1
|
Niraula S, Doderer JJ, Indulkar S, Berry KP, Hauser WL, L'Esperance OJ, Deng JZ, Keeter G, Rouse AG, Subramanian J. Excitation-inhibition imbalance disrupts visual familiarity in amyloid and non-pathology conditions. Cell Rep 2023; 42:111946. [PMID: 36640331 PMCID: PMC9939293 DOI: 10.1016/j.celrep.2022.111946] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/14/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Neuronal hyperactivity induces memory deficits in Alzheimer's disease. However, how hyperactivity disrupts memory is unclear. Using in vivo synaptic imaging in the mouse visual cortex, we show that structural excitatory-inhibitory synapse imbalance in the apical dendrites favors hyperactivity in early amyloidosis. Consistent with this, natural images elicit neuronal hyperactivity in these mice. Compensatory changes that maintain activity homeostasis disrupt functional connectivity and increase population sparseness such that a small fraction of neurons dominates population activity. These properties reduce the selectivity of neural response to natural images and render visual recognition memory vulnerable to interference. Deprivation of non-specific visual experiences improves the neural representation and behavioral expression of visual familiarity. In contrast, in non-pathological conditions, deprivation of non-specific visual experiences induces disinhibition, increases excitability, and disrupts visual familiarity. We show that disrupted familiarity occurs when the fraction of high-responsive neurons and the persistence of neural representation of a memory-associated stimulus are not constrained.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Julia J Doderer
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Shreya Indulkar
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Kalen P Berry
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - William L Hauser
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Oliver J L'Esperance
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Jasmine Z Deng
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Griffin Keeter
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Adam G Rouse
- Department of Neurosurgery, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
2
|
Nguyen LD, Fischer TT, Ehrlich BE. Pharmacological rescue of cognitive function in a mouse model of chemobrain. Mol Neurodegener 2021; 16:41. [PMID: 34174909 PMCID: PMC8235868 DOI: 10.1186/s13024-021-00463-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 06/09/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND After chemotherapy, many cancer survivors suffer from long-lasting cognitive impairment, colloquially known as "chemobrain." However, the trajectories of cognitive changes and the underlying mechanisms remain unclear. We previously established paclitaxel-induced inositol trisphosphate receptor (InsP3R)-dependent calcium oscillations as a mechanism for peripheral neuropathy, which was prevented by lithium pretreatment. Here, we investigated if a similar mechanism also underlay paclitaxel-induced chemobrain. METHOD Mice were injected with 4 doses of 20 mg/kg paclitaxel every other day to induced cognitive impairment. Memory acquisition was assessed with the displaced object recognition test. The morphology of neurons in the prefrontal cortex and the hippocampus was analyzed using Golgi-Cox staining, followed by Sholl analyses. Changes in protein expression were measured by Western blot. RESULTS Mice receiving paclitaxel showed impaired short-term spatial memory acquisition both acutely 5 days post injection and chronically 23 days post injection. Dendritic length and complexity were reduced in the hippocampus and the prefrontal cortex after paclitaxel injection. Concurrently, the expression of protein kinase C α (PKCα), an effector in the InsP3R pathway, was increased. Treatment with lithium before or shortly after paclitaxel injection rescued the behavioral, cellular, and molecular deficits observed. Similarly, memory and morphological deficits could be rescued by pretreatment with chelerythrine, a PKC inhibitor. CONCLUSION We establish the InsP3R calcium pathway and impaired neuronal morphology as mechanisms for paclitaxel-induced cognitive impairment. Our findings suggest lithium and PKC inhibitors as candidate agents for preventing chemotherapy-induced cognitive impairment.
Collapse
Affiliation(s)
- Lien D Nguyen
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA.,Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06520, USA.,Present Address: Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Tom T Fischer
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA.,Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA. .,Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
3
|
Du Q, Chang J, Cheng G, Zhao Y, Zhou W. Sunday Driver Mediates Multi-Compartment Golgi Outposts Defects Induced by Amyloid Precursor Protein. Front Neurosci 2021; 15:673684. [PMID: 34140878 PMCID: PMC8205063 DOI: 10.3389/fnins.2021.673684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
Golgi defects including Golgi fragmentation are pathological features of Alzheimer’s disease (AD). As a pathogenic factor in AD, amyloid precursor protein (APP) induces Golgi fragmentation in the soma. However, how APP regulates Golgi outposts (GOs) in dendrites remains unclear. Given that APP resides in and affects the movements of GOs, and in particular, reverses the distribution of multi-compartment GOs (mcGOs), we investigated the regulatory mechanism of mcGO movements in the Drosophila larvae. Knockdown experiments showed that the bidirectional mcGO movements were cooperatively controlled by the dynein heavy chain (Dhc) and kinesin heavy chain subunits. Notably, only Dhc mediated APP’s regulation of mcGO movements. Furthermore, by loss-of-function screening, the adaptor protein Sunday driver (Syd) was identified to mediate the APP-induced alteration of the direction of mcGO movements and dendritic defects. Collectively, by elucidating a model of bidirectional mcGO movements, we revealed the mechanism by which APP regulates the direction of mcGO movements. Our study therefore provides new insights into AD pathogenesis.
Collapse
Affiliation(s)
- Qianqian Du
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Chang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Guo Cheng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Yinyin Zhao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhou
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
The Interaction Between Contactin and Amyloid Precursor Protein and Its Role in Alzheimer’s Disease. Neuroscience 2020; 424:184-202. [DOI: 10.1016/j.neuroscience.2019.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023]
|
5
|
Protective effect of potassium 2-(l-hydroxypentyl)-benzoate on hippocampal neurons, synapses and dystrophic axons in APP/PS1 mice. Psychopharmacology (Berl) 2019; 236:2761-2771. [PMID: 31165206 DOI: 10.1007/s00213-019-05251-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/18/2019] [Indexed: 10/26/2022]
Abstract
RATIONALE As the hub of memory and space, hippocampus is very sensitive to a wide variety of injuries and is one of the earliest brain structures to develop neurodegenerative changes in AD. Previous research has showed a protective effect of potassium 2-(l-hydroxypentyl)-benzoate (PHPB) on cognitive deficits in animal models of AD. However, it is unclear whether this protective effect is associated with hippocampal alterations. OBJECTIVES The present study was conducted to evaluate the protective effect of PHPB on hippocampal neurodegenerative changes in middle-aged APP/PS1 mice. METHODS Ten-month-old male APP/PS1 transgenic mice and age-matched wild-type mice were randomly divided into three groups. PHPB-treated APP/PS1 group received 30 mg/kg PHPB by oral gavage once daily for 12 weeks. Wild-type group and APP/PS1 group received the same volume of water alone. Twelve weeks later, mice (13-month-old) were tested for in vivo 1H-MRS examination and then sacrificed for subsequent biochemical and pathological examinations using transmission electron microscopy, Golgi staining, immunohistochemistry, and western blotting. RESULTS We found that PHPB treatment significantly improved the micromorphology of hippocampal neurons and subcellular organelles, ameliorated synapse loss and presynaptic axonal dystrophy, increased hippocampal dendritic spine density and dendritic complexity, enhanced the expression of hippocampal synapse-associated proteins, and improved hippocampal metabolism in middle-aged APP/PS1 mice. CONCLUSIONS Our study showed for the first time the protective effect of PHPB on hippocampal neurons, synapses, and dystrophic axons in APP/PS1 mice, which to some extent revealed the possible mechanism for its ability to improve cognition in animal models of AD.
Collapse
|
6
|
Farhoodi R, Lansdell BJ, Kording KP. Quantifying How Staining Methods Bias Measurements of Neuron Morphologies. Front Neuroinform 2019; 13:36. [PMID: 31191283 PMCID: PMC6541099 DOI: 10.3389/fninf.2019.00036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 04/25/2019] [Indexed: 12/20/2022] Open
Abstract
The process through which neurons are labeled is a key methodological choice in measuring neuron morphology. However, little is known about how this choice may bias measurements. To quantify this bias we compare the extracted morphology of neurons collected from the same rodent species, experimental condition, gender distribution, age distribution, brain region and putative cell type, but obtained with 19 distinct staining methods. We found strong biases on measured features of morphology. These were largest in features related to the coverage of the dendritic tree (e.g., the total dendritic tree length). Understanding measurement biases is crucial for interpreting morphological data.
Collapse
Affiliation(s)
- Roozbeh Farhoodi
- Department of Mathematics, Sharif University of Technology, Tehran, Iran
| | | | - Konrad Paul Kording
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States.,Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
Zou X, Zhong L, Zhu C, Zhao H, Zhao F, Cui R, Gao S, Li B. Role of Leptin in Mood Disorder and Neurodegenerative Disease. Front Neurosci 2019; 13:378. [PMID: 31130833 PMCID: PMC6510114 DOI: 10.3389/fnins.2019.00378] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
The critical regulatory role of leptin in the neuroendocrine system has been widely reported. Significantly, leptin can improve learning and memory, affect hippocampal synaptic plasticity, exert neuroprotective efficacy and reduce the risk of several neuropsychiatric diseases. In terms of depression, leptin could modulate the levels of neurotransmitters, neurotrophic factors and reverse the dysfunction in the hypothalamic-pituitary-adrenal axis (HPA). At the same time, leptin affects neurological diseases during the regulation of metabolic homeostasis. With regards to neurodegenerative diseases, leptin can affect them via neuroprotection, mainly including Alzheimer's disease and Parkinson's disease. This review will summarize the mechanisms of leptin signaling within the neuroendocrine system with respect to these diseases and discuss the therapeutic potential of leptin.
Collapse
Affiliation(s)
- Xiaohan Zou
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Lili Zhong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Cuilin Zhu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Haisheng Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Shuohui Gao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Soluble Aβ Oligomers Impair Dipolar Heterodendritic Plasticity by Activation of mGluR in the Hippocampal CA1 Region. iScience 2018; 6:138-150. [PMID: 30240608 PMCID: PMC6137707 DOI: 10.1016/j.isci.2018.07.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/05/2018] [Accepted: 07/19/2018] [Indexed: 11/20/2022] Open
Abstract
Soluble Aβ oligomers (oAβs) contribute importantly to synaptotoxicity in Alzheimer disease (AD), but the mechanisms related to heterogeneity of synaptic functions at local circuits remain elusive. Nearly all studies of the effects of oAβs on hippocampal synaptic plasticity have only examined homosynaptic plasticity. Here we stimulated the Schaffer collaterals and then simultaneously recorded in stratum radiatum (apical dendrites) and stratum oriens (basal dendrites) of CA1 neurons. We found that the apical dendrites are significantly more vulnerable to oAβ-mediated synaptic dysfunction: the heterosynaptic basal dendritic long-term potentiation (LTP) remained unchanged, whereas the homosynaptic apical LTP was impaired. However, the heterosynaptic basal dendritic plasticity induced by either spaced 10-Hz bursts or low-frequency (1-Hz) stimulation was disrupted by oAβs in a mGluR5-dependent manner. These results suggest that different firing patterns in the same neurons may be selectively altered by soluble oAβs in an early phase of AD, before frank neurodegeneration.
Collapse
|
9
|
Mockett BG, Richter M, Abraham WC, Müller UC. Therapeutic Potential of Secreted Amyloid Precursor Protein APPsα. Front Mol Neurosci 2017; 10:30. [PMID: 28223920 PMCID: PMC5293819 DOI: 10.3389/fnmol.2017.00030] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/25/2017] [Indexed: 11/26/2022] Open
Abstract
Cleavage of the amyloid precursor protein (APP) by α-secretase generates an extracellularly released fragment termed secreted APP-alpha (APPsα). Not only is this process of interest due to the cleavage of APP within the amyloid-beta sequence, but APPsα itself has many physiological properties that suggest its great potential as a therapeutic target. For example, APPsα is neurotrophic, neuroprotective, neurogenic, a stimulator of protein synthesis and gene expression, and enhances long-term potentiation (LTP) and memory. While most early studies have been conducted in vitro, effectiveness in animal models is now being confirmed. These studies have revealed that either upregulating α-secretase activity, acutely administering APPsα or chronic delivery of APPsα via a gene therapy approach can effectively treat mouse models of Alzheimer's disease (AD) and other disorders such as traumatic head injury. Together these findings suggest the need for intensifying research efforts to harness the therapeutic potential of this multifunctional protein.
Collapse
Affiliation(s)
- Bruce G. Mockett
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of OtagoOtago, New Zealand
| | - Max Richter
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg UniversityHeidelberg, Germany
| | - Wickliffe C. Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of OtagoOtago, New Zealand
| | - Ulrike C. Müller
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg UniversityHeidelberg, Germany
| |
Collapse
|
10
|
Blazquez-Llorca L, Valero-Freitag S, Rodrigues EF, Merchán-Pérez Á, Rodríguez JR, Dorostkar MM, DeFelipe J, Herms J. High plasticity of axonal pathology in Alzheimer's disease mouse models. Acta Neuropathol Commun 2017; 5:14. [PMID: 28173876 PMCID: PMC5296955 DOI: 10.1186/s40478-017-0415-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/26/2017] [Indexed: 02/05/2023] Open
Abstract
Axonal dystrophies (AxDs) are swollen and tortuous neuronal processes that are associated with extracellular depositions of amyloid β (Aβ) and have been observed to contribute to synaptic alterations occurring in Alzheimer's disease. Understanding the temporal course of this axonal pathology is of high relevance to comprehend the progression of the disease over time. We performed a long-term in vivo study (up to 210 days of two-photon imaging) with two transgenic mouse models (dE9xGFP-M and APP-PS1xGFP-M). Interestingly, AxDs were formed only in a quarter of GFP-expressing axons near Aβ-plaques, which indicates a selective vulnerability. AxDs, especially those reaching larger sizes, had long lifetimes and appeared as highly plastic structures with large variations in size and shape and axonal sprouting over time. In the case of the APP-PS1 mouse only, the formation of new long axonal segments in dystrophic axons (re-growth phenomenon) was observed. Moreover, new AxDs could appear at the same point of the axon where a previous AxD had been located before disappearance (re-formation phenomenon). In addition, we observed that most AxDs were formed and developed during the imaging period, and numerous AxDs had already disappeared by the end of this time. This work is the first in vivo study analyzing quantitatively the high plasticity of the axonal pathology around Aβ plaques. We hypothesized that a therapeutically early prevention of Aβ plaque formation or their growth might halt disease progression and promote functional axon regeneration and the recovery of neural circuits.
Collapse
|
11
|
Iikuni S, Ono M, Tanimura K, Watanabe H, Yoshimura M, Saji H. Synthesis and biological evaluation of novel technetium-99m-labeled phenylquinoxaline derivatives as single photon emission computed tomography imaging probes targeting β-amyloid plaques in Alzheimer's disease. RSC Adv 2017. [DOI: 10.1039/c6ra28395k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The development of an imaging probe targeting β-amyloid (Aβ) plaques in Alzheimer's disease labeled with technetium-99m, the most commonly used radioisotope for clinical diagnoses, has been strongly anticipated.
Collapse
Affiliation(s)
- Shimpei Iikuni
- Department of Patho-Functional Bioanalysis
- Graduate School of Pharmaceutical Sciences
- Kyoto University
- Kyoto 606-8501
- Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis
- Graduate School of Pharmaceutical Sciences
- Kyoto University
- Kyoto 606-8501
- Japan
| | - Keiichi Tanimura
- Department of Patho-Functional Bioanalysis
- Graduate School of Pharmaceutical Sciences
- Kyoto University
- Kyoto 606-8501
- Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis
- Graduate School of Pharmaceutical Sciences
- Kyoto University
- Kyoto 606-8501
- Japan
| | - Masashi Yoshimura
- Department of Patho-Functional Bioanalysis
- Graduate School of Pharmaceutical Sciences
- Kyoto University
- Kyoto 606-8501
- Japan
| | - Hideo Saji
- Department of Patho-Functional Bioanalysis
- Graduate School of Pharmaceutical Sciences
- Kyoto University
- Kyoto 606-8501
- Japan
| |
Collapse
|
12
|
Song JM, Sung YM, Nam JH, Yoon H, Chung A, Moffat E, Jung M, Pak DTS, Kim J, Hoe HS. A Mercaptoacetamide-Based Class II Histone Deacetylase Inhibitor Increases Dendritic Spine Density via RasGRF1/ERK Pathway. J Alzheimers Dis 2016; 51:591-604. [PMID: 26890742 DOI: 10.3233/jad-150717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The accumulation of amyloid-β (Aβ) leads to the loss of dendritic spines and synapses, which is hypothesized to cause cognitive impairments in Alzheimer's disease (AD) patients. In our previous study, we demonstrated that a novel mercaptoacetamide-based class II histone deacetylase inhibitor (HDACI), known as W2, decreased Aβ levels and improved learning and memory in mice. However, the underlying mechanism of this effect is unknown. OBJECTIVE Because dendritic spine formation is associated with cognitive performance, here we investigated whether HDACI W2 regulates dendritic spine density and its molecular mechanism of action. METHODS To examine the effect of HDACI W2 on dendritic spine density, we conducted morphological analysis of dendritic spines using GFP transfection and Golgi staining. In addition, to determine the molecular mechanism of W2 effects on spines, we measured the levels of mRNAs and proteins involved in the Ras signaling pathway using quantitative real-time PCR, immunocytochemistry, and western analysis. RESULTS We found that HDACI W2 altered dendritic spine density and morphology in vitro and in vivo. Additionally, W2 increased the mRNA or protein levels of Ras GRF1 and phospho-ERK. Moreover, knockdown of RasGRF1 and inhibition of ERK activity prevented the W2-mediated spinogenesis in primary hippocampal neurons. CONCLUSION Our Class II-selective HDACI W2 promotes the formation and growth of dendritic spines in a RasGRF1 and ERK dependent manner in primary hippocampal neurons.
Collapse
Affiliation(s)
- Jung Min Song
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - You Me Sung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| | - Hyejin Yoon
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Andrew Chung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Emily Moffat
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Mira Jung
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, USA
| | - Jungsu Kim
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.,Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| |
Collapse
|
13
|
Somogyi A, Katonai Z, Alpár A, Wolf E. A Novel Form of Compensation in the Tg2576 Amyloid Mouse Model of Alzheimer's Disease. Front Cell Neurosci 2016; 10:152. [PMID: 27378850 PMCID: PMC4909742 DOI: 10.3389/fncel.2016.00152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/27/2016] [Indexed: 12/02/2022] Open
Abstract
One century after its first description, pathology of Alzheimer’s disease (AD) is still poorly understood. Amyloid-related dendritic atrophy and membrane alterations of susceptible brain neurons in AD, and in animal models of AD are widely recognized. However, little effort has been made to study the potential effects of combined morphological and membrane alterations on signal transfer and synaptic integration in neurons that build up affected neural networks in AD. In this study spatial reconstructions and electrophysiological measurements of layer II/III pyramidal neurons of the somatosensory cortex from wild-type (WT) and transgenic (TG) human amyloid precursor protein (hAPP) overexpressing Tg2576 mice were used to build faithful segmental cable models of these neurons. Local synaptic activities were simulated in various points of the dendritic arbors and properties of subthreshold dendritic impulse propagation and predictors of synaptic input pattern recognition ability were quantified and compared in modeled WT and TG neurons. Despite the widespread dendritic degeneration and membrane alterations in mutant mouse neurons, surprisingly little, or no change was detected in steady-state and 50 Hz sinusoidal voltage transfers, current transfers, and local and propagation delays of PSPs traveling along dendrites of TG neurons. Synaptic input pattern recognition ability was also predicted to be unaltered in TG neurons in two different soma-dendritic membrane models investigated. Our simulations predict the way how subthreshold dendritic signaling and pattern recognition are preserved in TG neurons: amyloid-related membrane alterations compensate for the pathological effects that dendritic atrophy has on subthreshold dendritic signal transfer and integration in layer II/III somatosensory neurons of this hAPP mouse model for AD. Since neither propagation of single PSPs nor integration of multiple PSPs (pattern recognition) changes in TG neurons, we conclude that AD-related neuronal hyperexcitability cannot be accounted for by altered subthreshold dendritic signaling in these neurons but hyperexcitability is related to changes in active membrane properties and network connectivity.
Collapse
Affiliation(s)
- Attila Somogyi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecen, Hungary; Kenézy Gyula Hospital Ltd., Department of Emergency MedicineDebrecen, Hungary
| | - Zoltán Katonai
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen Debrecen, Hungary
| | - Alán Alpár
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of SciencesBudapest, Hungary; Department of Anatomy, Semmelweis UniversityBudapest, Hungary
| | - Ervin Wolf
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen Debrecen, Hungary
| |
Collapse
|
14
|
Imaging of Cerebral Amyloid Angiopathy with Bivalent (99m)Tc-Hydroxamamide Complexes. Sci Rep 2016; 6:25990. [PMID: 27181612 PMCID: PMC4867616 DOI: 10.1038/srep25990] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 04/26/2016] [Indexed: 11/26/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA), characterized by the deposition of amyloid aggregates in the walls of cerebral vasculature, is a major factor in intracerebral hemorrhage and vascular cognitive impairment and is also associated closely with Alzheimer’s disease (AD). We previously reported 99mTc-hydroxamamide (99mTc-Ham) complexes with a bivalent amyloid ligand showing high binding affinity for β-amyloid peptide (Aβ(1–42)) aggregates present frequently in the form in AD. In this article, we applied them to CAA-specific imaging probes, and evaluated their utility for CAA-specific imaging. In vitro inhibition assay using Aβ(1–40) aggregates deposited mainly in CAA and a brain uptake study were performed for 99mTc-Ham complexes, and all 99mTc-Ham complexes with an amyloid ligand showed binding affinity for Aβ(1–40) aggregates and very low brain uptake. In vitro autoradiography of human CAA brain sections and ex vivo autoradiography of Tg2576 mice were carried out for bivalent 99mTc-Ham complexes ([99mTc]SB2A and [99mTc]BT2B), and they displayed excellent labeling of Aβ depositions in human CAA brain sections and high affinity and selectivity to CAA in transgenic mice. These results may offer new possibilities for the development of clinically useful CAA-specific imaging probes based on the 99mTc-Ham complex.
Collapse
|
15
|
Lee NJ, Song JM, Cho HJ, Sung YM, Lee T, Chung A, Hong SH, Cifelli JL, Rubinshtein M, Habib LK, Capule CC, Turner RS, Pak DTS, Yang J, Hoe HS. Hexa (ethylene glycol) derivative of benzothiazole aniline promotes dendritic spine formation through the RasGRF1-Ras dependent pathway. Biochim Biophys Acta Mol Basis Dis 2015; 1862:284-95. [PMID: 26675527 DOI: 10.1016/j.bbadis.2015.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/16/2015] [Accepted: 12/04/2015] [Indexed: 11/24/2022]
Abstract
Our recent study demonstrated that an amyloid-β binding molecule, BTA-EG4, increases dendritic spine number via Ras-mediated signaling. To potentially optimize the potency of the BTA compounds, we synthesized and evaluated an amyloid-β binding analog of BTA-EG4 with increased solubility in aqueous solution, BTA-EG6. We initially examined the effects of BTA-EG6 on dendritic spine formation and found that BTA-EG6-treated primary hippocampal neurons had significantly increased dendritic spine number compared to control treatment. In addition, BTA-EG6 significantly increased the surface level of AMPA receptors. Upon investigation into the molecular mechanism by which BTA-EG6 promotes dendritic spine formation, we found that BTA-EG6 may exert its effects on spinogenesis via RasGRF1-ERK signaling, with potential involvement of other spinogenesis-related proteins such as Cdc42 and CDK5. Taken together, our data suggest that BTA-EG6 boosts spine and synapse number, which may have a beneficial effect of enhancing neuronal and synaptic function in the normal healthy brain.
Collapse
Affiliation(s)
- Nathanael J Lee
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jung Min Song
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hyun-Ji Cho
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Cheomdan-ro, Dong-gu, Daegu 701-300, Republic of Korea
| | - You Me Sung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Taehee Lee
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Andrew Chung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Sung-Ha Hong
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jessica L Cifelli
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark Rubinshtein
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lila K Habib
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina C Capule
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - R Scott Turner
- Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Daniel T S Pak
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Cheomdan-ro, Dong-gu, Daegu 701-300, Republic of Korea.
| |
Collapse
|
16
|
Bullmann T, Seeger G, Stieler J, Hanics J, Reimann K, Kretzschmann TP, Hilbrich I, Holzer M, Alpár A, Arendt T. Tau phosphorylation-associated spine regression does not impair hippocampal-dependent memory in hibernating golden hamsters. Hippocampus 2015; 26:301-18. [DOI: 10.1002/hipo.22522] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/12/2015] [Accepted: 08/31/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Torsten Bullmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
- Frey Initiative Research Unit, RIKEN Quantitative Biology Center; Japan
| | - Gudrun Seeger
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Jens Stieler
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - János Hanics
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Katja Reimann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Tanja Petra Kretzschmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Isabel Hilbrich
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Max Holzer
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Alán Alpár
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Thomas Arendt
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| |
Collapse
|
17
|
Kim W, Noh H, Lee Y, Jeon J, Shanmugavadivu A, McPhie DL, Kim KS, Cohen BM, Seo H, Sonntag KC. MiR-126 Regulates Growth Factor Activities and Vulnerability to Toxic Insult in Neurons. Mol Neurobiol 2014; 53:95-108. [PMID: 25407931 DOI: 10.1007/s12035-014-8989-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/04/2014] [Indexed: 01/19/2023]
Abstract
Dysfunction of growth factor (GF) activities contributes to the decline and death of neurons during aging and in neurodegenerative diseases. In addition, neurons become more resistant to GF signaling with age. Micro (mi)RNAs are posttranscriptional regulators of gene expression that may be crucial to age- and disease-related changes in GF functions. MiR-126 is involved in regulating insulin/IGF-1/phosphatidylinositol-3-kinase (PI3K)/AKT and extracellular signal-regulated kinase (ERK) signaling, and we recently demonstrated a functional role of miR-126 in dopamine neuronal cell survival in models of Parkinson's disease (PD)-associated toxicity. Here, we show that elevated levels of miR-126 increase neuronal vulnerability to ubiquitous toxicity mediated by staurosporine (STS) or Alzheimer's disease (AD)-associated amyloid beta 1-42 peptides (Aβ1-42). The neuroprotective factors IGF-1, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and soluble amyloid precursor protein α (sAPPα) could diminish but not abrogate the toxic effects of miR-126. In miR-126 overexpressing neurons derived from Tg6799 familial AD model mice, we observed an increase in Aβ1-42 toxicity, but surprisingly, both Aβ1-42 and miR-126 promoted neurite sprouting. Pathway analysis revealed that miR-126 overexpression downregulated elements in the GF/PI3K/AKT and ERK signaling cascades, including AKT, GSK-3β, ERK, their phosphorylation, and the miR-126 targets IRS-1 and PIK3R2. Finally, inhibition of miR-126 was neuroprotective against both STS and Aβ1-42 toxicity. Our data provide evidence for a novel mechanism of regulating GF/PI3K signaling in neurons by miR-126 and suggest that miR-126 may be an important mechanistic link between metabolic dysfunction and neurotoxicity in general, during aging, and in the pathogenesis of specific neurological disorders, including PD and AD.
Collapse
Affiliation(s)
- Woori Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Haneul Noh
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Yenarae Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Jeha Jeon
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Arthi Shanmugavadivu
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Donna L McPhie
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Kwang-Soo Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Bruce M Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Kai C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223, 115 Mill Street, Belmont, MA, 02478, USA.
| |
Collapse
|
18
|
Zhang K, Zhao T, Huang X, Wu LY, Wu K, Zhu LL, Fan M. Notch1 mediates postnatal neurogenesis in hippocampus enhanced by intermittent hypoxia. Neurobiol Dis 2014; 64:66-78. [DOI: 10.1016/j.nbd.2013.12.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/23/2013] [Accepted: 12/12/2013] [Indexed: 12/17/2022] Open
|
19
|
Trafficking in neurons: Searching for new targets for Alzheimer's disease future therapies. Eur J Pharmacol 2013; 719:84-106. [DOI: 10.1016/j.ejphar.2013.07.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/11/2013] [Indexed: 11/22/2022]
|
20
|
Krezymon A, Richetin K, Halley H, Roybon L, Lassalle JM, Francès B, Verret L, Rampon C. Modifications of hippocampal circuits and early disruption of adult neurogenesis in the tg2576 mouse model of Alzheimer's disease. PLoS One 2013; 8:e76497. [PMID: 24086745 PMCID: PMC3785457 DOI: 10.1371/journal.pone.0076497] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 08/27/2013] [Indexed: 11/29/2022] Open
Abstract
At advanced stages of Alzheimer's disease, cognitive dysfunction is accompanied by severe alterations of hippocampal circuits that may largely underlie memory impairments. However, it is likely that anatomical remodeling in the hippocampus may start long before any cognitive alteration is detected. Using the well-described Tg2576 mouse model of Alzheimer's disease that develops progressive age-dependent amyloidosis and cognitive deficits, we examined whether specific stages of the disease were associated with the expression of anatomical markers of hippocampal dysfunction. We found that these mice develop a complex pattern of changes in their dentate gyrus with aging. Those include aberrant expression of neuropeptide Y and reduced levels of calbindin, reflecting a profound remodeling of inhibitory and excitatory circuits in the dentate gyrus. Preceding these changes, we identified severe alterations of adult hippocampal neurogenesis in Tg2576 mice. We gathered converging data in Tg2576 mice at young age, indicating impaired maturation of new neurons that may compromise their functional integration into hippocampal circuits. Thus, disruption of adult hippocampal neurogenesis occurred before network remodeling in this mouse model and therefore may account as an early event in the etiology of Alzheimer's pathology. Ultimately, both events may constitute key components of hippocampal dysfunction and associated cognitive deficits occurring in Alzheimer's disease.
Collapse
Affiliation(s)
- Alice Krezymon
- Université de Toulouse (UPS) Centre de Recherches sur la Cognition Animale, Toulouse, France
- Centre National de la Recherche Scientifique (CNRS) Centre de Recherches sur la Cognition Animale, Toulouse, France
| | - Kevin Richetin
- Université de Toulouse (UPS) Centre de Recherches sur la Cognition Animale, Toulouse, France
- Centre National de la Recherche Scientifique (CNRS) Centre de Recherches sur la Cognition Animale, Toulouse, France
| | - Hélène Halley
- Université de Toulouse (UPS) Centre de Recherches sur la Cognition Animale, Toulouse, France
- Centre National de la Recherche Scientifique (CNRS) Centre de Recherches sur la Cognition Animale, Toulouse, France
| | - Laurent Roybon
- Multi Park, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Jean-Michel Lassalle
- Université de Toulouse (UPS) Centre de Recherches sur la Cognition Animale, Toulouse, France
- Centre National de la Recherche Scientifique (CNRS) Centre de Recherches sur la Cognition Animale, Toulouse, France
| | - Bernard Francès
- Université de Toulouse (UPS) Centre de Recherches sur la Cognition Animale, Toulouse, France
- Centre National de la Recherche Scientifique (CNRS) Centre de Recherches sur la Cognition Animale, Toulouse, France
| | - Laure Verret
- Université de Toulouse (UPS) Centre de Recherches sur la Cognition Animale, Toulouse, France
- Centre National de la Recherche Scientifique (CNRS) Centre de Recherches sur la Cognition Animale, Toulouse, France
| | - Claire Rampon
- Université de Toulouse (UPS) Centre de Recherches sur la Cognition Animale, Toulouse, France
- Centre National de la Recherche Scientifique (CNRS) Centre de Recherches sur la Cognition Animale, Toulouse, France
| |
Collapse
|
21
|
Hafner A, Glavan G, Obermajer N, Živin M, Schliebs R, Kos J. Neuroprotective role of γ-enolase in microglia in a mouse model of Alzheimer's disease is regulated by cathepsin X. Aging Cell 2013; 12:604-14. [PMID: 23621429 DOI: 10.1111/acel.12093] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2013] [Indexed: 12/20/2022] Open
Abstract
γ-Enolase is a neurotrophic-like factor promoting growth, differentiation, survival and regeneration of neurons. Its neurotrophic activity is regulated by cysteine protease cathepsin X which cleaves the C-terminal end of the molecule. We have investigated the expression and colocalization of γ-enolase and cathepsin X in brains of Tg2576 mice overexpressing amyloid precursor protein. In situ hybridization of γ-enolase and cathepsin X revealed that mRNAs for both enzymes were expressed abundantly around amyloid plaques. Immunostaining demonstrated that the C-terminally cleaved form of γ-enolase was present in the immediate plaque vicinity, whereas the intact form, exhibiting neurotrophic activity, was observed in microglia cells in close proximity to senile plaque. The upregulation of γ-enolase in microglial cells in response to amyloid-β peptide (Aβ) was confirmed in mouse microglial cell line EOC 13.31 and primary microglia and medium enriched with γ-enolase proved to be neuroprotective against Aβ toxicity; however, the effect was reversed by cathepsin X proteolytic activity. These results demonstrate an upregulation of γ-enolase in microglia cells surrounding amyloid plaques in Tg2576 transgenic mice and demonstrate its neuroprotective role in amyloid-β-related neurodegeneration.
Collapse
Affiliation(s)
- Anja Hafner
- Department of Pharmaceutical Biology Faculty of Pharmacy University of Ljubljana Askerceva 7Ljubljana 1000Slovenia
| | - Gordana Glavan
- Institute of Pathophysiology Medical faculty University of Ljubljana Zaloska 4Ljubljana 1000Slovenia
- Department of Biology Biotechnical faculty University of Ljubljana Vecna pot 11Ljubljana 1000Slovenia
| | - Nataša Obermajer
- Department of Pharmaceutical Biology Faculty of Pharmacy University of Ljubljana Askerceva 7Ljubljana 1000Slovenia
- Department of Biotechnology Jožef Stefan Institute Jamova 39Ljubljana 1000Slovenia
| | - Marko Živin
- Institute of Pathophysiology Medical faculty University of Ljubljana Zaloska 4Ljubljana 1000Slovenia
| | - Reinhard Schliebs
- Department of Neurochemistry Paul Flechsig Institute for Brain Research University of Leipzig Jahnallee 59Leipzig 04109Germany
| | - Janko Kos
- Department of Pharmaceutical Biology Faculty of Pharmacy University of Ljubljana Askerceva 7Ljubljana 1000Slovenia
- Department of Biotechnology Jožef Stefan Institute Jamova 39Ljubljana 1000Slovenia
| |
Collapse
|
22
|
Daulatzai MA. Neurotoxic Saboteurs: Straws that Break the Hippo’s (Hippocampus) Back Drive Cognitive Impairment and Alzheimer’s Disease. Neurotox Res 2013; 24:407-59. [DOI: 10.1007/s12640-013-9407-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/06/2013] [Accepted: 06/17/2013] [Indexed: 12/29/2022]
|
23
|
RanBP9 aggravates synaptic damage in the mouse brain and is inversely correlated to spinophilin levels in Alzheimer's brain synaptosomes. Cell Death Dis 2013; 4:e667. [PMID: 23764848 PMCID: PMC3698550 DOI: 10.1038/cddis.2013.183] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We previously demonstrated that overexpression of RanBP9 led to enhanced Aβ generation in a variety of cell lines and primary neuronal cultures, and subsequently, we confirmed increased amyloid plaque burden in a mouse model of Alzheimer's disease (AD). In the present study, we found striking reduction of spinophilin protein levels when RanBP9 is overexpressed. At 12 months of age, we found spinophilin levels reduced by 70% (P<0.001) in the cortex of APΔE9/RanBP9 mice compared with that in wild-type (WT) controls. In the hippocampus, the spinophilin levels were reduced by 45% (P<0.01) in the APΔE9/RanBP9 mice. Spinophilin immunoreactivity was also reduced by 22% (P<0.01) and 12% (P<0.05) in the cortex of APΔE9/RanBP9 and APΔE9 mice, respectively. In the hippocampus, the reductions were 27% (P<0.001) and 14% (P<0.001) in the APΔE9/RanBP9 and APΔE9 mice, respectively. However, in the cerebellum, spinophilin levels were not altered in either APΔE9 or APΔE9/RanBP9 mice. Additionally, synaptosomal functional integrity was reduced under basal conditions by 39% (P<0.001) in the APΔE9/RanBP9 mice and ∼23% (P<0.001) in the APΔE9 mice compared with that in WT controls. Under ATP- and KCl-stimulated conditions, we observed higher mitochondrial activity in the WT and APΔE9 mice, but lower in the APΔE9/RanBP9 mice. Significantly, we confirmed the inverse relationship between RanBP9-N60 and spinophilin in the synaptosomes of Alzheimer's brains. More importantly, both APΔE9 and APΔE9/RanBP9 mice showed impaired learning and memory skills compared to WT controls. These data suggest that RanBP9 might play a crucial role in the loss of spines and synapses in AD.
Collapse
|
24
|
Amyloid plaque formation precedes dendritic spine loss. Acta Neuropathol 2012; 124:797-807. [PMID: 22993126 PMCID: PMC3508278 DOI: 10.1007/s00401-012-1047-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 09/07/2012] [Accepted: 09/09/2012] [Indexed: 11/01/2022]
Abstract
Amyloid-beta plaque deposition represents a major neuropathological hallmark of Alzheimer's disease. While numerous studies have described dendritic spine loss in proximity to plaques, much less is known about the kinetics of these processes. In particular, the question as to whether synapse loss precedes or follows plaque formation remains unanswered. To address this question, and to learn more about the underlying kinetics, we simultaneously imaged amyloid plaque deposition and dendritic spine loss by applying two-photon in vivo microscopy through a cranial window in double transgenic APPPS1 mice. As a result, we first observed that the rate of dendritic spine loss in proximity to plaques is the same in both young and aged animals. However, plaque size only increased significantly in the young cohort, indicating that spine loss persists even many months after initial plaque appearance. Tracking the fate of individual spines revealed that net spine loss is caused by increased spine elimination, with the rate of spine formation remaining constant. Imaging of dendritic spines before and during plaque formation demonstrated that spine loss around plaques commences at least 4 weeks after initial plaque formation. In conclusion, spine loss occurs, shortly but with a significant time delay, after the birth of new plaques, and persists in the vicinity of amyloid plaques over many months. These findings hence give further hope to the possibility that there is a therapeutic window between initial amyloid plaque deposition and the onset of structural damage at spines.
Collapse
|
25
|
Stress-induced grey matter loss determined by MRI is primarily due to loss of dendrites and their synapses. Mol Neurobiol 2012; 47:645-61. [PMID: 23138690 DOI: 10.1007/s12035-012-8365-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 10/18/2012] [Indexed: 12/24/2022]
Abstract
Stress, unaccompanied by signs of post-traumatic stress disorder, is known to decrease grey matter volume (GMV) in the anterior cingulate cortex (ACC) and hippocampus but not the amygdala in humans. We sought to determine if this was the case in stressed mice using high-resolution magnetic resonance imaging (MRI) and to identify the cellular constituents of the grey matter that quantitatively give rise to such changes. Stressed mice showed grey matter losses of 10 and 15 % in the ACC and hippocampus, respectively but not in the amygdala or the retrosplenial granular area (RSG). Concurrently, no changes in the number or volumes of the somas of neurons, astrocytes or oligodendrocytes were detected. A loss of synaptic spine density of up to 60 % occurred on different-order dendrites in the ACC and hippocampus (CA1) but not in the amygdala or RSG. The loss of spines was accompanied by decreases in cumulative dendritic length of neurons of over 40 % in the ACC and hippocampus (CA1) giving rise to decreases in volume of dendrites of 2.6 mm(3) for the former and 0.6 mm(3) for the latter, with no change in the amygdala or RSG. These values are similar to the MRI-determined loss of GMV following stress of 3.0 and 0.8 mm(3) in ACC and hippocampus, respectively, with no changes in the amygdala or RSG. This quantitative study is the first to relate GMV changes in the cortex measured with MRI to volume changes in cellular constituents of the grey matter.
Collapse
|
26
|
Pozueta J, Lefort R, Shelanski ML. Synaptic changes in Alzheimer's disease and its models. Neuroscience 2012; 251:51-65. [PMID: 22687952 DOI: 10.1016/j.neuroscience.2012.05.050] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 05/14/2012] [Accepted: 05/17/2012] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder characterized by a progressive loss of cognition and the presence of two hallmark lesions, senile plaques (SP) and neurofibrillary tangles (NFT), which result from the accumulation and deposition of the β-amyloid peptide (Aβ) and the aggregation of hyperphosphorylated tau protein, respectively. Initially, it was thought that Aβ fibrils, which make up SP, were the root cause of the massive neurodegeneration usual found in AD brains. Over time, the longstanding emphasis on fibrillar Aβ deposits and neuronal death slowly gave way to a new paradigm involving soluble oligomeric forms of Aβ, which play a prominent role in triggering the cognitive deficits by specifically targeting synapses and disrupting synaptic signaling pathways. While this paradigm is widely accepted today in the AD field, the molecular details have not been fully elucidated. In this review, we address some of the important evidence, which has led to the Aβ oligomer-centric hypothesis as well as some of the key findings concerning the effects of Aβ oligomers on synapses at a morphological and functional level. Understanding how Aβ oligomers target synapses provides an important framework for ongoing AD research, which can lead to the development of successful therapeutic strategies designed to alter or perhaps reverse the course of the disease.
Collapse
Affiliation(s)
- J Pozueta
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain and Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, United States
| | | | | |
Collapse
|
27
|
Spines, plasticity, and cognition in Alzheimer's model mice. Neural Plast 2011; 2012:319836. [PMID: 22203915 PMCID: PMC3238410 DOI: 10.1155/2012/319836] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 10/27/2011] [Indexed: 01/03/2023] Open
Abstract
The pathological hallmarks of Alzheimer's disease (AD)--widespread synaptic and neuronal loss and the pathological accumulation of amyloid-beta peptide (Aβ) in senile plaques, as well as hyperphosphorylated tau in neurofibrillary tangles--have been known for many decades, but the links between AD pathology and dementia and effective therapeutic strategies remain elusive. Transgenic mice have been developed based on rare familial forms of AD and frontotemporal dementia, allowing investigators to test in detail the structural, functional, and behavioral consequences of AD-associated pathology. Here, we review work on transgenic AD models that investigate the degeneration of dendritic spine structure, synaptic function, and cognition. Together, these data support a model of AD pathogenesis in which soluble Aβ initiates synaptic dysfunction and loss, as well as pathological changes in tau, which contribute to both synaptic and neuronal loss. These changes in synapse structure and function as well as frank synapse and neuronal loss contribute to the neural system dysfunction which causes cognitive deficits. Understanding the underpinnings of dementia in AD will be essential to develop and evaluate therapeutic approaches for this widespread and devastating disease.
Collapse
|
28
|
Jung CKE, Herms J. Role of APP for dendritic spine formation and stability. Exp Brain Res 2011; 217:463-70. [PMID: 22094714 DOI: 10.1007/s00221-011-2939-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 11/03/2011] [Indexed: 11/27/2022]
Abstract
The amyloid precursor protein (APP) is transported in high amounts to the presynaptic endings where its function is still unknown. Several studies indicate that lack of APP or its overexpression affects the number of dendritic spines, the postsynaptic compartment of excitatory synapses. Since synapse loss has been identified as one of the most important structural correlates of cognitive decline in Alzheimer's diseases (AD), the physiological function of APP at synapses, specifically at dendritic spines, has come into focus in AD research. This review intends to give an overview of the very controversial results on APP expression on dendritic spine number in the mouse brain.
Collapse
Affiliation(s)
- Christian K E Jung
- Center of Neuropathology and Prion Research, Ludwig Maximilians Universität, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | | |
Collapse
|
29
|
Babus LW, Little EM, Keenoy KE, Minami SS, Chen E, Song JM, Caviness J, Koo SY, Pak DTS, Rebeck GW, Turner RS, Hoe HS. Decreased dendritic spine density and abnormal spine morphology in Fyn knockout mice. Brain Res 2011; 1415:96-102. [PMID: 21872217 DOI: 10.1016/j.brainres.2011.07.059] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 07/14/2011] [Accepted: 07/28/2011] [Indexed: 10/17/2022]
Abstract
Fyn is a Src-family tyrosine kinase that affects long term potentiation (LTP), synapse formation, and learning and memory. Fyn is also implicated in dendritic spine formation both in vitro and in vivo. However, whether Fyn's regulation of dendritic spine formation is brain-region specific and age-dependent is unknown. In the present study, we systematically examined whether Fyn altered dendritic spine density and morphology in the cortex and hippocampus and if these effects were age-dependent. We found that Fyn knockout mice trended toward a decrease in dendritic spine density in cortical layers II/III, but not in the hippocampus, at 1 month of age. Additionally, Fyn knockout mice had significantly decreased dendritic spine density in both the cortex and hippocampus at 3 months and 1 year, and Fyn's effect on dendritic spine density was age-dependent in the hippocampus. Moreover, Fyn knockout mice had wider spines at the three time points (1 month, 3 months, 1 year) in the cortex. These findings suggest that Fyn regulates dendritic spine number and morphology over time and provide further support for Fyn's role in maintaining proper synaptic function in vivo.
Collapse
Affiliation(s)
- Lenard W Babus
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC 20057-1464, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Metal ionophore treatment restores dendritic spine density and synaptic protein levels in a mouse model of Alzheimer's disease. PLoS One 2011; 6:e17669. [PMID: 21412423 PMCID: PMC3055881 DOI: 10.1371/journal.pone.0017669] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 02/10/2011] [Indexed: 12/28/2022] Open
Abstract
We have previously demonstrated that brief treatment of APP transgenic mice with metal ionophores (PBT2, Prana Biotechnology) rapidly and markedly improves learning and memory. To understand the potential mechanisms of action underlying this phenomenon we examined hippocampal dendritic spine density, and the levels of key proteins involved in learning and memory, in young (4 months) and old (14 months) female Tg2576 mice following brief (11 days) oral treatment with PBT2 (30 mg/kg/d). Transgenic mice exhibited deficits in spine density compared to littermate controls that were significantly rescued by PBT2 treatment in both the young (+17%, p<0.001) and old (+32%, p<0.001) animals. There was no effect of PBT2 on spine density in the control animals. In the transgenic animals, PBT2 treatment also resulted in significant increases in brain levels of CamKII (+57%, p = 0.005), spinophilin (+37%, p = 0.04), NMDAR1A (+126%, p = 0.02), NMDAR2A (+70%, p = 0.05), pro-BDNF (+19%, p = 0.02) and BDNF (+19%, p = 0.04). While PBT2-treatment did not significantly alter neurite-length in vivo, it did increase neurite outgrowth (+200%, p = 0.006) in cultured cells, and this was abolished by co-incubation with the transition metal chelator, diamsar. These data suggest that PBT2 may affect multiple aspects of snaptic health/efficacy. In Alzheimer's disease therefore, PBT2 may restore the uptake of physiological metal ions trapped within extracellular β-amyloid aggregates that then induce biochemical and anatomical changes to improve cognitive function.
Collapse
|
31
|
Alpár A, Ueberham U, Lendvai D, Naumann N, Rohn S, Gáti G, Arendt T, Gärtner U. Activity-induced dendrite and dendritic spine development in human amyloid precursor protein transgenic mice. Int J Dev Neurosci 2011; 29:107-14. [PMID: 21277971 DOI: 10.1016/j.ijdevneu.2011.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 12/29/2010] [Accepted: 01/10/2011] [Indexed: 10/18/2022] Open
Abstract
The amyloid precursor protein is essential for proper neuronal function but an imbalance in processing or metabolism or its overexpression lead to severe malfunction of the brain. The present study focused on dendritic morphology of hippocampal neurons in mice overexpressing the wild-type human amyloid precursor protein (hAPP). In addition, we examined whether enhanced physical activity may affect hAPP-related morphological changes. Overexpression of hAPP resulted in significant enlargement of dendrites, especially within the basal dendritic field but had no effect on spine density. Enhanced physical activity only moderately potentiated hAPP induced changes in dendritic size. Physical activity dependent increases in spine density were, however, augmented by hAPP overexpression. The results suggest that enhanced levels of wild-type hAPP do not result in degenerative changes of neuronal morphology, but rather promote dendritic growth.
Collapse
Affiliation(s)
- Alán Alpár
- Department of Anatomy, Histology and Embryology, Semmelweis University Medical School, Tűzoltó u. 58, H-1450 Budapest, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
AbstractIn neuronal circuits, excitatory synaptic transmission predominantly occurs at postsynaptic protrusions called dendritic spines. Spines are highly plastic structures capable of formation, enlargement, shrinkage, and elimination over time. Individual spine morphology is widely variable, and evidence suggests these differences in morphology are relevant to spine function. Recent reports provide evidence that spine structural plasticity underlies functional synaptic changes, including those seen in animal models of learning and memory plasticity. Conversely, impairments in cognitive functions, such as those commonly seen in aging, have recently been linked to and correlated with alterations in spine density and morphology. In addition, dendritic spine density and morphology also appear to be altered in various transgenic animal models of neurodegenerative diseases. Ultimately, an understanding of the synaptic basis of age- and disease-related cognitive impairments may lead to the development of drug treatments that can restore or protect synaptic profiles in neural circuits that mediate cognition.
Collapse
|
33
|
Calcineurin inhibition with FK506 ameliorates dendritic spine density deficits in plaque-bearing Alzheimer model mice. Neurobiol Dis 2010; 41:650-4. [PMID: 21134458 DOI: 10.1016/j.nbd.2010.11.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 10/19/2010] [Accepted: 11/26/2010] [Indexed: 11/20/2022] Open
Abstract
Synapse loss is the strongest correlate of cognitive decline in Alzheimer's disease, and synapses are an attractive therapeutic target due to their plastic nature that allows for potential recovery with intervention. We have previously demonstrated in transgenic mice that form senile plaques that dendrites surrounding plaques become dystrophic and lose postsynaptic dendritic spines. Furthermore, we found strong evidence that plaque-associated dendritic changes are mediated by calcineurin, a calcium-dependent phosphatase involved in cell signaling, using in vitro models and genetically encoded inhibitors in mouse models. In this study, we pharmacologically inhibited calcineurin with FK506 treatment to test the hypothesis that calcineurin inhibition will allow recovery of plaque-associated synapse loss. We found that in plaque bearing transgenic mice, short term (1 week) FK506 treatment results in an amelioration of dendritic spine loss. We also observe an effect on spine morphology in wild-type mice with FK506 treatment. These data show that systemic FK506 administration, and hence calcineurin inhibition, may be neuroprotective for amyloid beta induced synaptic alterations.
Collapse
|
34
|
Sperling RA, Dickerson BC, Pihlajamaki M, Vannini P, LaViolette PS, Vitolo OV, Hedden T, Becker JA, Rentz DM, Selkoe DJ, Johnson KA. Functional alterations in memory networks in early Alzheimer's disease. Neuromolecular Med 2010; 12:27-43. [PMID: 20069392 PMCID: PMC3036844 DOI: 10.1007/s12017-009-8109-7] [Citation(s) in RCA: 416] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 12/02/2009] [Indexed: 12/22/2022]
Abstract
The hallmark clinical symptom of early Alzheimer's disease (AD) is episodic memory impairment. Recent functional imaging studies suggest that memory function is subserved by a set of distributed networks, which include both the medial temporal lobe (MTL) system and the set of cortical regions collectively referred to as the default network. Specific regions of the default network, in particular, the posteromedial cortices, including the precuneus and posterior cingulate, are selectively vulnerable to early amyloid deposition in AD. These regions are also thought to play a key role in both memory encoding and retrieval, and are strongly functionally connected to the MTL. Multiple functional magnetic resonance imaging (fMRI) studies during memory tasks have revealed alterations in these networks in patients with clinical AD. Similar functional abnormalities have been detected in subjects at-risk for AD, including those with genetic risk and older individuals with mild cognitive impairment. Recently, we and other groups have found evidence of functional alterations in these memory networks even among cognitively intact older individuals with occult amyloid pathology, detected by PET amyloid imaging. Taken together, these findings suggest that the pathophysiological process of AD exerts specific deleterious effects on these distributed memory circuits, even prior to clinical manifestations of significant memory impairment. Interestingly, some of the functional alterations seen in prodromal AD subjects have taken the form of increases in activity relative to baseline, rather than a loss of activity. It remains unclear whether these increases in fMRI activity may be compensatory to maintain memory performance in the setting of early AD pathology or instead, represent evidence of excitotoxicity and impending neuronal failure. Recent studies have also revealed disruption of the intrinsic connectivity of these networks observable even during the resting state in early AD and asymptomatic individuals with high amyloid burden. Research is ongoing to determine if these early network alterations will serve as sensitive predictors of clinical decline, and eventually, as markers of pharmacological response to potential disease-modifying treatments for AD.
Collapse
Affiliation(s)
- Reisa A Sperling
- Department of Neurology, Center for Alzheimer's Research and Treatment, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Synaptotagmins (Syts) are transmembrane proteins involved in the regulation of membrane trafficking. Here, we summarize literature data that provide growing evidence that several Syts are involved in the pathophysiological mechanisms of temporal lobe epilepsy and Parkinson's disease, as well as few reports related to brain ischemia and Alzheimer's disease (AD). We also report new data from our laboratories, showing changes of the expression of several Syts in Tg2576 mouse model of AD that may be related to neuroinflammation surrounding the beta-amyloid plaques. Furthermore, we demonstrate N-methyl-D-aspartate receptor-mediated upregulation of Syt 4 mRNA in a model of excitotoxic striatal lesion induced by unilateral striatal injection of quinolinic acid, associating the upregulation of Syt 4 with mechanisms of excitotoxicity. We propose that pharmacological manipulation of Syt expression in animal models of neurodegeneration should be further explored, as it may help to clarify the role of individual Syt isoforms in the regulation of membrane trafficking in neurodegeneration.
Collapse
Affiliation(s)
- Gordana Glavan
- Medical Faculty, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | | | | |
Collapse
|
36
|
Abstract
The three human alleles of apolipoprotein E (APOE) differentially influence outcome after CNS injury and affect one's risk of developing Alzheimer's disease (AD). It remains unclear how ApoE isoforms contribute to various AD-related pathological changes (e.g., amyloid plaques and synaptic and neuron loss). Here, we systematically examined whether apoE isoforms (E2, E3, E4) exhibit differential effects on dendritic spine density and morphology in APOE targeted replacement (TR) mice, which lack AD pathological changes. Using Golgi staining, we found age-dependent effects of APOE4 on spine density in the cortex. The APOE4 TR mice had significantly reduced spine density at three independent time points (4 weeks, 3 months, and 1 year, 27.7% +/- 7.4%, 24.4% +/- 8.6%, and 55.6% +/- 10.5%, respectively) compared with APOE3 TR mice and APOE2 TR mice. Additionally, in APOE4 TR mice, shorter spines were evident compared with other APOE TR mice at 1 year. APOE2 TR mice exhibited longer spines as well as significantly increased apical dendritic arborization in the cortex compared with APOE4 and APOE3 TR mice at 4 weeks. However, there were no differences in spine density across APOE genotypes in hippocampus. These findings demonstrate that apoE isoforms differentially affect dendritic complexity and spine formation, suggesting a role for APOE genotypes not only in acute and chronic brain injuries including AD, but also in normal brain functions.
Collapse
|
37
|
Reversal of long-term dendritic spine alterations in Alzheimer disease models. Proc Natl Acad Sci U S A 2009; 106:16877-82. [PMID: 19805389 DOI: 10.1073/pnas.0908706106] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synapse loss is strongly correlated with cognitive impairment in Alzheimer's disease (AD). We have previously reported the loss of dendritic spines and the presence of dystrophic neurites in both the hippocampi of transgenic mice overexpressing amyloid precursor protein (APP) and in the human brain affected with AD. In the studies reported here we have asked whether the acute alterations in dendritic spines induced by Abeta, as well as the chronic loss of spine density seen in hAPP transgenic mice, are reversible by treatments that restore the cAMP/PKA/CREB signaling pathway or proteasome function to control levels. The results show that both rolipram and TAT-HA-Uch-L1 restore spine density to near control conditions, even in elderly mice. The results suggest that changes in dendritic structure and function that occur after Abeta elevation are reversible even after long periods of time, and that one could envision therapeutic approaches to AD based on this restoration that could work independently of therapies aimed at lowering Abeta levels in the brain.
Collapse
|
38
|
Knafo S, Venero C, Merino-Serrais P, Fernaud-Espinosa I, Gonzalez-Soriano J, Ferrer I, Santpere G, DeFelipe J. Morphological alterations to neurons of the amygdala and impaired fear conditioning in a transgenic mouse model of Alzheimer's disease. J Pathol 2009; 219:41-51. [PMID: 19449368 DOI: 10.1002/path.2565] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
39
|
Kwan AC, Duff K, Gouras GK, Webb WW. Optical visualization of Alzheimer's pathology via multiphoton-excited intrinsic fluorescence and second harmonic generation. OPTICS EXPRESS 2009; 17:3679-89. [PMID: 19259208 PMCID: PMC2977950 DOI: 10.1364/oe.17.003679] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Intrinsic optical emissions, such as autofluorescence and second harmonic generation (SHG), are potentially useful for functional fluorescence imaging and biomedical disease diagnosis for neurodegenerative diseases such as Alzheimer's disease (AD). Here, using multiphoton and SHG microscopy, we identified sources of intrinsic emissions in ex vivo, acute brain slices from AD transgenic mouse models. We observed autofluorescence and SHG at senile plaques as well as characterized their emission spectra. The utility of intrinsic emissions was demonstrated by imaging senile plaque autofluorescence in conjunction with SHG from microtubule arrays to assess the polarity of microtubules near pathological lesions. Our results suggest that tissues from AD transgenic models contain distinct intrinsic emissions, which can provide valuable information about the disease mechanisms.
Collapse
Affiliation(s)
- Alex C. Kwan
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York 14853, USA
| | - Karen Duff
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York City, New York 10032, USA
| | - Gunnar K. Gouras
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York City, New York 10021, USA
| | - Watt W. Webb
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
40
|
Rocher AB, Kinson MS, Luebke JI. Significant structural but not physiological changes in cortical neurons of 12-month-old Tg2576 mice. Neurobiol Dis 2008; 32:309-18. [PMID: 18721884 DOI: 10.1016/j.nbd.2008.07.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 06/26/2008] [Accepted: 07/25/2008] [Indexed: 11/29/2022] Open
Abstract
Amyloid-beta (Abeta) plays a key role in the etiology of Alzheimer's disease, and pyramidal cell dendrites exposed to Abeta exhibit dramatic structural alterations, including reduced dendritic spine densities. To determine whether such structural alterations lead to electrophysiological changes, whole-cell patch clamp recordings with biocytin filling were used to assess both the electrophysiological and morphological properties of layer 3 pyramidal cells in frontal cortical slices prepared from 12-month-old Tg2576 amyloid precursor protein (APP) mutant vs. wild-type (Wt) mice. Tg2576 cells exhibited significantly increased dendritic lengths and volumes and decreased spine densities, while the total number of spines was not different from Wt. Tg2576 and Wt cells did not differ with regard to passive membrane, action potential firing or glutamatergic spontaneous excitatory postsynaptic current properties. Thus, overexpression of mutated APP in young Tg2576 mice leads to significant changes in neuronal morphological properties which do not have readily apparent functional consequences.
Collapse
Affiliation(s)
- Anne B Rocher
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | |
Collapse
|
41
|
Knobloch M, Mansuy IM. Dendritic spine loss and synaptic alterations in Alzheimer's disease. Mol Neurobiol 2008; 37:73-82. [PMID: 18438727 DOI: 10.1007/s12035-008-8018-z] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Accepted: 03/28/2008] [Indexed: 01/04/2023]
Abstract
Dendritic spines are tiny protrusions along dendrites, which constitute major postsynaptic sites for excitatory synaptic transmission. These spines are highly motile and can undergo remodeling even in the adult nervous system. Spine remodeling and the formation of new synapses are activity-dependent processes that provide a basis for memory formation. A loss or alteration of these structures has been described in patients with neurodegenerative disorders such as Alzheimer's disease (AD), and in mouse models for these disorders. Such alteration is thought to be responsible for cognitive deficits long before or even in the absence of neuronal loss, but the underlying mechanisms are poorly understood. This review will describe recent findings and discoveries on the loss or alteration of dendritic spines induced by the amyloid beta (Abeta) peptide in the context of AD.
Collapse
Affiliation(s)
- Marlen Knobloch
- Division of Psychiatry Research, University of Zurich, 8008 Zurich, Switzerland
| | | |
Collapse
|
42
|
Nathalie Lacor P. Advances on the understanding of the origins of synaptic pathology in AD. Curr Genomics 2007; 8:486-508. [PMID: 19415125 PMCID: PMC2647163 DOI: 10.2174/138920207783769530] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 12/20/2007] [Accepted: 12/20/2007] [Indexed: 12/14/2022] Open
Abstract
Although Alzheimer's disease (AD) was first discovered a century ago, we are still facing a lack of definitive diagnosis during the patient's lifetime and are unable to prescribe a curative treatment. However, the past 10 years have seen a "revamping" of the main hypothesis about AD pathogenesis and the hope to foresee possible treatment. AD is no longer considered an irreversible disease. A major refinement of the classic beta-amyloid cascade describing amyloid fibrils as neurotoxins has been made to integrate the key scientific evidences demonstrating that the first pathological event occurring in AD early stages affects synaptic function and maintenance. A concept fully compatible with synapse loss being the best pathological correlate of AD rather than other described neuropathological hallmarks (amyloid plaques, neurofibrillary tangles or neuronal death). The notion that synaptic alterations might be reverted, thus offering a potential curability, was confirmed by immunotherapy experiments targeting beta-amyloid protein in transgenic AD mice in which cognitive functions were improved despite no reduction in the amyloid plaques burden. The updated amyloid cascade now integrates the synapse failure triggered by soluble Abeta-oligomers. Still no consensus has been reached on the most toxic Abeta conformations, neither on their site of production nor on their extra- versus intra-cellular actions. Evidence shows that soluble Abeta oligomers or ADDLs bind selectively to neurons at their synaptic loci, and trigger major changes in synapse composition and morphology, which ultimately leads to dendritic spine loss. However, the exact mechanism is not yet fully understood but is suspected to involve some membrane receptor(s).
Collapse
|
43
|
Alpár A, Ueberham U, Seeger G, Arendt T, Gärtner U. Effects of wild-type and mutant human amyloid precursor protein on cortical afferent network. Neuroreport 2007; 18:1247-50. [PMID: 17632276 DOI: 10.1097/wnr.0b013e3282202829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease is characterized by severe neuronal disintegration supposed to be partly associated with amyloid pathology. Recently, we described morphological alterations of pyramidal cell structure in transgenic mice expressing wild-type or mutant human amyloid precursor protein (hAPP) (strains B6-Py8.9 and Tg2576), which are unrelated to direct plaque-associated changes. In this study, we focused on the pattern of cortical afferent connections in these transgenic mice. The quantity of cholinergic afferents is increased in both transgenic lines. Glutamatergic intra- and interhemispheric afferents are augmented in B6-Py8.9 mice but decreased in Tg2576 mice. Furthermore, perisomatic inhibition of pyramidal neurons was found to be reduced in Tg2576 mice. Findings suggest different effects of wild-type and mutant hAPP on neuronal connectivity.
Collapse
Affiliation(s)
- Alán Alpár
- Department of Anatomy, Histology and Embryology, Semmelweis University Medical School, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
44
|
Dickstein DL, Kabaso D, Rocher AB, Luebke JI, Wearne SL, Hof PR. Changes in the structural complexity of the aged brain. Aging Cell 2007; 6:275-84. [PMID: 17465981 PMCID: PMC2441530 DOI: 10.1111/j.1474-9726.2007.00289.x] [Citation(s) in RCA: 273] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Structural changes of neurons in the brain during aging are complex and not well understood. Neurons have significant homeostatic control of essential brain functions, including synaptic excitability, gene expression, and metabolic regulation. Any deviations from the norm can have severe consequences as seen in aging and injury. In this review, we present some of the structural adaptations that neurons undergo throughout normal and pathological aging and discuss their effects on electrophysiological properties and cognition. During aging, it is evident that neurons undergo morphological changes such as a reduction in the complexity of dendrite arborization and dendritic length. Spine numbers are also decreased, and because spines are the major sites for excitatory synapses, changes in their numbers could reflect a change in synaptic densities. This idea has been supported by studies that demonstrate a decrease in the overall frequency of spontaneous glutamate receptor-mediated excitatory responses, as well as a decrease in the levels of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid and N-methyl-d-aspartate receptor expression. Other properties such as gamma-aminobutyric acid A receptor-mediated inhibitory responses and action potential firing rates are both significantly increased with age. These findings suggest that age-related neuronal dysfunction, which must underlie observed decline in cognitive function, probably involves a host of other subtle changes within the cortex that could include alterations in receptors, loss of dendrites, and spines and myelin dystrophy, as well as the alterations in synaptic transmission. Together these multiple alterations in the brain may constitute the substrate for age-related loss of cognitive function.
Collapse
Affiliation(s)
- Dara L Dickstein
- Department of Neuroscience, Mount Sinai School of Medicine, New York 10029, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Bai W, Zhou X, Ji L, Cheng J, Wong STC. Automatic dendritic spine analysis in two-photon laser scanning microscopy images. Cytometry A 2007; 71:818-26. [PMID: 17654649 DOI: 10.1002/cyto.a.20431] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Dendritic spine expression plays an important role in the central nervous system. Modern fluorescence microscopy and green fluorescent protein technology have facilitated the research on spines. To quantitatively analyze the spines in fluorescence microscopy images, an automatic dendritic spine analysis method is proposed. Because of the limit of axial resolution, our method is designed to process the projection image along the z-axis and analyze the lateral spines. The method can automatically extract the dendrite centerlines and segment the spines along the dendrites according to width-based criteria. The criteria utilize a common morphological feature of the spines. It can detect some shapes of spines missed by previous methods. In addition, the proposed method is automatic once a few parameters are set. Spine numbers, lengths, and densities, which biologists are interested in, are analyzed both manually and automatically. The results of the two methods match well. The proposed method provides automatic and accurate dendritic spine analysis. It can serve as a useful tool for spine image analysis to avoid tedious manual labor.
Collapse
Affiliation(s)
- Wenjia Bai
- Bioinformatics Division, TNLIST and Department of Automation, Tsinghua University, Beijing, PR China
| | | | | | | | | |
Collapse
|