neurovascular unit in 2-month-old SHR, leading to neuronal hypoxia in the brain of 9-month-old animals. To identify genes and pathways involved in the development of SVD, we compared the gene expression profile in the cortex of 2 and 9-month-old of SHR with age-matched normotensive Wistar Kyoto (WKY) rats using microarray-based technology. The results revealed significant differences in expression of genes involved in energy and lipid metabolisms, mitochondrial functions, oxidative stress and ischemic responses between both groups. These results strongly suggest that SHR suffer from chronic hypoxia, and therefore are unable to tolerate ischemia-like conditions, and are more vulnerable to high-energy needs than WKY. This molecular analysis gives new insights about pathways accounting for the development of SVD.
Collapse
Affiliation(s)
- Marie-Françoise Ritz
- Department of Biomedicine, Brain Tumor Biology Laboratory, Pharmazentrum, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | | | | | | |
Collapse
|
33
|
Kulkarni VA, Firestein BL. The dendritic tree and brain disorders. Mol Cell Neurosci 2012; 50:10-20. [DOI: 10.1016/j.mcn.2012.03.005] [Citation(s) in RCA: 242] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 03/09/2012] [Indexed: 01/21/2023] Open
|
34
|
Inhibition of JNK by a peptide inhibitor reduces traumatic brain injury-induced tauopathy in transgenic mice. J Neuropathol Exp Neurol 2012; 71:116-29. [PMID: 22249463 DOI: 10.1097/nen.0b013e3182456aed] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) is a major environmental risk factor for subsequent development of Alzheimer disease (AD). Pathological features that are common to AD and many tauopathies are neurofibrillary tangles (NFTs) and neuropil threads composed of hyperphosphorylated tau. Axonal accumulations of total and phospho-tau have been observed within hours to weeks, and intracytoplasmic NFTs have been documented years after severe TBI in humans. We previously reported that controlled cortical impact TBI accelerated tau pathology in young 3xTg-AD mice. Here, we used this TBI mouse model to investigate mechanisms responsible for increased tau phosphorylation and accumulation after brain trauma. We found that TBI resulted in abnormal axonal accumulation of several kinases that phosphorylate tau. Notably, c-Jun N-terminal kinase (JNK) was markedly activated in injured axons and colocalized with phospho-tau. We found that moderate reduction of JNK activity (40%) by a peptide inhibitor, D-JNKi1, was sufficient to reduce total and phospho-tau accumulations in axons of these mice with TBI. Longer-term studies will be required to determine whether reducing acute tau pathology proves beneficial in brain trauma.
Collapse
|
35
|
Changes in hippocampal synapses and learning-memory abilities in a streptozotocin-treated rat model and intervention by using fasudil hydrochloride. Neuroscience 2012; 200:120-9. [DOI: 10.1016/j.neuroscience.2011.10.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 10/15/2011] [Accepted: 10/17/2011] [Indexed: 01/03/2023]
|
36
|
Valero J, España J, Parra-Damas A, Martín E, Rodríguez-Álvarez J, Saura CA. Short-term environmental enrichment rescues adult neurogenesis and memory deficits in APP(Sw,Ind) transgenic mice. PLoS One 2011; 6:e16832. [PMID: 21347380 PMCID: PMC3036721 DOI: 10.1371/journal.pone.0016832] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 01/17/2011] [Indexed: 12/12/2022] Open
Abstract
Epidemiological studies indicate that intellectual activity prevents or delays the onset of Alzheimer's disease (AD). Similarly, cognitive stimulation using environmental enrichment (EE), which increases adult neurogenesis and functional integration of newborn neurons into neural circuits of the hippocampus, protects against memory decline in transgenic mouse models of AD, but the mechanisms involved are poorly understood. To study the therapeutic benefits of cognitive stimulation in AD we examined the effects of EE in hippocampal neurogenesis and memory in a transgenic mouse model of AD expressing the human mutant β-amyloid (Aβ) precursor protein (APPSw,Ind). By using molecular markers of new generated neurons (bromodeoxiuridine, NeuN and doublecortin), we found reduced neurogenesis and decreased dendritic length and projections of doublecortin-expressing cells of the dentate gyrus in young APPSw,Ind transgenic mice. Moreover, we detected a lower number of mature neurons (NeuN positive) in the granular cell layer and a reduced volume of the dentate gyrus that could be due to a sustained decrease in the incorporation of new generated neurons. We found that short-term EE for 7 weeks efficiently ameliorates early hippocampal-dependent spatial learning and memory deficits in APPSw,Ind transgenic mice. The cognitive benefits of enrichment in APPSw,Ind transgenic mice were associated with increased number, dendritic length and projections to the CA3 region of the most mature adult newborn neurons. By contrast, Aβ levels and the total number of neurons in the dentate gyrus were unchanged by EE in APPSw,Ind mice. These results suggest that promoting the survival and maturation of adult generated newborn neurons in the hippocampus may contribute to cognitive benefits in AD mouse models.
Collapse
Affiliation(s)
- Jorge Valero
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Judit España
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Arnaldo Parra-Damas
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elsa Martín
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Rodríguez-Álvarez
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos A. Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
37
|
Lin YC, Koleske AJ. Mechanisms of synapse and dendrite maintenance and their disruption in psychiatric and neurodegenerative disorders. Annu Rev Neurosci 2011; 33:349-78. [PMID: 20367247 DOI: 10.1146/annurev-neuro-060909-153204] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Emerging evidence indicates that once established, synapses and dendrites can be maintained for long periods, if not for the organism's entire lifetime. In contrast to the wealth of knowledge regarding axon, dendrite, and synapse development, we understand comparatively little about the cellular and molecular mechanisms that enable long-term synapse and dendrite maintenance. Here, we review how the actin cytoskeleton and its regulators, adhesion receptors, and scaffolding proteins mediate synapse and dendrite maintenance. We examine how these mechanisms are reinforced by trophic signals passed between the pre- and postsynaptic compartments. We also discuss how synapse and dendrite maintenance mechanisms are compromised in psychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yu-Chih Lin
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520-8024, USA.
| | | |
Collapse
|
38
|
Couch BA, DeMarco GJ, Gourley SL, Koleske AJ. Increased dendrite branching in AbetaPP/PS1 mice and elongation of dendrite arbors by fasudil administration. J Alzheimers Dis 2010; 20:1003-8. [PMID: 20413901 DOI: 10.3233/jad-2010-091114] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Amyloid-beta (Abeta) overproduction and dendrite arbor atrophy are hallmarks of Alzheimer's disease. The RhoA GTPase (Rho) signals through Rho kinase (ROCK) to control cytoskeletal dynamics and regulate neuron structure. Hyperactive Rho signaling destabilizes neurons leading to dendritic regression that can be rescued by genetic or pharmacological reduction of ROCK signaling. To understand what effect reduced ROCK signaling has on the dendrite arbors of mice that overproduce Abeta, we administered the ROCK inhibitor fasudil to AbetaPP/PS1 transgenic mice. We report that increased dendrite branching occurs in AbetaPP/PS1 mice and that fasudil promotes lengthening of the dendrite arbors of CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Brian A Couch
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
39
|
Morse TM, Carnevale NT, Mutalik PG, Migliore M, Shepherd GM. Abnormal Excitability of Oblique Dendrites Implicated in Early Alzheimer's: A Computational Study. Front Neural Circuits 2010; 4. [PMID: 20725509 PMCID: PMC2901152 DOI: 10.3389/fncir.2010.00016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 05/04/2010] [Indexed: 11/13/2022] Open
Abstract
The integrative properties of cortical pyramidal dendrites are essential to the neural basis of cognitive function, but the impact of amyloid beta protein (abeta) on these properties in early Alzheimer's is poorly understood. In animal models, electrophysiological studies of proximal dendrites have shown that abeta induces hyperexcitability by blocking A-type K+ currents (I(A)), disrupting signal integration. The present study uses a computational approach to analyze the hyperexcitability induced in distal dendrites beyond the experimental recording sites. The results show that back-propagating action potentials in the dendrites induce hyperexcitability and excessive calcium concentrations not only in the main apical trunk of pyramidal cell dendrites, but also in their oblique dendrites. Evidence is provided that these thin branches are particularly sensitive to local reductions in I(A). The results suggest the hypothesis that the oblique branches may be most vulnerable to disruptions of I(A) by early exposure to abeta, and point the way to further experimental analysis of these actions as factors in the neural basis of the early decline of cognitive function in Alzheimer's.
Collapse
Affiliation(s)
- Thomas M Morse
- Department of Neurobiology, Yale University School of Medicine New Haven, CT, USA
| | | | | | | | | |
Collapse
|
40
|
Previtera ML, Langhammer CG, Firestein BL. Effects of substrate stiffness and cell density on primary hippocampal cultures. J Biosci Bioeng 2010; 110:459-70. [PMID: 20547372 DOI: 10.1016/j.jbiosc.2010.04.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 04/01/2010] [Accepted: 04/14/2010] [Indexed: 01/06/2023]
Abstract
Previous studies have shown that dendrites are influenced by substrate stiffness when neurons are plated in either pure or mixed cultures. However, because substrate rigidity can also affect other aspects of culture development known to impact dendrite branching, such as overall cell number, it is unclear whether substrate stiffness exerts a direct or indirect effect on dendrite morphology. In this study, we determine whether substrate stiffness plays a critical role in regulating dendrite branching independent of cell number. We plated primary mixed hippocampal cultures on soft and stiff gels, with Young's moduli of 1 kPa and 7 kPa, respectively. We found that neurons plated on stiffer substrates showed increased branching relative to neurons grown on softer substrates at the same cell number. On the stiff gels, we also observed a cell number-dependent effect, in which increasing initial plating density decreased dendrite branching. This change correlates with an increase in extracellular glutamate. We concluded that both cell number and substrate stiffness play roles in determining dendrite branching, and that the two effects are independent of one another.
Collapse
Affiliation(s)
- Michelle L Previtera
- Molecular Biosciences Graduate Program, Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | | | | |
Collapse
|
41
|
Shetty AK, Hattiangady B, Rao MS, Shuai B. Deafferentation enhances neurogenesis in the young and middle aged hippocampus but not in the aged hippocampus. Hippocampus 2010; 21:631-46. [PMID: 20333732 DOI: 10.1002/hipo.20776] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2010] [Indexed: 12/13/2022]
Abstract
Increased neurogenesis in the dentate gyrus (DG) after brain insults such as excitotoxic lesions, seizures, or stroke is a well known phenomenon in the young hippocampus. This plasticity reflects an innate compensatory response of neural stem cells (NSCs) in the young hippocampus to preserve function or minimize damage after injury. However, injuries to the middle-aged and aged hippocampi elicit either no or dampened neurogenesis response, which could be due to an altered plasticity of NSCs and/or the hippocampus with age. We examined whether the plasticity of NSCs to increase neurogenesis in response to a milder injury such as partial deafferentation is preserved during aging. We quantified DG neurogenesis in the hippocampus of young, middle-aged, and aged F344 rats after partial deafferentation. A partial deafferentation of the left hippocampus without any apparent cell loss was induced via administration of Kainic acid (0.5 μg in 1.0 μl) into the right lateral ventricle of the brain. In this model, degeneration of CA3 pyramidal neurons and dentate hilar neurons in the right hippocampus results in loss of commissural axons which leads to partial deafferentation of the dendrites of dentate granule cells and CA1-CA3 pyramidal neurons in the left hippocampus. Quantification of newly born cells that are added to the dentate granule cell layer at postdeafferentation days 4-15 using 5'-bromodeoxyuridine (BrdU) labeling revealed greatly increased addition of newly born cells (∼three fold increase) in the deafferented young and middle-aged hippocampi but not in the deafferented aged hippocampus. Measurement of newly born neurons using doublecortin (DCX) immunostaining also revealed similar findings. Analyses using BrdU-DCX dual immunofluorescence demonstrated no changes in neuronal fate-choice decision of newly born cells after deafferentation, in comparison to the age-matched naive hippocampus in all age groups. Thus, the plasticity of hippocampal NSCs to increase DG neurogenesis in response to a milder injury such as partial hippocampal deafferentation is preserved until middle age but lost at old age.
Collapse
Affiliation(s)
- Ashok K Shetty
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, North Carolina, USA.
| | | | | | | |
Collapse
|
42
|
Dendritic vulnerability in neurodegenerative disease: insights from analyses of cortical pyramidal neurons in transgenic mouse models. Brain Struct Funct 2010; 214:181-99. [PMID: 20177698 DOI: 10.1007/s00429-010-0244-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 02/05/2010] [Indexed: 12/27/2022]
Abstract
In neurodegenerative disorders, such as Alzheimer's disease, neuronal dendrites and dendritic spines undergo significant pathological changes. Because of the determinant role of these highly dynamic structures in signaling by individual neurons and ultimately in the functionality of neuronal networks that mediate cognitive functions, a detailed understanding of these changes is of paramount importance. Mutant murine models, such as the Tg2576 APP mutant mouse and the rTg4510 tau mutant mouse have been developed to provide insight into pathogenesis involving the abnormal production and aggregation of amyloid and tau proteins, because of the key role that these proteins play in neurodegenerative disease. This review showcases the multidimensional approach taken by our collaborative group to increase understanding of pathological mechanisms in neurodegenerative disease using these mouse models. This approach includes analyses of empirical 3D morphological and electrophysiological data acquired from frontal cortical pyramidal neurons using confocal laser scanning microscopy and whole-cell patch-clamp recording techniques, combined with computational modeling methodologies. These collaborative studies are designed to shed insight on the repercussions of dystrophic changes in neocortical neurons, define the cellular phenotype of differential neuronal vulnerability in relevant models of neurodegenerative disease, and provide a basis upon which to develop meaningful therapeutic strategies aimed at preventing, reversing, or compensating for neurodegenerative changes in dementia.
Collapse
|
43
|
|
44
|
Fujiwara H, Tabuchi M, Yamaguchi T, Iwasaki K, Furukawa K, Sekiguchi K, Ikarashi Y, Kudo Y, Higuchi M, Saido TC, Maeda S, Takashima A, Hara M, Yaegashi N, Kase Y, Arai H. A traditional medicinal herbPaeonia suffruticosaand its active constituent 1,2,3,4,6-penta-O-galloyl-β-d-glucopyranose have potent anti-aggregation effects on Alzheimer’s amyloid β proteinsin vitroandin vivo. J Neurochem 2009; 109:1648-57. [DOI: 10.1111/j.1471-4159.2009.06069.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
45
|
Alonso ADC, Corbo CP. Novel therapeutics based on tau/microtubule dynamics: WO2008084483. Expert Opin Ther Pat 2009; 19:1335-8. [DOI: 10.1517/13543770902887815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
46
|
Jan A, Gokce O, Luthi-Carter R, Lashuel HA. The ratio of monomeric to aggregated forms of Abeta40 and Abeta42 is an important determinant of amyloid-beta aggregation, fibrillogenesis, and toxicity. J Biol Chem 2008; 283:28176-89. [PMID: 18694930 PMCID: PMC2661389 DOI: 10.1074/jbc.m803159200] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 08/06/2008] [Indexed: 11/06/2022] Open
Abstract
Aggregation and fibril formation of amyloid-beta (Abeta) peptides Abeta40 and Abeta42 are central events in the pathogenesis of Alzheimer disease. Previous studies have established the ratio of Abeta40 to Abeta42 as an important factor in determining the fibrillogenesis, toxicity, and pathological distribution of Abeta. To better understand the molecular basis underlying the pathologic consequences associated with alterations in the ratio of Abeta40 to Abeta42, we probed the concentration- and ratio-dependent interactions between well defined states of the two peptides at different stages of aggregation along the amyloid formation pathway. We report that monomeric Abeta40 alters the kinetic stability, solubility, and morphological properties of Abeta42 aggregates and prevents their conversion into mature fibrils. Abeta40, at approximately equimolar ratios (Abeta40/Abeta42 approximately 0.5-1), inhibits (> 50%) fibril formation by monomeric Abeta42, whereas inhibition of protofibrillar Abeta42 fibrillogenesis is achieved at lower, substoichiometric ratios (Abeta40/Abeta42 approximately 0.1). The inhibitory effect of Abeta40 on Abeta42 fibrillogenesis is reversed by the introduction of excess Abeta42 monomer. Additionally, monomeric Abeta42 and Abeta40 are constantly recycled and compete for binding to the ends of protofibrillar and fibrillar Abeta aggregates. Whereas the fibrillogenesis of both monomeric species can be seeded by fibrils composed of either peptide, Abeta42 protofibrils selectively seed the fibrillogenesis of monomeric Abeta42 but not monomeric Abeta40. Finally, we also show that the amyloidogenic propensities of different individual and mixed Abeta species correlates with their relative neuronal toxicities. These findings, which highlight specific points in the amyloid peptide equilibrium that are highly sensitive to the ratio of Abeta40 to Abeta42, carry important implications for the pathogenesis and current therapeutic strategies of Alzheimer disease.
Collapse
Affiliation(s)
- Asad Jan
- Laboratory of Molecular Neurobiology and Neuroproteomics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | |
Collapse
|
47
|
Inhibition of Rho via Arg and p190RhoGAP in the postnatal mouse hippocampus regulates dendritic spine maturation, synapse and dendrite stability, and behavior. J Neurosci 2007; 27:10982-92. [PMID: 17928439 DOI: 10.1523/jneurosci.0793-07.2007] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The RhoA (Rho) GTPase is a master regulator of dendrite morphogenesis. Rho activation in developing neurons slows dendrite branch dynamics, yielding smaller, less branched dendrite arbors. Constitutive activation of Rho in mature neurons causes dendritic spine loss and dendritic regression, indicating that Rho can affect dendritic structure and function even after dendrites have developed. However, it is unclear whether and how endogenous Rho modulates dendrite and synapse morphology after dendrite arbor development has occurred. We demonstrate that a Rho inhibitory pathway involving the Arg tyrosine kinase and p190RhoGAP is essential for synapse and dendrite stability during late postnatal development. Hippocampal CA1 pyramidal dendrites develop normally in arg-/- mice, reaching their mature size by postnatal day 21 (P21). However, dendritic spines do not undergo the normal morphological maturation in these mice, leading to a loss of hippocampal synapses and dendritic branches by P42. Coincident with this synapse and dendrite loss, arg-/- mice exhibit progressive deficits in a hippocampus-dependent object recognition behavioral task. p190RhoGAP localizes to dendritic spines, and its activity is reduced in arg-/- hippocampus, leading to increased Rho activity. Although mutations in p190rhogap enhance dendritic regression resulting from decreased Arg levels, reducing gene dosage of the Rho effector ROCKII can suppress the dendritic regression observed in arg-/- mice. Together, these data indicate that signaling through Arg and p190RhoGAP acts late during synaptic refinement to promote dendritic spine maturation and synapse/dendrite stability by attenuating synaptic Rho activity.
Collapse
|
48
|
Vallotton P, Lagerstrom R, Sun C, Buckley M, Wang D, De Silva M, Tan SS, Gunnersen JM. Automated analysis of neurite branching in cultured cortical neurons using HCA-Vision. Cytometry A 2007; 71:889-95. [PMID: 17868085 DOI: 10.1002/cyto.a.20462] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Manual neuron tracing is a very labor-intensive task. In the drug screening context, the sheer number of images to process means that this approach is unrealistic. Moreover, the lack of reproducibility, objectivity, and auditing capability of manual tracing is limiting even in the context of smaller studies. We have developed fast, sensitive, and reliable algorithms for the purpose of detecting and analyzing neurites in cell cultures, and we have integrated them in software called HCA-Vision, suitable for the research environment. We validate the software on images of cortical neurons by comparing results obtained using HCA-Vision with those obtained using an established semi-automated tracing solution (NeuronJ). The effect of the Sez-6 deletion was characterized in detail. Sez-6 null neurons exhibited a significant increase in neurite branching, although the neurite field area was unchanged due to a reduction in mean branch length. HCA-Vision delivered considerable speed benefits and reliable traces.
Collapse
Affiliation(s)
- Pascal Vallotton
- CSIRO, Mathematical and Information Sciences, North Ryde, NSW, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Reynolds MR, Reyes JF, Fu Y, Bigio EH, Guillozet-Bongaarts AL, Berry RW, Binder LI. Tau nitration occurs at tyrosine 29 in the fibrillar lesions of Alzheimer's disease and other tauopathies. J Neurosci 2006; 26:10636-45. [PMID: 17050703 PMCID: PMC6674733 DOI: 10.1523/jneurosci.2143-06.2006] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The neurodegenerative tauopathies are a clinically diverse group of diseases typified by the pathological self-assembly of the microtubule-associated tau protein. Although tau nitration is believed to influence the pathogenesis of these diseases, the precise residues modified, and the resulting effects on tau function, remain enigmatic. Previously, we demonstrated that nitration at residue Tyr29 markedly inhibits the ability of tau to self-associate and stabilize the microtubule lattice (Reynolds et al., 2005b, 2006). Here, we report the first monoclonal antibody to detect nitration in a protein-specific and site-selective manner. This reagent, termed Tau-nY29, recognizes tau only when nitrated at residue Tyr29. It does not cross-react with wild-type tau, tau mutants singly nitrated at Tyr18, Tyr197, and Tyr394, or other proteins known to be nitrated in neurodegenerative diseases. By Western blot analysis, Tau-nY29 detects soluble tau and paired helical filament tau from severely affected Alzheimer's brain but fails to recognize tau from normal aged brain. This observation suggests that nitration at Tyr29 is a disease-related event that may alter the intrinsic ability of tau to self-polymerize. In Alzheimer's brain, Tau-nY29 labels the fibrillar triad of tau lesions, including neurofibrillary tangles, neuritic plaques, and, to a lesser extent, neuropil threads. Intriguingly, although Tau-nY29 stains both the neuronal and glial tau pathology of Pick disease, it detects only the neuronal pathology in corticobasal degeneration and progressive supranuclear palsy without labeling the predominant glial pathology. Collectively, our findings provide the first direct evidence that site-specific tau nitration is linked to the progression of the neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Matthew R Reynolds
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Schönheit B, Glöckner F, Ohm TG. Apolipoprotein E polymorphism and dendritic shape in hippocampal interneurons. Neurobiol Aging 2006; 28:677-86. [PMID: 17023092 DOI: 10.1016/j.neurobiolaging.2006.03.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Revised: 03/16/2006] [Accepted: 03/31/2006] [Indexed: 11/20/2022]
Abstract
The apolipoprotein E genetic polymorphism exerts a well described influence on Alzheimer's disease (AD) risk, although the pathogenetic mechanism is still not clear. Increasing evidence points to a diminished neuroplasticity in apolipoprotein E varepsilon4-allele carriers. But, alternatively or additionally, developmental differences in dendritic geometry may be associated with the polymorphism. We morphometrically examined the dendritic ramification of CA1 Parvalbumin-positive GABAergic hippocampal neurons (n=571) in matched pairs of aged non-demented individuals with different apolipoprotein E genotype. We chose Parvalbumin-positive interneurons since they lack potentially confounding AD-like cytoskeletal changes. To minimize the risk of transneuronal dendritic changes due to significant deafferentation we focused on non-demented individuals. In this chosen paradigm, neither the disease-associated apolipoprotein E varepsilon4-allele nor the apolipoprotein E varepsilon2-allele had a significant impact on dendritic shape when compared to the most common allelic variant apolipoprotein E varepsilon3/3. At least with respect to the studied cell type, the data suggest that the apolipoprotein E polymorphism does not modulate the original formation of dendrites in vivo, contrary to conclusions drawn from in vitro studies on neurite outgrowth.
Collapse
Affiliation(s)
- Bärbel Schönheit
- Institute of Integrative Neuroanatomy, Department of Clinical Cell- and Neurobiology, Charité, 10098 Berlin, Germany
| | | | | |
Collapse
|