1
|
Caiola HO, Wu Q, Li J, Wang XF, Soni S, Monahan K, Wagner GC, Pang ZP, Zhang H. Neuronal connectivity, behavioral, and transcriptional alterations associated with the loss of MARK2. FASEB J 2024; 38:e70124. [PMID: 39436150 DOI: 10.1096/fj.202400454r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/03/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Neuronal connectivity is essential for adaptive brain responses and can be modulated by dendritic spine plasticity and the intrinsic excitability of individual neurons. Dysregulation of these processes can lead to aberrant neuronal activity, which has been associated with numerous neurological disorders including autism, epilepsy, and Alzheimer's disease. Nonetheless, the molecular mechanisms underlying abnormal neuronal connectivity remain unclear. We previously found that the serine/threonine kinase Microtubule Affinity Regulating Kinase 2 (MARK2), also known as Partitioning Defective 1b (Par1b), is important for the formation of dendritic spines in vitro. However, despite its genetic association with several neurological disorders, the in vivo impact of MARK2 on neuronal connectivity and cognitive functions remains unclear. Here, we demonstrate that the loss of MARK2 in vivo results in changes to dendritic spine morphology, which in turn leads to a decrease in excitatory synaptic transmission. Additionally, the loss of MARK2 produces substantial impairments in learning and memory, reduced anxiety, and defective social behavior. Notably, MARK2 deficiency results in heightened seizure susceptibility. Consistent with this observation, electrophysiological analysis of hippocampal slices indicates underlying neuronal hyperexcitability in MARK2-deficient neurons. Finally, RNAseq analysis reveals transcriptional changes in genes regulating synaptic transmission and ion homeostasis. These results underscore the in vivo role of MARK2 in governing synaptic connectivity, neuronal excitability, and cognitive functions.
Collapse
Affiliation(s)
- Hanna O Caiola
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Qian Wu
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Junlong Li
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Xue-Feng Wang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Shaili Soni
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Kevin Monahan
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey, USA
| | - George C Wagner
- Department of Psychology, Rutgers University, Piscataway, New Jersey, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| |
Collapse
|
2
|
Rentsch P, Ganesan K, Langdon A, Konen LM, Vissel B. Toward the development of a sporadic model of Alzheimer's disease: comparing pathologies between humanized APP and the familial J20 mouse models. Front Aging Neurosci 2024; 16:1421900. [PMID: 39040546 PMCID: PMC11260812 DOI: 10.3389/fnagi.2024.1421900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/03/2024] [Indexed: 07/24/2024] Open
Abstract
Background Finding successful therapies for individuals with Alzheimer's disease (AD) remains an ongoing challenge. One contributing factor is that the mouse models commonly used in preclinical research primarily mimic the familial form of AD, whereas the vast majority of human cases are sporadic. Accordingly, for a sporadic mouse model of AD, incorporating the multifactorial aspects of the disease is of utmost importance. Methods In the current study, we exposed humanized Aβ knock-in mice (hAβ-KI) to weekly low-dose lipopolysaccharide (LPS) injections until 24 weeks of age and compared the development of AD pathologies to the familial AD mouse model known as the J20 mice. Results At the early time point of 24 weeks, hAβ-KI mice and J20 mice exhibited spatial memory impairments in the Barnes maze. Strikingly, both hAβ-KI mice and J20 mice showed significant loss of dendritic spines when compared to WT controls, despite the absence of Aβ plaques in hAβ-KI mice at 24 weeks of age. Glial cell numbers remained unchanged in hAβ-KI mice compared to WT, and LPS exposure in hAβ-KI mice did not result in memory deficits and failed to exacerbate any other examined AD pathology. Conclusion The study highlights the potential of hAβ-KI mice as a model for sporadic AD, demonstrating early cognitive deficits and synaptic alterations despite no evidence of Aβ plaque formation. These findings underscore the importance of considering multifactorial influences in sporadic AD pathogenesis and the need for innovative models to advance our understanding and treatment strategies for this complex disease.
Collapse
Affiliation(s)
- Peggy Rentsch
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
- UNSW St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Kiruthika Ganesan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Alexander Langdon
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Lyndsey M. Konen
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
- UNSW St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
3
|
Tzavellas NP, Tsamis KI, Katsenos AP, Davri AS, Simos YV, Nikas IP, Bellos S, Lekkas P, Kanellos FS, Konitsiotis S, Labrakakis C, Vezyraki P, Peschos D. Firing Alterations of Neurons in Alzheimer's Disease: Are They Merely a Consequence of Pathogenesis or a Pivotal Component of Disease Progression? Cells 2024; 13:434. [PMID: 38474398 DOI: 10.3390/cells13050434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, yet its underlying causes remain elusive. The conventional perspective on disease pathogenesis attributes alterations in neuronal excitability to molecular changes resulting in synaptic dysfunction. Early hyperexcitability is succeeded by a progressive cessation of electrical activity in neurons, with amyloid beta (Aβ) oligomers and tau protein hyperphosphorylation identified as the initial events leading to hyperactivity. In addition to these key proteins, voltage-gated sodium and potassium channels play a decisive role in the altered electrical properties of neurons in AD. Impaired synaptic function and reduced neuronal plasticity contribute to a vicious cycle, resulting in a reduction in the number of synapses and synaptic proteins, impacting their transportation inside the neuron. An understanding of these neurophysiological alterations, combined with abnormalities in the morphology of brain cells, emerges as a crucial avenue for new treatment investigations. This review aims to delve into the detailed exploration of electrical neuronal alterations observed in different AD models affecting single neurons and neuronal networks.
Collapse
Affiliation(s)
- Nikolaos P Tzavellas
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Konstantinos I Tsamis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Andreas P Katsenos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Athena S Davri
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Yannis V Simos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Ilias P Nikas
- Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Stefanos Bellos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Panagiotis Lekkas
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Foivos S Kanellos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Spyridon Konitsiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Charalampos Labrakakis
- Department of Biological Applications and Technology, University of Ioannina, 451 10 Ioannina, Greece
| | - Patra Vezyraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | - Dimitrios Peschos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| |
Collapse
|
4
|
Chen J, Chen JS, Li S, Zhang F, Deng J, Zeng LH, Tan J. Amyloid Precursor Protein: A Regulatory Hub in Alzheimer's Disease. Aging Dis 2024; 15:201-225. [PMID: 37307834 PMCID: PMC10796103 DOI: 10.14336/ad.2023.0308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/08/2023] [Indexed: 06/14/2023] Open
Abstract
Decades of research have demonstrated an incontrovertible role of amyloid-β (Aβ) in the etiology of Alzheimer's disease (AD). However, the overemphasis on the pathological impacts of Aβ may obscure the role of its metabolic precursor, amyloid precursor protein (APP), as a significant hub in the occurrence and progression of AD. The complicated enzymatic processing, ubiquitous receptor-like properties, and abundant expression of APP in the brain, as well as its close links with systemic metabolism, mitochondrial function and neuroinflammation, imply that APP plays multifaceted roles in AD. In this review, we briefly describe the evolutionarily conserved biological characteristics of APP, including its structure, functions and enzymatic processing. We also discuss the possible involvement of APP and its enzymatic metabolites in AD, both detrimental and beneficial. Finally, we describe pharmacological agents or genetic approaches with the capability to reduce APP expression or inhibit its cellular internalization, which can ameliorate multiple aspects of AD pathologies and halt disease progression. These approaches provide a basis for further drug development to combat this terrible disease.
Collapse
Affiliation(s)
- Jiang Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Jun-Sheng Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Song Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Fengning Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Jie Deng
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Jun Tan
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Niraula S, Yan SS, Subramanian J. Amyloid Pathology Impairs Experience-Dependent Inhibitory Synaptic Plasticity. J Neurosci 2024; 44:e0702232023. [PMID: 38050105 PMCID: PMC10860629 DOI: 10.1523/jneurosci.0702-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023] Open
Abstract
Alzheimer's disease patients and mouse models exhibit aberrant neuronal activity and altered excitatory-to-inhibitory synaptic ratio. Using multicolor two-photon microscopy, we test how amyloid pathology alters the structural dynamics of excitatory and inhibitory synapses and their adaptation to altered visual experience in vivo in the visual cortex. We show that the baseline dynamics of mature excitatory synapses and their adaptation to visual deprivation are not altered in amyloidosis. Likewise, the baseline dynamics of inhibitory synapses are not affected. In contrast, visual deprivation fails to induce inhibitory synapse loss in amyloidosis, a phenomenon observed in nonpathological conditions. Intriguingly, inhibitory synapse loss associated with visual deprivation in nonpathological mice is accompanied by subtle broadening of spontaneous but not visually evoked calcium transients. However, such broadening does not manifest in the context of amyloidosis. We also show that excitatory and inhibitory synapse loss is locally clustered under the nonpathological state. In contrast, a fraction of synapse loss is not locally clustered in amyloidosis, indicating an impairment in inhibitory synapse adaptation to changes in excitatory synaptic activity.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
- Department of Surgery, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York 10032
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
6
|
Milham LT, Morris GP, Konen LM, Rentsch P, Avgan N, Vissel B. Quantification of AMPA receptor subunits and RNA editing-related proteins in the J20 mouse model of Alzheimer's disease by capillary western blotting. Front Mol Neurosci 2024; 16:1338065. [PMID: 38299128 PMCID: PMC10828003 DOI: 10.3389/fnmol.2023.1338065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction Accurate modelling of molecular changes in Alzheimer's disease (AD) dementia is crucial for understanding the mechanisms driving neuronal pathology and for developing treatments. Synaptic dysfunction has long been implicated as a mechanism underpinning memory dysfunction in AD and may result in part from changes in adenosine deaminase acting on RNA (ADAR) mediated RNA editing of the GluA2 subunit of AMPA receptors and changes in AMPA receptor function at the post synaptic cleft. However, few studies have investigated changes in proteins which influence RNA editing and notably, AD studies that focus on studying changes in protein expression, rather than changes in mRNA, often use traditional western blotting. Methods Here, we demonstrate the value of automated capillary western blotting to investigate the protein expression of AMPA receptor subunits (GluA1-4), the ADAR RNA editing proteins (ADAR1-3), and proteins known to regulate RNA editing (PIN1, WWP2, FXR1P, and CREB1), in the J20 AD mouse model. We describe extensive optimisation and validation of the automated capillary western blotting method, demonstrating the use of total protein to normalise protein load, in addition to characterising the optimal protein/antibody concentrations to ensure accurate protein quantification. Following this, we assessed changes in proteins of interest in the hippocampus of 44-week-old J20 AD mice. Results We observed an increase in the expression of ADAR1 p110 and GluA3 and a decrease in ADAR2 in the hippocampus of 44-week-old J20 mice. These changes signify a shift in the balance of proteins that play a critical role at the synapse. Regression analysis revealed unique J20-specific correlations between changes in AMPA receptor subunits, ADAR enzymes, and proteins that regulate ADAR stability in J20 mice, highlighting potential mechanisms mediating RNA-editing changes found in AD. Discussion Our findings in J20 mice generally reflect changes seen in the human AD brain. This study underlines the importance of novel techniques, like automated capillary western blotting, to assess protein expression in AD. It also provides further evidence to support the hypothesis that a dysregulation in RNA editing-related proteins may play a role in the initiation and/or progression of AD.
Collapse
Affiliation(s)
- Luke T. Milham
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Gary P. Morris
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Lyndsey M. Konen
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Peggy Rentsch
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Nesli Avgan
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
7
|
Caiola HO, Wu Q, Soni S, Wang XF, Monahan K, Pang ZP, Wagner GC, Zhang H. Neuronal connectivity, behavioral, and transcriptional alterations associated with the loss of MARK2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.569759. [PMID: 38105965 PMCID: PMC10723285 DOI: 10.1101/2023.12.05.569759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Neuronal connectivity is essential for adaptive brain responses and can be modulated by dendritic spine plasticity and the intrinsic excitability of individual neurons. Dysregulation of these processes can lead to aberrant neuronal activity, which has been associated with numerous neurological disorders including autism, epilepsy, and Alzheimer's disease. Nonetheless, the molecular mechanisms underlying aberrant neuronal connectivity remains unclear. We previously found that the serine/threonine kinase Microtubule Affinity Regulating Kinase 2 (MARK2), also known as Partitioning Defective 1b (Par1b), is important for the formation of dendritic spines in vitro. However, despite its genetic association with several neurological disorders, the in vivo impact of MARK2 on neuronal connectivity and cognitive functions remains unclear. Here, we demonstrate that loss of MARK2 in vivo results in changes to dendritic spine morphology, which in turn leads to a decrease in excitatory synaptic transmission. Additionally, loss of MARK2 produces substantial impairments in learning and memory, anxiety, and social behavior. Notably, MARK2 deficiency results in heightened seizure susceptibility. Consistent with this observation, RNAseq analysis reveals transcriptional changes in genes regulating synaptic transmission and ion homeostasis. These findings underscore the in vivo role of MARK2 in governing synaptic connectivity, cognitive functions, and seizure susceptibility.
Collapse
|
8
|
Niraula S, Yan SS, Subramanian J. Amyloid pathology impairs experience-dependent inhibitory synaptic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.539450. [PMID: 37205469 PMCID: PMC10187277 DOI: 10.1101/2023.05.04.539450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Alzheimer's disease patients and mouse models exhibit aberrant neuronal activity and altered excitatory-to-inhibitory synaptic ratio. Using multicolor two-photon microscopy, we test how amyloid pathology alters the structural dynamics of excitatory and inhibitory synapses and their adaptation to altered visual experience in vivo in the visual cortex. We show that the baseline dynamics of mature excitatory synapses and their adaptation to visual deprivation are not altered in amyloidosis. Likewise, the baseline dynamics of inhibitory synapses are not affected. In contrast, visual deprivation fails to induce inhibitory synapse loss in amyloidosis, a phenomenon observed in nonpathological conditions. Intriguingly, inhibitory synapse loss associated with visual deprivation in nonpathological mice is accompanied by the broadening of spontaneous but not visually evoked calcium transients. However, such broadening does not manifest in the context of amyloidosis. We also show that excitatory and inhibitory synapse loss is locally clustered under the nonpathological state. In contrast, a fraction of synapse loss is not locally clustered in amyloidosis, indicating an impairment in inhibitory synapse adaptation to changes in excitatory synaptic activity.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
- Department of Surgery, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
9
|
Wright AL, Konen LM, Mockett BG, Morris GP, Singh A, Burbano LE, Milham L, Hoang M, Zinn R, Chesworth R, Tan RP, Royle GA, Clark I, Petrou S, Abraham WC, Vissel B. The Q/R editing site of AMPA receptor GluA2 subunit acts as an epigenetic switch regulating dendritic spines, neurodegeneration and cognitive deficits in Alzheimer's disease. Mol Neurodegener 2023; 18:65. [PMID: 37759260 PMCID: PMC10537207 DOI: 10.1186/s13024-023-00632-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/03/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND RNA editing at the Q/R site of GluA2 occurs with ~99% efficiency in the healthy brain, so that the majority of AMPARs contain GluA2(R) instead of the exonically encoded GluA2(Q). Reduced Q/R site editing infcreases AMPA receptor calcium permeability and leads to dendritic spine loss, neurodegeneration, seizures and learning impairments. Furthermore, GluA2 Q/R site editing is impaired in Alzheimer's disease (AD), raising the possibility that unedited GluA2(Q)-containing AMPARs contribute to synapse loss and neurodegeneration in AD. If true, then inhibiting expression of unedited GluA2(Q), while maintaining expression of GluA2(R), may be a novel strategy of preventing synapse loss and neurodegeneration in AD. METHODS We engineered mice with the 'edited' arginine codon (CGG) in place of the unedited glutamine codon (CAG) at position 607 of the Gria2 gene. We crossbred this line with the J20 mouse model of AD and conducted anatomical, electrophysiological and behavioural assays to determine the impact of eliminating unedited GluA2(Q) expression on AD-related phenotypes. RESULTS Eliminating unedited GluA2(Q) expression in AD mice prevented dendritic spine loss and hippocampal CA1 neurodegeneration as well as improved working and reference memory in the radial arm maze. These phenotypes were improved independently of Aβ pathology and ongoing seizure susceptibility. Surprisingly, our data also revealed increased spine density in non-AD mice with exonically encoded GluA2(R) as compared to their wild-type littermates, suggesting an unexpected and previously unknown role for unedited GluA2(Q) in regulating dendritic spines. CONCLUSION The Q/R editing site of the AMPA receptor subunit GluA2 may act as an epigenetic switch that regulates dendritic spines, neurodegeneration and memory deficits in AD.
Collapse
Affiliation(s)
- Amanda L Wright
- St Vincent's Clinical School, St Vincent's Hospital Sydney, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, 2010, Australia
- School of Rural Medicine, Charles Sturt University, Orange, NSW, 2800, Australia
| | - Lyndsey M Konen
- Centre for Neuroscience and Regenerative Medicine, St Vincent's Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia
| | - Bruce G Mockett
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Box 56, Dunedin, 9054, New Zealand
| | - Gary P Morris
- Centre for Neuroscience and Regenerative Medicine, St Vincent's Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7005, Australia
| | - Anurag Singh
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Box 56, Dunedin, 9054, New Zealand
| | - Lisseth Estefania Burbano
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Luke Milham
- St Vincent's Clinical School, St Vincent's Hospital Sydney, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, 2010, Australia
- Centre for Neuroscience and Regenerative Medicine, St Vincent's Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia
| | - Monica Hoang
- School of Pharmacy, University of Waterloo, Kitchener, ON, N2G 1C5, Canada
| | - Raphael Zinn
- Centre for Neuroscience and Regenerative Medicine, St Vincent's Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia
| | - Rose Chesworth
- School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Richard P Tan
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, NSW, 2050, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Gordon A Royle
- Middlemore Hospital, Counties Manukau DHB, Otahuhu, Auckland, 1062, New Zealand
- Faculty of Medical and Health Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand
| | - Ian Clark
- Research School of Biology, Australian National University, Canberra, ACT, 0200, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Box 56, Dunedin, 9054, New Zealand
| | - Bryce Vissel
- St Vincent's Clinical School, St Vincent's Hospital Sydney, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, 2010, Australia.
- Centre for Neuroscience and Regenerative Medicine, St Vincent's Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
10
|
Oroszi T, Geerts E, Rajadhyaksha R, Nyakas C, van Heuvelen MJG, van der Zee EA. Whole-body vibration ameliorates glial pathological changes in the hippocampus of hAPP transgenic mice, but does not affect plaque load. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:5. [PMID: 36941713 PMCID: PMC10026461 DOI: 10.1186/s12993-023-00208-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the core cause of dementia in elderly populations. One of the main hallmarks of AD is extracellular amyloid beta (Aβ) accumulation (APP-pathology) associated with glial-mediated neuroinflammation. Whole-Body Vibration (WBV) is a passive form of exercise, but its effects on AD pathology are still unknown. METHODS Five months old male J20 mice (n = 26) and their wild type (WT) littermates (n = 24) were used to investigate the effect of WBV on amyloid pathology and the healthy brain. Both J20 and WT mice underwent WBV on a vibration platform or pseudo vibration treatment. The vibration intervention consisted of 2 WBV sessions of 10 min per day, five days per week for five consecutive weeks. After five weeks of WBV, the balance beam test was used to assess motor performance. Brain tissue was collected to quantify Aβ deposition and immunomarkers of astrocytes and microglia. RESULTS J20 mice have a limited number of plaques at this relatively young age. Amyloid plaque load was not affected by WBV. Microglia activation based on IBA1-immunostaining was significantly increased in the J20 animals compared to the WT littermates, whereas CD68 expression was not significantly altered. WBV treatment was effective to ameliorate microglia activation based on morphology in both J20 and WT animals in the Dentate Gyrus, but not so in the other subregions. Furthermore, GFAP expression based on coverage was reduced in J20 pseudo-treated mice compared to the WT littermates and it was significantly reserved in the J20 WBV vs. pseudo-treated animals. Further, only for the WT animals a tendency of improved motor performance was observed in the WBV group compared to the pseudo vibration group. CONCLUSION In accordance with the literature, we detected an early plaque load, reduced GFAP expression and increased microglia activity in J20 mice at the age of ~ 6 months. Our findings indicate that WBV has beneficial effects on the early progression of brain pathology. WBV restored, above all, the morphology of GFAP positive astrocytes to the WT level that could be considered the non-pathological and hence "healthy" level. Next experiments need to be performed to determine whether WBV is also affective in J20 mice of older age or other AD mouse models.
Collapse
Affiliation(s)
- Tamas Oroszi
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.
- Research Center for Molecular Exercise Science, Hungarian University of Sports Science, Budapest, Hungary.
| | - Eva Geerts
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Reuben Rajadhyaksha
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Csaba Nyakas
- Research Center for Molecular Exercise Science, Hungarian University of Sports Science, Budapest, Hungary
- Department of Morphology and Physiology, Health Science Faculty, Semmelweis Univesity, Budapest, Hungary
| | - Marieke J G van Heuvelen
- Department of Human Movement Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Eddy A van der Zee
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| |
Collapse
|
11
|
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
|
12
|
Lanooij SD, Eisel ULM, van der Zee EA, Kas MJH. Variation in Group Composition Alters an Early-Stage Social Phenotype in hAPP-Transgenic J20 Mice. J Alzheimers Dis 2023; 93:211-224. [PMID: 36970900 DOI: 10.3233/jad-221126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
BACKGROUND Altered social behavior is one of the symptoms of Alzheimer's disease (AD) that results in social withdrawal and loneliness and provides a major burden on patients and their relatives. Furthermore, loneliness is associated with an increased risk to develop AD and related dementias. OBJECTIVE We aimed to investigate if altered social behavior is an early indicator of amyloid-β (Aβ) pathology in J20 mice, and if co-housing with wild type (WT) mice can positively influence this social phenotype. METHODS The social phenotype of group-housed mice was assessed using an automated behavioral scoring system for longitudinal recordings. Female mice were housed in a same-genotype (4 J20 or WT mice per colony) or mixed-genotype (2 J20 mice + 2 WT mice) colony. At 10 weeks of age, their behavior was assessed for five consecutive days. RESULTS J20 mice showed increased locomotor activity and social sniffing, and reduced social contact compared to WT mice housed in same-genotype colonies. Mixed-genotype housing reduced the social sniffing duration of J20 mice, increased social contact frequency of J20 mice, and increased nest hide by WT mice. CONCLUSION Thus, altered social behavior can be used as an early indicator of Aβ-pathology in female J20 mice. Additionally, when co-housed with WT mice, their social sniffing phenotype is not expressed and their social contact phenotype is reduced. Our findings highlight the presence of a social phenotype in the early stages of AD and indicate a role for social environment variation in the expression of social behavior of WT and J20 mice.
Collapse
Affiliation(s)
- Suzanne D Lanooij
- Groningen Institute for Evolutionary Life Sciences (GELIFES), Neurobiology, University of Groningen, Groningen, The Netherlands
| | - Ulrich L M Eisel
- Groningen Institute for Evolutionary Life Sciences (GELIFES), Neurobiology, University of Groningen, Groningen, The Netherlands
| | - Eddy A van der Zee
- Groningen Institute for Evolutionary Life Sciences (GELIFES), Neurobiology, University of Groningen, Groningen, The Netherlands
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences (GELIFES), Neurobiology, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Flores J, Fillion ML, LeBlanc AC. Caspase-1 inhibition improves cognition without significantly altering amyloid and inflammation in aged Alzheimer disease mice. Cell Death Dis 2022; 13:864. [PMID: 36220815 PMCID: PMC9553979 DOI: 10.1038/s41419-022-05290-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022]
Abstract
Human genetic and animal model studies indicate that brain microglial inflammation is a primary driver of cognitive impairment in Alzheimer Disease (AD). Inflammasome-activated Caspase-1 (Casp1) is associated with both AD microglial inflammation and neuronal degeneration. In mice, Casp1 genetic ablation or VX-765 small molecule inhibition of Casp1 given at onset of cognitive deficits strongly supports the association between microglial inflammation and cognitive impairment. Here, VX-765 significantly improved episodic and spatial memory impairment eight months after the onset of cognitive impairment in aged AD mice with significant amyloid beta peptide (Aβ) accumulation and microglial inflammation. Unexpectedly, while cognitive improvement was associated with dendritic spine density and hippocampal synaptophysin level recovery, VX-765 only slightly decreased Aβ deposition and did not alter biochemically-measured Aβ levels. Furthermore, increased hippocampal Iba1+-microglia, GFAP+-astrocytes, IL-1β, and TNF-α levels were unaltered by VX-765. These results support the hypothesis that neuronal degeneration, not Aβ or microglial inflammation, drives cognitive impairment in AD.
Collapse
Affiliation(s)
- Joseph Flores
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research at Jewish General Hospital, Montréal, QC Canada
| | - Marie-Lyne Fillion
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research at Jewish General Hospital, Montréal, QC Canada
| | - Andréa C. LeBlanc
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research at Jewish General Hospital, Montréal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montréal, QC Canada
| |
Collapse
|
14
|
Li JM, Yang FH, Chao MW, Tseng CY. Swimming exercise prevents hippocampal dendritic spine changes and memory loss caused by aging: An application of a new semi-automated spine analysis software. Mol Cell Neurosci 2022; 121:103755. [PMID: 35850447 DOI: 10.1016/j.mcn.2022.103755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/06/2022] [Accepted: 07/10/2022] [Indexed: 11/17/2022] Open
Abstract
Dendritic spines are small, ratchet-like protrusions on neuronal dendrites that form synapses for receiving neuronal messages. Dendritic spine morphology is associated with synapse function. If neurons degrade or are damaged, the spine morphology of neurons changes. Given that most commercially available spine analysis software is expensive and complex, this study investigated a semi-automated spine analysis software, CTSpine, and used previously published data to verify the accuracy of the analysis results of this software. We also applied CTSpine to understand whether aging causes alterations in the hippocampal spine morphology and whether physical exercise can impede dendritic spine changes in 20 male Sprague Dawley rats. The spines of pyramidal cells in the hippocampal Cornu Ammonis 1 (CA1) region in the aging group were more enriched in filopodium type pattern than those in the control group, whereas the spines of the exercised aging group showed a similar pattern to that of the control. No significant changes were observed in neuronal dendritic spines in other hippocampal regions. However, long-term hippocampal memory was considerably decreased in the aging group, which was reversed to some extent in the exercised aging group. CTSpine, a self-developed semi-automatic spine analysis software, showed results similar to those noted in published data and can be effectively applied to the study of dendritic patterns, including neurodevelopment and disease.
Collapse
Affiliation(s)
- Jun-Ming Li
- Psychiatry Department, Taoyuan Armed Forces General Hospital, No. 168, Zhongxing Rd., Longtan Dist, Taoyuan City 32551, Taiwan.
| | - Fu-Hua Yang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli District, Taoyuan 320, Taiwan.
| | - Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli District, Taoyuan 320, Taiwan.
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli District, Taoyuan 320, Taiwan.
| |
Collapse
|
15
|
Eskandari E, Eaves CJ. Paradoxical roles of caspase-3 in regulating cell survival, proliferation, and tumorigenesis. J Cell Biol 2022; 221:213213. [PMID: 35551578 PMCID: PMC9106709 DOI: 10.1083/jcb.202201159] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 11/22/2022] Open
Abstract
Caspase-3 is a widely expressed member of a conserved family of proteins, generally recognized for their activated proteolytic roles in the execution of apoptosis in cells responding to specific extrinsic or intrinsic inducers of this mode of cell death. However, accumulating evidence indicates that caspase-3 also plays key roles in regulating the growth and homeostatic maintenance of both normal and malignant cells and tissues in multicellular organisms. Given that yeast possess an ancestral caspase-like gene suggests that the caspase-3 protein may have acquired different functions later during evolution to better meet the needs of more complex multicellular organisms, but without necessarily losing all of the functions of its ancestral yeast precursor. This review provides an update on what has been learned about these interesting dichotomous roles of caspase-3, their evolution, and their potential relevance to malignant as well as normal cell biology.
Collapse
Affiliation(s)
- Ebrahim Eskandari
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Connie J. Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada,Correspondence to Connie J. Eaves:
| |
Collapse
|
16
|
Zussy C, John R, Urgin T, Otaegui L, Vigor C, Acar N, Canet G, Vitalis M, Morin F, Planel E, Oger C, Durand T, Rajshree SL, Givalois L, Devarajan PV, Desrumaux C. Intranasal Administration of Nanovectorized Docosahexaenoic Acid (DHA) Improves Cognitive Function in Two Complementary Mouse Models of Alzheimer’s Disease. Antioxidants (Basel) 2022; 11:antiox11050838. [PMID: 35624701 PMCID: PMC9137520 DOI: 10.3390/antiox11050838] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are a class of fatty acids that are closely associated with the development and function of the brain. The most abundant PUFA is docosahexaenoic acid (DHA, 22:6 n-3). In humans, low plasmatic concentrations of DHA have been associated with impaired cognitive function, low hippocampal volumes, and increased amyloid deposition in the brain. Several studies have reported reduced brain DHA concentrations in Alzheimer’s disease (AD) patients’ brains. Although a number of epidemiological studies suggest that dietary DHA consumption may protect the elderly from developing cognitive impairment or dementia including AD, several review articles report an inconclusive association between omega-3 PUFAs intake and cognitive decline. The source of these inconsistencies might be because DHA is highly oxidizable and its accessibility to the brain is limited by the blood–brain barrier. Thus, there is a pressing need for new strategies to improve DHA brain supply. In the present study, we show for the first time that the intranasal administration of nanovectorized DHA reduces Tau phosphorylation and restores cognitive functions in two complementary murine models of AD. These results pave the way for the development of a new approach to target the brain with DHA for the prevention or treatment of this devastating disease.
Collapse
Affiliation(s)
- Charleine Zussy
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Rijo John
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Mumbai 400019, India; (R.J.); (S.L.R.); (P.V.D.)
| | - Théo Urgin
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Léa Otaegui
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Claire Vigor
- IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34095 Montpellier, France; (C.V.); (C.O.); (T.D.)
| | - Niyazi Acar
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Geoffrey Canet
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Mathieu Vitalis
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Françoise Morin
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, CR-CHUQ, Québec City, QC G1V 0A6, Canada; (F.M.); (E.P.)
| | - Emmanuel Planel
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, CR-CHUQ, Québec City, QC G1V 0A6, Canada; (F.M.); (E.P.)
| | - Camille Oger
- IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34095 Montpellier, France; (C.V.); (C.O.); (T.D.)
| | - Thierry Durand
- IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34095 Montpellier, France; (C.V.); (C.O.); (T.D.)
| | - Shinde L. Rajshree
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Mumbai 400019, India; (R.J.); (S.L.R.); (P.V.D.)
| | - Laurent Givalois
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, CR-CHUQ, Québec City, QC G1V 0A6, Canada; (F.M.); (E.P.)
| | - Padma V. Devarajan
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Mumbai 400019, India; (R.J.); (S.L.R.); (P.V.D.)
| | - Catherine Desrumaux
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
- LIPSTIC LabEx, 21000 Dijon, France
- Correspondence: ; Tel.: +33-467-14-36-89; Fax: +33-467-14-33-86
| |
Collapse
|
17
|
Therapeutic potential of Nlrp1 inflammasome, Caspase-1, or Caspase-6 against Alzheimer disease cognitive impairment. Cell Death Differ 2022; 29:657-669. [PMID: 34625662 PMCID: PMC8901623 DOI: 10.1038/s41418-021-00881-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/08/2022] Open
Abstract
The sequential activation of Nucleotide-binding oligomerization domain, Leucine rich Repeat and Pyrin domain containing protein 1 (Nlrp1) inflammasome, Caspase-1 (Casp1), and Caspase-6 (Casp6) is implicated in primary human neuron cultures and Alzheimer Disease (AD) neurodegeneration. To validate the Nlrp1-Casp1-Casp6 pathway in vivo, the APPSwedish/Indiana J20 AD transgenic mouse model was generated on either a Nlrp1, Casp1 or Casp6 null genetic background and mice were studied at 4-5 months of age. Episodic memory deficits assessed with novel object recognition were normalized by genetic ablation of Nlrp1, Casp1, or Casp6 in J20 mice. Spatial learning deficits, assessed with the Barnes Maze, were normalized in genetically ablated J20, whereas memory recall was normalized in J20/Casp1-/- and J20/Casp6-/-, and improved in J20/Nlrp1-/- mice. Hippocampal CA1 dendritic spine density of the mushroom subtype was reduced in J20, and normalized in genetically ablated J20 mice. Reduced J20 hippocampal dentate gyrus and CA3 synaptophysin levels were normalized in genetically ablated J20. Increased Iba1+-microglia in the hippocampus and cortex of J20 brains were normalized by Casp1 and Casp6 ablation and reduced by Nlrp1 ablation. Increased pro-inflammatory cytokines, TNF-α and CXCL1, in the J20 hippocampus were normalized by Nlrp1 or Casp1 genetic ablation. CXCL1 was also normalized by Casp6 genetic ablation. IFN-γ was increased and total amyloid β peptide was decreased in genetically ablated Nlrp1, Casp1 or Casp6 J20 hippocampi. We conclude that Nlrp1, Casp1, or Casp6 are implicated in AD-related cognitive impairment, inflammation, and amyloidogenesis. These results indicate that Nlrp1, Casp1, and Casp6 represent rational therapeutic targets against cognitive impairment and inflammation in AD.
Collapse
|
18
|
Cuestas Torres DM, Cardenas FP. Synaptic plasticity in Alzheimer's disease and healthy aging. Rev Neurosci 2021; 31:245-268. [PMID: 32250284 DOI: 10.1515/revneuro-2019-0058] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
The strength and efficiency of synaptic connections are affected by the environment or the experience of the individual. This property, called synaptic plasticity, is directly related to memory and learning processes and has been modeled at the cellular level. These types of cellular memory and learning models include specific stimulation protocols that generate a long-term strengthening of the synapses, called long-term potentiation, or a weakening of the said long-term synapses, called long-term depression. Although, for decades, researchers have believed that the main cause of the cognitive deficit that characterizes Alzheimer's disease (AD) and aging was the loss of neurons, the hypothesis of an imbalance in the cellular and molecular mechanisms of synaptic plasticity underlying this deficit is currently widely accepted. An understanding of the molecular and cellular changes underlying the process of synaptic plasticity during the development of AD and aging will direct future studies to specific targets, resulting in the development of much more efficient and specific therapeutic strategies. In this review, we classify, discuss, and describe the main findings related to changes in the neurophysiological mechanisms of synaptic plasticity in excitatory synapses underlying AD and aging. In addition, we suggest possible mechanisms in which aging can become a high-risk factor for the development of AD and how its development could be prevented or slowed.
Collapse
Affiliation(s)
- Diana Marcela Cuestas Torres
- Departamento de Psicología and Departamento de Biología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| | - Fernando P Cardenas
- Departamento de Psicología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| |
Collapse
|
19
|
Dávila-Bouziguet E, Casòliba-Melich A, Targa-Fabra G, Galera-López L, Ozaita A, Maldonado R, Ávila J, Delgado-García JM, Gruart A, Soriano E, Pascual M. Functional protection in J20/VLW mice: a model of non-demented with Alzheimer's disease neuropathology. Brain 2021; 145:729-743. [PMID: 34424282 DOI: 10.1093/brain/awab319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/19/2021] [Accepted: 07/28/2021] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease comprises amyloid-β and hyperphosphorylated Tau accumulation, imbalanced neuronal activity, aberrant oscillatory rhythms, and cognitive deficits. Non-Demented with Alzheimer's disease Neuropathology (NDAN) defines a novel clinical entity with amyloid-β and Tau pathologies but preserved cognition. The mechanisms underlying such neuroprotection remain undetermined and animal models of NDAN are currently unavailable. We demonstrate that J20/VLW mice (accumulating amyloid-β and hyperphosphorylated Tau) exhibit preserved hippocampal rhythmic activity and cognition, as opposed to J20 and VLW animals, which show significant alterations. Furthermore, we show that the overexpression of mutant human Tau in coexistence with amyloid-β accumulation renders a particular hyperphosphorylated Tau signature in hippocampal interneurons. The GABAergic septohippocampal pathway, responsible for hippocampal rhythmic activity, is preserved in J20/VLW mice, in contrast to single mutants. Our data highlight J20/VLW mice as a suitable animal model in which to explore the mechanisms driving cognitive preservation in NDAN. Moreover, they suggest that a differential Tau phosphorylation pattern in hippocampal interneurons prevents the loss of GABAergic septohippocampal innervation and alterations in local field potentials, thereby avoiding cognitive deficits.
Collapse
Affiliation(s)
- Eva Dávila-Bouziguet
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), Spain
| | - Arnau Casòliba-Melich
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), Spain
| | - Georgina Targa-Fabra
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), Spain
| | - Lorena Galera-López
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Andrés Ozaita
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Jesús Ávila
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), Spain.,Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Neurobiology Laboratory, Madrid, Spain
| | - José M Delgado-García
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Neurobiology Laboratory, Madrid, Spain.,Division of Neurosciences, Pablo de Olavide University, Seville, Spain
| | - Agnès Gruart
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Neurobiology Laboratory, Madrid, Spain.,Division of Neurosciences, Pablo de Olavide University, Seville, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), Spain
| | - Marta Pascual
- Department of Cell Biology, Physiology and Immunology, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED, ISCIII), Spain
| |
Collapse
|
20
|
Kim HR, Jang JH, Ham H, Choo SH, Park J, Kang SH, Hwangbo S, Jang H, Na DL, Seo SW, Baek JH, Kim HJ. A Case of Early-Onset Alzheimer's Disease Mimicking Schizophrenia in a Patient with Presenilin 1 Mutation (S170P). J Alzheimers Dis 2021; 83:1025-1031. [PMID: 34366354 DOI: 10.3233/jad-210650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Atypical psychological symptoms frequently occur in early-onset Alzheimer's disease (EOAD), which makes it difficult to differentiate it from other psychiatric disorders. We report the case of a 28-year-old woman with EOAD, carrying a presenilin-1 mutation (S170P), who was initially misdiagnosed with schizophrenia because of prominent psychiatric symptoms in the first 1-2 years of the disease. Amyloid-β positron emission tomography (PET) showed remarkably high tracer uptake in the striatum and thalamus. Tau PET showed widespread cortical uptake and relatively low uptake in the subcortical and medial temporal regions. Our case advocates for considering EOAD diagnosis for young patients with psychiatric and atypical cognitive symptoms.
Collapse
Affiliation(s)
- Hang-Rai Kim
- Department of Neurology, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Republic of Korea.,Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Ja Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Honggi Ham
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Department of Digital Health, SAIHST, Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea.,Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seung Ho Choo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeongho Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Hoon Kang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Song Hwangbo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Ji Hyun Baek
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.,Department of Digital Health, SAIHST, Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Xu P, Chang JC, Zhou X, Wang W, Bamkole M, Wong E, Bettayeb K, Jiang LL, Huang T, Luo W, Xu H, Nairn AC, Flajolet M, Ip NY, Li YM, Greengard P. GSAP regulates lipid homeostasis and mitochondrial function associated with Alzheimer's disease. J Exp Med 2021; 218:e20202446. [PMID: 34156424 PMCID: PMC8222926 DOI: 10.1084/jem.20202446] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/22/2021] [Accepted: 05/26/2021] [Indexed: 11/04/2022] Open
Abstract
Biochemical, pathogenic, and human genetic data confirm that GSAP (γ-secretase activating protein), a selective γ-secretase modulatory protein, plays important roles in Alzheimer's disease (AD) and Down's syndrome. However, the molecular mechanism(s) underlying GSAP-dependent pathogenesis remains largely elusive. Here, through unbiased proteomics and single-nuclei RNAseq, we identified that GSAP regulates multiple biological pathways, including protein phosphorylation, trafficking, lipid metabolism, and mitochondrial function. We demonstrated that GSAP physically interacts with the Fe65-APP complex to regulate APP trafficking/partitioning. GSAP is enriched in the mitochondria-associated membrane (MAM) and regulates lipid homeostasis through the amyloidogenic processing of APP. GSAP deletion generates a lipid environment unfavorable for AD pathogenesis, leading to improved mitochondrial function and the rescue of cognitive deficits in an AD mouse model. Finally, we identified a novel GSAP single-nucleotide polymorphism that regulates its brain transcript level and is associated with an increased AD risk. Together, our findings indicate that GSAP impairs mitochondrial function through its MAM localization and that lowering GSAP expression reduces pathological effects associated with AD.
Collapse
Affiliation(s)
- Peng Xu
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Jerry C. Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science and Technology Parks, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease, and Drug Development, Shenzhen–Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Wei Wang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Michael Bamkole
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karima Bettayeb
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Lu-Lin Jiang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Timothy Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Wenjie Luo
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Angus C. Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, CT
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| | - Nancy Y. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science and Technology Parks, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease, and Drug Development, Shenzhen–Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Program of Pharmacology and Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY
| |
Collapse
|
22
|
Pentkowski NS, Rogge-Obando KK, Donaldson TN, Bouquin SJ, Clark BJ. Anxiety and Alzheimer's disease: Behavioral analysis and neural basis in rodent models of Alzheimer's-related neuropathology. Neurosci Biobehav Rev 2021; 127:647-658. [PMID: 33979573 DOI: 10.1016/j.neubiorev.2021.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 04/28/2021] [Accepted: 05/05/2021] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) pathology is commonly associated with cognitive decline but is also composed of neuropsychiatric symptoms including psychological distress and alterations in mood, including anxiety and depression. Emotional dysfunction in AD is frequently modeled using tests of anxiety-like behavior in transgenic rodents. These tests often include the elevated plus-maze, light/dark test and open field test. In this review, we describe prototypical behavioral paradigms used to examine emotional dysfunction in transgenic models of AD, specifically anxiety-like behavior. Next, we summarize the results of studies examining anxiety-like behavior in transgenic rodents, noting that the behavioral outcomes using these paradigms have produced inconsistent results. We suggest that future research will benefit from using a battery of tests to examine emotional behavior in transgenic AD models. We conclude by discussing putative, overlapping neurobiological mechanisms underlying AD-related neuropathology, stress and anxiety-like behavior reported in AD models.
Collapse
Affiliation(s)
- Nathan S Pentkowski
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico.
| | | | - Tia N Donaldson
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico
| | - Samuel J Bouquin
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico
| | - Benjamin J Clark
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico.
| |
Collapse
|
23
|
Caspase Activation and Caspase-Mediated Cleavage of APP Is Associated with Amyloid β-Protein-Induced Synapse Loss in Alzheimer's Disease. Cell Rep 2021; 31:107839. [PMID: 32610140 DOI: 10.1016/j.celrep.2020.107839] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/29/2020] [Accepted: 06/08/2020] [Indexed: 01/09/2023] Open
Abstract
Amyloid β-protein (Aβ) toxicity is hypothesized to play a seminal role in Alzheimer's disease (AD) pathogenesis. However, it remains unclear how Aβ causes synaptic dysfunction and synapse loss. We hypothesize that one mechanism of Aβ-induced synaptic injury is related to the cleavage of amyloid β precursor protein (APP) at position D664 by caspases that release the putatively cytotoxic C31 peptide. In organotypic slice cultures derived from mice with a knock-in mutation in the APP gene (APP D664A) to inhibit caspase cleavage, Aβ-induced synaptic injury is markedly reduced in two models of Aβ toxicity. Loss of dendritic spines is also attenuated in mice treated with caspase inhibitors. Importantly, the time-dependent dendritic spine loss is correlated with localized activation of caspase-3 but is absent in APP D664A cultures. We propose that the APP cytosolic domain plays an essential role in Aβ-induced synaptic damage in the injury pathway mediated by localized caspase activation.
Collapse
|
24
|
mTOR Attenuation with Rapamycin Reverses Neurovascular Uncoupling and Memory Deficits in Mice Modeling Alzheimer's Disease. J Neurosci 2021; 41:4305-4320. [PMID: 33888602 DOI: 10.1523/jneurosci.2144-20.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Vascular dysfunction is a universal feature of aging and decreased cerebral blood flow has been identified as an early event in the pathogenesis of Alzheimer's disease (AD). Cerebrovascular dysfunction in AD includes deficits in neurovascular coupling (NVC), a mechanism that ensures rapid delivery of energy substrates to active neurons through the blood supply. The mechanisms underlying NVC impairment in AD, however, are not well understood. We have previously shown that mechanistic/mammalian target of rapamycin (mTOR) drives cerebrovascular dysfunction in models of AD by reducing the activity of endothelial nitric oxide synthase (eNOS), and that attenuation of mTOR activity with rapamycin is sufficient to restore eNOS-dependent cerebrovascular function. Here we show mTOR drives NVC impairments in an AD model through the inhibition of neuronal NOS (nNOS)- and non-NOS-dependent components of NVC, and that mTOR attenuation with rapamycin is sufficient to restore NVC and even enhance it above WT responses. Restoration of NVC and concomitant reduction of cortical amyloid-β levels effectively treated memory deficits in 12-month-old hAPP(J20) mice. These data indicate that mTOR is a critical driver of NVC dysfunction and underlies cognitive impairment in an AD model. Together with our previous findings, the present studies suggest that mTOR promotes cerebrovascular dysfunction in AD, which is associated with early disruption of nNOS activation, through its broad negative impact on nNOS as well as on non-NOS components of NVC. Our studies highlight the potential of mTOR attenuation as an efficacious treatment for AD and potentially other neurologic diseases of aging.SIGNIFICANCE STATEMENT Failure of the blood flow response to neuronal activation [neurovascular coupling (NVC)] in a model of AD precedes the onset of AD-like cognitive symptoms and is driven, to a large extent, by mammalian/mechanistic target of rapamycin (mTOR)-dependent inhibition of nitric oxide synthase activity. Our studies show that mTOR also drives AD-like failure of non-nitric oxide (NO)-mediated components of NVC. Thus, mTOR attenuation may serve to treat AD, where we find that neuronal NO synthase is profoundly reduced early in disease progression, and potentially other neurologic diseases of aging with cerebrovascular dysfunction as part of their etiology.
Collapse
|
25
|
Sokolova D, Childs T, Hong S. Insight into the role of phosphatidylserine in complement-mediated synapse loss in Alzheimer's disease. Fac Rev 2021; 10:19. [PMID: 33718936 PMCID: PMC7946395 DOI: 10.12703/r/10-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The innate immune system plays an integral role in the brain. Synaptic pruning, a fundamental process in developmental circuit refinement, is partially mediated by neuroimmune signalling at the synapse. In particular, microglia, the major tissue-resident macrophages of the brain, and the classical complement cascade, an innate immune pathway that aids in the clearance of unwanted material, have been implicated in mediating synapse elimination. Emerging data suggest that improper signalling of the innate immune pathway at the synapse leads to pathological synapse loss in age-related neurodegenerative diseases, including Alzheimer's disease. Now the key questions are whether synapses are targeted by complement and, if so, which synapses are vulnerable to elimination. Here, we review recent work implicating C1q, the initiator of the classical complement cascade, and surrounding glia as mediators of synapse loss. We examine how synapses could undergo apoptosis-like pathways in the Alzheimer brain, which may lead to the externalisation of phosphatidylserine on synapses. Finally, we discuss potential roles for microglia and astrocytes in this 'synaptic apoptosis'. Critical insight into neuroimmune regulatory pathways on synapses will be key to developing effective targets against pathological synapse loss in dementia.
Collapse
Affiliation(s)
- Dimitra Sokolova
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London WC1E 6BT, UK
| | - Thomas Childs
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London WC1E 6BT, UK
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
26
|
Shields LY, Li H, Nguyen K, Kim H, Doric Z, Garcia JH, Gill TM, Haddad D, Vossel K, Calvert M, Nakamura K. Mitochondrial fission is a critical modulator of mutant APP-induced neural toxicity. J Biol Chem 2021; 296:100469. [PMID: 33639169 PMCID: PMC8042169 DOI: 10.1016/j.jbc.2021.100469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 11/30/2022] Open
Abstract
Alterations in mitochondrial fission may contribute to the pathophysiology of several neurodegenerative diseases, including Alzheimer's disease (AD). However, we understand very little about the normal functions of fission or how fission disruption may interact with AD-associated proteins to modulate pathogenesis. Here we show that loss of the central mitochondrial fission protein dynamin-related protein 1 (Drp1) in CA1 and other forebrain neurons markedly worsens the learning and memory of mice expressing mutant human amyloid precursor protein (hAPP) in neurons. In cultured neurons, Drp1KO and hAPP converge to produce mitochondrial Ca2+ (mitoCa2+) overload, despite decreasing mitochondria-associated ER membranes (MAMs) and cytosolic Ca2+. This mitoCa2+ overload occurs independently of ATP levels. These findings reveal a potential mechanism by which mitochondrial fission protects against hAPP-driven pathology.
Collapse
Affiliation(s)
- Lauren Y Shields
- Gladstone Institute of Neurological Disease, San Francisco, California, USA; Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Huihui Li
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Kevin Nguyen
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Hwajin Kim
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Zak Doric
- Gladstone Institute of Neurological Disease, San Francisco, California, USA; Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Joseph H Garcia
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - T Michael Gill
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Dominik Haddad
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Keith Vossel
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Meredith Calvert
- Gladstone Institute of Neurological Disease, San Francisco, California, USA
| | - Ken Nakamura
- Gladstone Institute of Neurological Disease, San Francisco, California, USA; Department of Neurology, University of California, San Francisco, San Francisco, California, USA; Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
27
|
AhR/miR-23a-3p/PKCα axis contributes to memory deficits in ovariectomized and normal aging female mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:79-91. [PMID: 33738140 PMCID: PMC7940705 DOI: 10.1016/j.omtn.2021.02.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/14/2021] [Indexed: 12/14/2022]
Abstract
The mechanism of estrogen deficiency-induced cognitive impairment is still not fully elucidated. In this study, we assessed the effect of microRNA (miRNA) on the memory of long-term estrogen-deficient mice after ovariectomy (OVX) and normal aging. We observed that 5-month OVX and 22-month-old normal aging female mice showed significantly impaired spatial and object recognition memory, declined hippocampal long-term potentiation (LTP), and decreased hippocampal protein kinase C α (PKCα) protein. Quantitative real-time PCR analysis showed upregulated miRNA-23a-3p (miR-23a-3p) in the hippocampus of 5-month OVX and 22-month-old female mice. In vitro, overexpression of miR-23a-3p downregulated PKCα by binding the 3¢ UTRs of Prkca mRNAs, which was prevented by its antisense oligonucleotide AMO-23a. In vivo, adeno-associated virus-mediated overexpression of miR-23a-3p (AAV-pre-miR-23a-3p) suppressed hippocampal PKCα and impaired the memory of mice. Chromatin immunoprecipitation analysis showed that aryl hydrocarbon receptor (AhR) binds the promoter region of miR-23a-3p. The AhR-dependent downregulation of PKCα could be prevented by AMO-23a as well. Furthermore, knockdown of miR-23a-3p using AAV-AMO-23a rescued the cognitive and electrophysiological impairments of OVX and normal aging female mice. We conclude that long-term estrogen deficiency impairs cognition and hippocampal LTP by activating the AhR/miR-23a-3p/PKCα axis. The knockdown of miR-23a-3p may be a potentially valuable therapeutic strategy for estrogen deficiency-induced memory deficits.
Collapse
|
28
|
Machine learning classifies predictive kinematic features in a mouse model of neurodegeneration. Sci Rep 2021; 11:3950. [PMID: 33597593 PMCID: PMC7889656 DOI: 10.1038/s41598-021-82694-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 01/21/2021] [Indexed: 01/31/2023] Open
Abstract
Motor deficits are observed in Alzheimer's disease (AD) prior to the appearance of cognitive symptoms. To investigate the role of amyloid proteins in gait disturbances, we characterized locomotion in APP-overexpressing transgenic J20 mice. We used three-dimensional motion capture to characterize quadrupedal locomotion on a treadmill in J20 and wild-type mice. Sixteen J20 mice and fifteen wild-type mice were studied at two ages (4- and 13-month). A random forest (RF) classification algorithm discriminated between the genotypes within each age group using a leave-one-out cross-validation. The balanced accuracy of the RF classification was 92.3 ± 5.2% and 93.3 ± 4.5% as well as False Negative Rate (FNR) of 0.0 ± 0.0% and 0.0 ± 0.0% for the 4-month and 13-month groups, respectively. Feature ranking algorithms identified kinematic features that when considered simultaneously, achieved high genotype classification accuracy. The identified features demonstrated an age-specific kinematic profile of the impact of APP-overexpression. Trunk tilt and unstable hip movement patterns were important in classifying the 4-month J20 mice, whereas patterns of shoulder and iliac crest movement were critical for classifying 13-month J20 mice. Examining multiple kinematic features of gait simultaneously could also be developed to classify motor disorders in humans.
Collapse
|
29
|
Poon CH, Wang Y, Fung ML, Zhang C, Lim LW. Rodent Models of Amyloid-Beta Feature of Alzheimer's Disease: Development and Potential Treatment Implications. Aging Dis 2020; 11:1235-1259. [PMID: 33014535 PMCID: PMC7505263 DOI: 10.14336/ad.2019.1026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide and causes severe financial and social burdens. Despite much research on the pathogenesis of AD, the neuropathological mechanisms remain obscure and current treatments have proven ineffective. In the past decades, transgenic rodent models have been used to try to unravel this disease, which is crucial for early diagnosis and the assessment of disease-modifying compounds. In this review, we focus on transgenic rodent models used to study amyloid-beta pathology in AD. We also discuss their possible use as promising tools for AD research. There is still no effective treatment for AD and the development of potent therapeutics are urgently needed. Many molecular pathways are susceptible to AD, ranging from neuroinflammation, immune response, and neuroplasticity to neurotrophic factors. Studying these pathways may shed light on AD pathophysiology as well as provide potential targets for the development of more effective treatments. This review discusses the advantages and limitations of these models and their potential therapeutic implications for AD.
Collapse
Affiliation(s)
- Chi Him Poon
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingyi Wang
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Lung Fung
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chengfei Zhang
- 2Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
30
|
Flores J, Noël A, Foveau B, Beauchet O, LeBlanc AC. Pre-symptomatic Caspase-1 inhibitor delays cognitive decline in a mouse model of Alzheimer disease and aging. Nat Commun 2020; 11:4571. [PMID: 32917871 PMCID: PMC7486940 DOI: 10.1038/s41467-020-18405-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Early therapeutic interventions are essential to prevent Alzheimer Disease (AD). The association of several inflammation-related genetic markers with AD and the early activation of pro-inflammatory pathways in AD suggest inflammation as a plausible therapeutic target. Inflammatory Caspase-1 has a significant impact on AD-like pathophysiology and Caspase-1 inhibitor, VX-765, reverses cognitive deficits in AD mouse models. Here, a one-month pre-symptomatic treatment of Swedish/Indiana mutant amyloid precursor protein (APPSw/Ind) J20 and wild-type mice with VX-765 delays both APPSw/Ind- and age-induced episodic and spatial memory deficits. VX-765 delays inflammation without considerably affecting soluble and aggregated amyloid beta peptide (Aβ) levels. Episodic memory scores correlate negatively with microglial activation. These results suggest that Caspase-1-mediated inflammation occurs early in the disease and raise hope that VX-765, a previously Food and Drug Administration-approved drug for human CNS clinical trials, may be a useful drug to prevent the onset of cognitive deficits and brain inflammation in AD.
Collapse
Affiliation(s)
- Joseph Flores
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Anastasia Noël
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Bénédicte Foveau
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Olivier Beauchet
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Medicine, Division of Geriatric Medicine, Sir Mortimer B. Davis - Jewish General Hospital, Montreal, Quebec, Canada.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Andréa C LeBlanc
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
31
|
Rodriguez GA, Barrett GM, Duff KE, Hussaini SA. Chemogenetic attenuation of neuronal activity in the entorhinal cortex reduces Aβ and tau pathology in the hippocampus. PLoS Biol 2020; 18:e3000851. [PMID: 32822389 PMCID: PMC7467290 DOI: 10.1371/journal.pbio.3000851] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/02/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
High levels of the amyloid-beta (Aβ) peptide have been shown to disrupt neuronal function and induce hyperexcitability, but it is unclear what effects Aβ-associated hyperexcitability may have on tauopathy pathogenesis or propagation in vivo. Using a novel transgenic mouse line to model the impact of human APP (hAPP)/Aβ accumulation on tauopathy in the entorhinal cortex–hippocampal (EC-HIPP) network, we demonstrate that hAPP overexpression aggravates EC-Tau aggregation and accelerates pathological tau spread into the hippocampus. In vivo recordings revealed a strong role for hAPP/Aβ, but not tau, in the emergence of EC neuronal hyperactivity and impaired theta rhythmicity. Chronic chemogenetic attenuation of EC neuronal hyperactivity led to reduced hAPP/Aβ accumulation and reduced pathological tau spread into downstream hippocampus. These data strongly support the hypothesis that in Alzheimer’s disease (AD), Aβ-associated hyperactivity accelerates the progression of pathological tau along vulnerable neuronal circuits, and demonstrates the utility of chronic, neuromodulatory approaches in ameliorating AD pathology in vivo. A novel, triple transgenic mouse model of Alzheimer's disease reveals that amyloid beta-associated neuronal hyperactivity and network dysfunction accelerates the spread of pathological tau from the entorhinal cortex into the hippocampus. Chronic attenuation of neuronal activity using chemogenetics reduces this effect, supporting a role for neuronal hyperactivity in Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Gustavo A. Rodriguez
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Geoffrey M. Barrett
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Karen E. Duff
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
- UK Dementia Research Institute at University College London, London, United Kingdom
- * E-mail: (SAH); (KED)
| | - S. Abid Hussaini
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
- * E-mail: (SAH); (KED)
| |
Collapse
|
32
|
Oberman K, Gouweleeuw L, Hoogerhout P, Eisel ULM, van Riet E, Schoemaker RG. Vaccination Prevented Short-Term Memory Loss, but Deteriorated Long-Term Spatial Memory in Alzheimer's Disease Mice, Independent of Amyloid-β Pathology. J Alzheimers Dis Rep 2020; 4:261-280. [PMID: 32904788 PMCID: PMC7458552 DOI: 10.3233/adr-200213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Soluble oligomeric amyloid-β (Aβ), rather than Aβ plaques, seems to be the culprit in Alzheimer’s disease (AD). Accordingly, a new concept vaccine of small cyclic peptide conjugates, selectively targeting oligomeric Aβ, has been developed. Objective: Study the therapeutic potential of this new vaccine in a mouse model for AD. Methods: J20 mice, overexpressing human amyloid precursor protein, were validated for an AD-like phenotype. Then, J20 mice were vaccinated at 2, 3, and 4 months of age and AD phenotype was evaluated at 6, 9, and 12 months of age; or at 9, 10, and 11 months with evaluation at 12 months. Effects on Aβ pathology were studied by plaque load (immunohistochemistry; 6E10) and antibody titers against Aβ (ELISA). AD behavioral phenotype was evaluated by performance in a battery of cognitive tests. Results: J20 mice displayed age-related Aβ plaque development and an AD-like behavioral phenotype. A consistent antibody response to the cyclic peptides was, however, not extended to Aβ, leaving plaque load unaffected. Nevertheless, immunization at young ages prevented working- and short-term spatial memory loss, but deteriorated long-term spatial learning and memory, at 12 months of age. Immunization at later ages did not affect any measured parameter. Conclusion: J20 mice provide a relevant model for AD to study potential anti-Aβ treatment. Early vaccination prevented short-term memory loss at later ages, but deteriorated long-term spatial memory, however without affecting Aβ pathology. Later vaccination had no effects, but optimal timing may require further investigation.
Collapse
Affiliation(s)
- Klaske Oberman
- Department of Neurobiology GELIFES, University Groningen, The Netherlands
| | - Leonie Gouweleeuw
- Department of Neurobiology GELIFES, University Groningen, The Netherlands
| | | | - Ulrich L M Eisel
- Department of Neurobiology GELIFES, University Groningen, The Netherlands
| | | | - Regien G Schoemaker
- Department of Neurobiology GELIFES, University Groningen, The Netherlands.,University Medical Center Groningen, The Netherlands
| |
Collapse
|
33
|
Ferreira S, Pitman KA, Wang S, Summers BS, Bye N, Young KM, Cullen CL. Amyloidosis is associated with thicker myelin and increased oligodendrogenesis in the adult mouse brain. J Neurosci Res 2020; 98:1905-1932. [PMID: 32557778 PMCID: PMC7540704 DOI: 10.1002/jnr.24672] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/03/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022]
Abstract
In Alzheimer's disease, amyloid plaque formation is associated with the focal death of oligodendrocytes and soluble amyloid β impairs the survival of oligodendrocytes in vitro. However, the response of oligodendrocyte progenitor cells (OPCs) to early amyloid pathology remains unclear. To explore this, we performed a histological, electrophysiological, and behavioral characterization of transgenic mice expressing a pathological form of human amyloid precursor protein (APP), containing three single point mutations associated with the development of familial Alzheimer's disease (PDGFB‐APPSw.Ind, also known as J20 mice). PDGFB‐APPSw.Ind transgenic mice had impaired survival from weaning, were hyperactive by 2 months of age, and developed amyloid plaques by 6 months of age, however, their spatial memory remained intact over this time course. Hippocampal OPC density was normal in P60‐P180 PDGFB‐APPSw.Ind transgenic mice and, by performing whole‐cell patch‐clamp electrophysiology, we found that their membrane properties, including their response to kainate (100 µM), were largely normal. However, by P100, the response of hippocampal OPCs to GABA was elevated in PDGFB‐APPSw.Ind transgenic mice. We also found that the nodes of Ranvier were shorter, the paranodes longer, and the myelin thicker for hippocampal axons in young adult PDGFB‐APPSw.Ind transgenic mice compared with wildtype littermates. Additionally, oligodendrogenesis was normal in young adulthood, but increased in the hippocampus, entorhinal cortex, and fimbria of PDGFB‐APPSw.Ind transgenic mice as pathology developed. As the new oligodendrocytes were not associated with a change in total oligodendrocyte number, these cells are likely required for cell replacement.
Collapse
Affiliation(s)
- Solène Ferreira
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Shiwei Wang
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Benjamin S Summers
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Bye
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
34
|
Castanho I, Murray TK, Hannon E, Jeffries A, Walker E, Laing E, Baulf H, Harvey J, Bradshaw L, Randall A, Moore K, O'Neill P, Lunnon K, Collier DA, Ahmed Z, O'Neill MJ, Mill J. Transcriptional Signatures of Tau and Amyloid Neuropathology. Cell Rep 2020; 30:2040-2054.e5. [PMID: 32049030 PMCID: PMC7016505 DOI: 10.1016/j.celrep.2020.01.063] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 11/21/2019] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is associated with the intracellular aggregation of hyperphosphorylated tau and the accumulation of β-amyloid in the neocortex. We use transgenic mice harboring human tau (rTg4510) and amyloid precursor protein (J20) mutations to investigate transcriptional changes associated with the progression of tau and amyloid pathology. rTg4510 mice are characterized by widespread transcriptional differences in the entorhinal cortex with changes paralleling neuropathological burden across multiple brain regions. Differentially expressed transcripts overlap with genes identified in genetic studies of familial and sporadic AD. Systems-level analyses identify discrete co-expression networks associated with the progressive accumulation of tau that are enriched for genes and pathways previously implicated in AD pathology and overlap with co-expression networks identified in human AD cortex. Our data provide further evidence for an immune-response component in the accumulation of tau and reveal molecular pathways associated with the progression of AD neuropathology.
Collapse
Affiliation(s)
- Isabel Castanho
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Tracey K Murray
- Eli Lilly & Co., Erl Wood Manor, Sunninghill Road, Windlesham GU20 6PH, UK
| | - Eilis Hannon
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Aaron Jeffries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Emma Walker
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Emma Laing
- Eli Lilly & Co., Erl Wood Manor, Sunninghill Road, Windlesham GU20 6PH, UK
| | - Hedley Baulf
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Joshua Harvey
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Lauren Bradshaw
- Eli Lilly & Co., Erl Wood Manor, Sunninghill Road, Windlesham GU20 6PH, UK
| | - Andrew Randall
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Karen Moore
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Paul O'Neill
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Katie Lunnon
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - David A Collier
- Eli Lilly & Co., Erl Wood Manor, Sunninghill Road, Windlesham GU20 6PH, UK
| | - Zeshan Ahmed
- Eli Lilly & Co., Erl Wood Manor, Sunninghill Road, Windlesham GU20 6PH, UK
| | - Michael J O'Neill
- Eli Lilly & Co., Erl Wood Manor, Sunninghill Road, Windlesham GU20 6PH, UK
| | - Jonathan Mill
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK.
| |
Collapse
|
35
|
Abstract
Endogenous retroviruses (ERVs) consist of interspersed genomic elements derived from retroviral infections that invaded our ancestral germ lines. Notably, ERVs compose 8 to 10% of the human and mouse genomes. Until recently, ERVs were considered unimportant, so-called “junk” DNA. However, this naïve characterization has changed dramatically as distinct ERV-related functions are revealed in heath and disease. In this study, we demonstrate that chronic ERV activation is associated with cognitive impairment, measured with hippocampus-related tasks, in a mouse model. We confirm these findings in an independent mouse model of acute retroviral activation and show that cognitive deficits are mitigated in the absence of the retroviral RNA sensor protein MAVS. Our results point to an underappreciated therapeutic modality for impaired cognition. Retrotransposons compose a staggering 40% of the mammalian genome. Among them, endogenous retroviruses (ERV) represent sequences that closely resemble the proviruses created from exogenous retroviral infection. ERVs make up 8 to 10% of human and mouse genomes and range from evolutionarily ancient sequences to recent acquisitions. Studies in Drosophila have provided a causal link between genomic retroviral elements and cognitive decline; however, in mammals, the role of ERVs in learning and memory remains unclear. Here we studied 2 independent murine models for ERV activation: muMT strain (lacking B cells and antibody production) and intracerebroventricular injection of streptozotocin (ICVI-STZ). We conducted behavioral assessments (contextual fear memory and spatial learning), as well as gene and protein analysis (RNA sequencing, PCR, immunohistochemistry, and western blot assays). Mice lacking mitochondrial antiviral-signaling protein (MAVS) and mice lacking stimulator of IFN genes protein (STING), 2 downstream sensors of ERV activation, provided confirmation of ERV impact. We found that muMT mice and ICVI-STZ mice induced hippocampal ERV activation, as shown by increased gene and protein expression of the Gag sequence of the transposable element intracisternal A-particle. ERV activation was accompanied by significant hippocampus-related memory impairment in both models. Notably, the deficiency of the MAVS pathway was protective against ICVI-STZ–induced cognitive pathology. Overall, our results demonstrate that ERV activation is associated with cognitive impairment in mice. Moreover, they provide a molecular target for strategies aimed at attenuating retroviral element sensing, via MAVS, to treat dementia and neuropsychiatric disorders.
Collapse
|
36
|
Espinosa-Oliva AM, García-Revilla J, Alonso-Bellido IM, Burguillos MA. Brainiac Caspases: Beyond the Wall of Apoptosis. Front Cell Neurosci 2019; 13:500. [PMID: 31749689 PMCID: PMC6848387 DOI: 10.3389/fncel.2019.00500] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/22/2019] [Indexed: 01/04/2023] Open
Abstract
For the last two decades, caspases, a family of cysteine-aspartic proteases, have evolved from being considered solely as regulators of apoptosis or inflammation to having a wider range of functions. In this mini review, we focus on the most recent “non-apoptotic” roles of caspases in the CNS, particularly in neurons, astrocytes and oligodendrocytes. Non-apoptotic caspase functions in microglia have already been reviewed extensively elsewhere. Here we discuss the involvement of caspases in the activation of the inflammasome, autophagy, and non-apoptotic forms of cell death such as necroptosis and pyroptosis. Also, we review the involvement of caspases in synapses and the processing of aggregates key to neurodegenerative diseases such as Parkinson’s, Alzheimer’s and Huntington’s diseases. Likewise, we mention the recently described involvement of caspases in mitochondrial biogenesis, which is a function independent of the enzymatic activity. We conclude discussing the relevance that “new” functions of caspases have in the CNS and the future of this field of research.
Collapse
Affiliation(s)
- Ana María Espinosa-Oliva
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Juan García-Revilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Isabel María Alonso-Bellido
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| | - Miguel Angel Burguillos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC, Seville, Spain
| |
Collapse
|
37
|
Butterfield DA, Boyd-Kimball D. Redox proteomics and amyloid β-peptide: insights into Alzheimer disease. J Neurochem 2019; 151:459-487. [PMID: 30216447 PMCID: PMC6417976 DOI: 10.1111/jnc.14589] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/15/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disorder associated with aging and characterized pathologically by the presence of senile plaques, neurofibrillary tangles, and neurite and synapse loss. Amyloid beta-peptide (1-42) [Aβ(1-42)], a major component of senile plaques, is neurotoxic and induces oxidative stress in vitro and in vivo. Redox proteomics has been used to identify proteins oxidatively modified by Aβ(1-42) in vitro and in vivo. In this review, we discuss these proteins in the context of those identified to be oxidatively modified in animal models of AD, and human studies including familial AD, pre-clinical AD (PCAD), mild cognitive impairment (MCI), early AD, late AD, Down syndrome (DS), and DS with AD (DS/AD). These redox proteomics studies indicate that Aβ(1-42)-mediated oxidative stress occurs early in AD pathogenesis and results in altered antioxidant and cellular detoxification defenses, decreased energy yielding metabolism and mitochondrial dysfunction, excitotoxicity, loss of synaptic plasticity and cell structure, neuroinflammation, impaired protein folding and degradation, and altered signal transduction. Improved access to biomarker imaging and the identification of lifestyle interventions or treatments to reduce Aβ production could be beneficial in preventing or delaying the progression of AD. This article is part of the special issue "Proteomics".
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH 44601
| |
Collapse
|
38
|
Mouse induced pluripotent stem cells-derived Alzheimer's disease cerebral organoid culture and neural differentiation disorders. Neurosci Lett 2019; 711:134433. [PMID: 31421155 DOI: 10.1016/j.neulet.2019.134433] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, characterized by cognitive impairment. However, the pathogenesis of AD are very complicated, and the theories of Aβ and neurofibrillary tangles cannot explain all pathological alterations and clinical symptoms. Here, we used three-dimensional (3D) neural organoids culture derived from mouse induced pluripotent stem cells (iPSCs) to investigate the pathological mechanisms of AD. In this study, AD cerebral organoids were generated by overexpressing familial AD mutations (APP and PS1 genes) in mouse induced pluripotent stem cells, so that the early pathogenesis of AD could be investigated well with protein and cellular phenotype analyses. The results showed that AD cerebral organoids appeared some AD pathological alterations, and high levels of Aβ and p-Tau were induced as well. Furthermore, the number of GFAP-positive astrocytes and glutamatergic excitatory neurons increased significantly, but the number of GABAergic interneurons decreased. In conclusion, we suggest that cerebral organoids are a suitable AD model for scientific study, and that will provide us a novel insight into the understanding of the pathogenesis of AD.
Collapse
|
39
|
Wagner JM, Sichler ME, Schleicher EM, Franke TN, Irwin C, Löw MJ, Beindorff N, Bouter C, Bayer TA, Bouter Y. Analysis of Motor Function in the Tg4-42 Mouse Model of Alzheimer's Disease. Front Behav Neurosci 2019; 13:107. [PMID: 31156407 PMCID: PMC6533559 DOI: 10.3389/fnbeh.2019.00107] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common form of dementia. Hallmarks of AD are memory impairments and cognitive deficits, but non-cognitive impairments, especially motor dysfunctions are also associated with the disease and may even precede classic clinical symptoms. With an aging society and increasing hospitalization of the elderly, motor deficits are of major interest to improve independent activities in daily living. Consistent with clinical findings, a variety of AD mouse models develop motor deficits as well. We investigated the motor function of 3- and 7-month-old Tg4-42 mice in comparison to wild-type controls and 5XFAD mice and discuss the results in context with several other AD mouse model. Our study shows impaired balance and motor coordination in aged Tg4-42 mice in the balance beam and rotarod test, while general locomotor activity and muscle strength is not impaired at 7 months. The cerebellum is a major player in the regulation and coordination of balance and locomotion through practice. Particularly, the rotarod test is able to detect cerebellar deficits. Furthermore, supposed cerebellar impairment was verified by 18F-FDG PET/MRI. Aged Tg4-42 mice showed reduced cerebellar glucose metabolism in the 18F-FDG PET. Suggesting that, deficits in coordination and balance are most likely due to cerebellar impairment. In conclusion, Tg4-42 mice develop motor deficits before memory deficits, without confounding memory test. Thus, making the Tg4-42 mouse model a good model to study the effects on cognitive decline of therapies targeting motor impairments.
Collapse
Affiliation(s)
- Jannek M. Wagner
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Marius E. Sichler
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Eva M. Schleicher
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Timon N. Franke
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Maximilian Johannes Löw
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center, Charité – University Medicine Berlin, Berlin, Germany
| | - Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Thomas A. Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| |
Collapse
|
40
|
Ameen-Ali KE, Simpson JE, Wharton SB, Heath PR, Sharp PS, Brezzo G, Berwick J. The Time Course of Recognition Memory Impairment and Glial Pathology in the hAPP-J20 Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2019; 68:609-624. [DOI: 10.3233/jad-181238] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Kamar E. Ameen-Ali
- Department of Psychology, University of Sheffield, Sheffield, UK
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle-Upon-Tyne, UK
| | - Julie E. Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Stephen B. Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R. Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul S. Sharp
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Gaia Brezzo
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Jason Berwick
- Department of Psychology, University of Sheffield, Sheffield, UK
| |
Collapse
|
41
|
Mondragón-Rodríguez S, Gu N, Fasano C, Peña-Ortega F, Williams S. Functional Connectivity between Hippocampus and Lateral Septum is Affected in Very Young Alzheimer’s Transgenic Mouse Model. Neuroscience 2019; 401:96-105. [DOI: 10.1016/j.neuroscience.2018.12.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/20/2018] [Accepted: 12/22/2018] [Indexed: 12/29/2022]
|
42
|
Quartey MO, Nyarko JNK, Pennington PR, Heistad RM, Chaharyn BM, Wei Z, Bainbridge D, Baker GB, Mousseau DD. Age- and sex-dependent profiles of APP fragments and key secretases align with changes in despair-like behavior and cognition in young APPSwe/Ind mice. Biochem Biophys Res Commun 2019; 511:454-459. [PMID: 30803762 DOI: 10.1016/j.bbrc.2019.02.083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/15/2019] [Indexed: 12/11/2022]
Abstract
Biological sex exerts distinct influences on brain levels of the β-amyloid (Aβ) peptide in both clinical depression and Alzheimer disease (AD), yet studies in animal models focus primarily on males. We examined behavioral 'despair'/depression (using the tail-suspension test) and memory (using the novel object recognition task) in J20 (hAPPSwe/Ind) mice. Three month-old male (but not female) J20 mice exhibited less despair-like behavior, but more evidence of cognitive deficits. In young J20 mice, only soluble Aβ peptides -primarily Aβ(1-40)- were detected. There was no evidence of an effect on despair-like behavior in the six month-old J20 mice, although cognitive deficits were now evident in both sexes, and coincided with a greater proportion of the neurotoxic Aβ(1-42) species (in soluble as well as insoluble fractions). This age-dependent shift in Aβ peptide profile coincided with reduced expression of glycosylated species of ADAM-10 (α-secretase) and BACE1 (β-secretase), and an increased co-immunoprecipitation of presenilin-1 with nicastrin (components of the γ-secretase complex). Sex-dependent changes in depression-related monoaminergic, e.g. serotonin and dopamine (but not noradrenaline), systems were evident already in young J20 mice. It is critical to acknowledge that sex-dependent APP-related phenotypes might differentially influence modifiable depression-related monoaminergic signalling at some of the earliest pathological stages of clinical AD.
Collapse
Affiliation(s)
- Maa O Quartey
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada
| | - Jennifer N K Nyarko
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada
| | - Paul R Pennington
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada
| | - Ryan M Heistad
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada
| | - Bradley M Chaharyn
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada
| | - Zelan Wei
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada
| | - Dennis Bainbridge
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada
| | - Glen B Baker
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Canada.
| |
Collapse
|
43
|
Nyarko JNK, Quartey MO, Baker GB, Mousseau DD. Can Animal Models Inform on the Relationship between Depression and Alzheimer Disease? CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2019; 64:18-29. [PMID: 29685068 PMCID: PMC6364140 DOI: 10.1177/0706743718772514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The focus on the β-amyloid (Aβ) peptide in clinical Alzheimer disease (AD) as well as in animal models of AD has perhaps biased our understanding of what contributes to the heterogeneity in disease onset and progression. Part of this heterogeneity could reflect the various neuropsychiatric risk factors that present with common symptomatology and can predispose the brain to AD-like changes. One such risk factor is depression. Animal models, particularly mouse models carrying variants of AD-related gene(s), many of which lead to an accumulation of Aβ, suggest that a fundamental shift in depression-related monoaminergic systems (including serotonin and noradrenaline) is a strong indicator of the altered cellular function associated with the earlier(est) stages of AD-related pathology. These changes in monoaminergic neurochemistry could provide for relevant targets for intervention in clinical AD and/or could support a polypharmacy strategy, which might include the targeting of Aβ, in vulnerable populations. Future studies must also include female mice as well as male mice in animal model studies on the relationship between depression and AD.
Collapse
Affiliation(s)
- Jennifer N K Nyarko
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Maa O Quartey
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Glen B Baker
- 2 Department of Psychiatry, Neuroscience and Mental Health Institute, Neurochemical Research Unit, University of Alberta, Edmonton, Alberta, Canada
| | - Darrell D Mousseau
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
44
|
Ziegler-Waldkirch S, Meyer-Luehmann M. The Role of Glial Cells and Synapse Loss in Mouse Models of Alzheimer's Disease. Front Cell Neurosci 2018; 12:473. [PMID: 30618627 PMCID: PMC6297249 DOI: 10.3389/fncel.2018.00473] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/20/2018] [Indexed: 11/13/2022] Open
Abstract
Synapse loss has detrimental effects on cellular communication, leading to network disruptions within the central nervous system (CNS) such as in Alzheimer’s disease (AD). AD is characterized by a progressive decline of memory function, cognition, neuronal and synapse loss. The two main neuropathological hallmarks are amyloid-β (Aβ) plaques and neurofibrillary tangles. In the brain of AD patients and in mouse models of AD several morphological and functional changes, such as microgliosis and astrogliosis around Aβ plaques, as well as dendritic and synaptic alterations, are associated with these lesions. In this review article, we will summarize the current literature on synapse loss in mouse models of AD and discuss current and prospective treatments for AD.
Collapse
Affiliation(s)
- Stephanie Ziegler-Waldkirch
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
45
|
Tosh JL, Rickman M, Rhymes E, Norona FE, Clayton E, Mucke L, Isaacs AM, Fisher EM, Wiseman FK. The integration site of the APP transgene in the J20 mouse model of Alzheimer's disease. Wellcome Open Res 2018; 2:84. [PMID: 29062914 PMCID: PMC5645710 DOI: 10.12688/wellcomeopenres.12237.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 02/02/2023] Open
Abstract
Background: Transgenic animal models are a widely used and powerful tool to investigate human disease and develop therapeutic interventions. Making a transgenic mouse involves random integration of exogenous DNA into the host genome that can have the effect of disrupting endogenous gene expression. The J20 mouse model of Alzheimer's disease (AD) is a transgenic overexpresser of human APP with familial AD mutations and has been extensively utilised in preclinical studies and our aim was to determine the genomic location of the J20 transgene insertion. Methods: We used a combination of breeding strategy and Targeted Locus Amplification with deep sequencing to identify the insertion site of the J20 transgene array. To assess RNA and protein expression of Zbtb20, we used qRT-PCR and Western Blotting. Results: We demonstrate that the J20 transgene construct has inserted within the genetic locus of endogenous mouse gene Zbtb20 on chromosome 16 in an array , disrupting expression of mRNA from this gene in adult hippocampal tissue, while expression of Zbtb20 protein remains unchanged. We note that the endogenous mouse App gene also lies on chromosome 16, although 42 Mb from the Zbtb20 locus. Conclusions: These data will be useful for future studies utilising this popular model of AD, particularly those investigating gene interactions between the J20 APP transgene and other genes present on Mmu16 in the mouse.
Collapse
Affiliation(s)
- Justin L. Tosh
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Matthew Rickman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ellie Rhymes
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Frances E. Norona
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Emma Clayton
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease and University of California, San Francisco, CA, 4158, USA
| | - Adrian M. Isaacs
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Elizabeth M.C. Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| | - Frances K. Wiseman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| |
Collapse
|
46
|
Tosh JL, Rickman M, Rhymes E, Norona FE, Clayton E, Mucke L, Isaacs AM, Fisher EM, Wiseman FK. The integration site of the APP transgene in the J20 mouse model of Alzheimer's disease. Wellcome Open Res 2018; 2:84. [PMID: 29062914 PMCID: PMC5645710 DOI: 10.12688/wellcomeopenres.12237.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2018] [Indexed: 02/02/2023] Open
Abstract
Background: Transgenic animal models are a widely used and powerful tool to investigate human disease and develop therapeutic interventions. Making a transgenic mouse involves random integration of exogenous DNA into the host genome that can have the effect of disrupting endogenous gene expression. The J20 mouse model of Alzheimer's disease (AD) is a transgenic overexpresser of human APP with familial AD mutations and has been extensively utilised in preclinical studies and our aim was to determine the genomic location of the J20 transgene insertion. Methods: We used a combination of breeding strategy and Targeted Locus Amplification with deep sequencing to identify the insertion site of the J20 transgene array. To assess RNA and protein expression of Zbtb20, we used qRT-PCR and Western Blotting. Results: We demonstrate that the J20 transgene construct has inserted within the genetic locus of endogenous mouse gene Zbtb20 on chromosome 16 in an array , disrupting expression of mRNA from this gene in adult hippocampal tissue. Preliminary data suggests that ZBTB20 protein levels remain unchanged in this tissue, however further study is necessary. We note that the endogenous mouse App gene also lies on chromosome 16, although 42 Mb from the Zbtb20 locus. Conclusions: These data will be useful for future studies utilising this popular model of AD, particularly those investigating gene interactions between the J20 APP transgene and other genes present on Mmu16 in the mouse.
Collapse
Affiliation(s)
- Justin L. Tosh
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Matthew Rickman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Ellie Rhymes
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Frances E. Norona
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Emma Clayton
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease and University of California, San Francisco, CA, 4158, USA
| | - Adrian M. Isaacs
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Elizabeth M.C. Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| | - Frances K. Wiseman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK,
| |
Collapse
|
47
|
Caspase-1 inhibition alleviates cognitive impairment and neuropathology in an Alzheimer's disease mouse model. Nat Commun 2018; 9:3916. [PMID: 30254377 PMCID: PMC6156230 DOI: 10.1038/s41467-018-06449-x] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is an intractable progressive neurodegenerative disease characterized by cognitive decline and dementia. An inflammatory neurodegenerative pathway, involving Caspase-1 activation, is associated with human age-dependent cognitive impairment and several classical AD brain pathologies. Here, we show that the nontoxic and blood-brain barrier permeable small molecule Caspase-1 inhibitor VX-765 dose-dependently reverses episodic and spatial memory impairment, and hyperactivity in the J20 mouse model of AD. Cessation of VX-765 results in the reappearance of memory deficits in the mice after 1 month and recommencement of treatment re-establishes normal cognition. VX-765 prevents progressive amyloid beta peptide deposition, reverses brain inflammation, and normalizes synaptophysin protein levels in mouse hippocampus. Consistent with these findings, Caspase-1 null J20 mice are protected from episodic and spatial memory deficits, neuroinflammation and Aβ accumulation. These results provide in vivo proof of concept for Caspase-1 inhibition against AD cognitive deficits and pathologies.
Collapse
|
48
|
Da Mesquita S, Louveau A, Vaccari A, Smirnov I, Cornelison RC, Kingsmore KM, Contarino C, Onengut-Gumuscu S, Farber E, Raper D, Viar KE, Powell RD, Baker W, Dabhi N, Bai R, Cao R, Hu S, Rich SS, Munson JM, Lopes MB, Overall CC, Acton ST, Kipnis J. Functional aspects of meningeal lymphatics in ageing and Alzheimer's disease. Nature 2018; 560:185-191. [PMID: 30046111 PMCID: PMC6085146 DOI: 10.1038/s41586-018-0368-8] [Citation(s) in RCA: 808] [Impact Index Per Article: 134.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 06/15/2018] [Indexed: 01/26/2023]
Abstract
Ageing is a major risk factor for many neurological pathologies, but its mechanisms remain unclear. Unlike other tissues, the parenchyma of the central nervous system (CNS) lacks lymphatic vasculature and waste products are removed partly through a paravascular route. (Re)discovery and characterization of meningeal lymphatic vessels has prompted an assessment of their role in waste clearance from the CNS. Here we show that meningeal lymphatic vessels drain macromolecules from the CNS (cerebrospinal and interstitial fluids) into the cervical lymph nodes in mice. Impairment of meningeal lymphatic function slows paravascular influx of macromolecules into the brain and efflux of macromolecules from the interstitial fluid, and induces cognitive impairment in mice. Treatment of aged mice with vascular endothelial growth factor C enhances meningeal lymphatic drainage of macromolecules from the cerebrospinal fluid, improving brain perfusion and learning and memory performance. Disruption of meningeal lymphatic vessels in transgenic mouse models of Alzheimer's disease promotes amyloid-β deposition in the meninges, which resembles human meningeal pathology, and aggravates parenchymal amyloid-β accumulation. Meningeal lymphatic dysfunction may be an aggravating factor in Alzheimer's disease pathology and in age-associated cognitive decline. Thus, augmentation of meningeal lymphatic function might be a promising therapeutic target for preventing or delaying age-associated neurological diseases.
Collapse
Affiliation(s)
- Sandro Da Mesquita
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
| | - Antoine Louveau
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Andrea Vaccari
- Virginia Image and Video Analysis Laboratory, Department of Electrical and Computer Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Igor Smirnov
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - R Chase Cornelison
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kathryn M Kingsmore
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Christian Contarino
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Department of Mathematics, University of Trento, Povo, Italy
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Emily Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Daniel Raper
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Department of Neurosurgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Kenneth E Viar
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Romie D Powell
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Wendy Baker
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Nisha Dabhi
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Robin Bai
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Rui Cao
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Song Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jennifer M Munson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA, USA
| | - M Beatriz Lopes
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Christopher C Overall
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Scott T Acton
- Virginia Image and Video Analysis Laboratory, Department of Electrical and Computer Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
49
|
Mondragón-Rodríguez S, Gu N, Manseau F, Williams S. Alzheimer's Transgenic Model Is Characterized by Very Early Brain Network Alterations and β-CTF Fragment Accumulation: Reversal by β-Secretase Inhibition. Front Cell Neurosci 2018; 12:121. [PMID: 29867356 PMCID: PMC5952042 DOI: 10.3389/fncel.2018.00121] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/13/2018] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is defined by the presence of amyloid-β (Aβ) and tau protein aggregates. However, increasing data is suggesting that brain network alterations rather than protein deposition could account for the early pathogenesis of the disease. In the present study, we performed in vitro extracellular field recordings in the CA1/subiculum area of the hippocampus from 30 days old J20-TG-AD mice. Here, we found that theta oscillations were significantly less rhythmic than those recorded from control group. In addition, J20 mice displayed significantly less theta-gamma cross-frequency coupling (CFC) as peak modulation indexes for slow (25-45 Hz) and fast (150-250 Hz) gamma frequency oscillations were reduced. Because inhibitory parvalbumin (PV) cells play a vital role in coordinating hippocampal theta and gamma oscillations, whole-cell patch-clamp recordings and extracellular stimulation were performed to access their intrinsic and synaptic properties. Whereas neither the inhibitory output of local interneurons to pyramidal cells (PCs) (inhibitory→PC) nor the excitatory output of PCs to PV cells (PC→PV) differed between control and J20 animals, the intrinsic excitability of PV cells was reduced in J20 mice compared to controls. Interestingly, optogenetic activation of PV interneurons which can directly drive theta oscillations in the hippocampus, did not rescue CFC impairments, suggesting the latter did not simply result from alteration of the underlying theta rhythm. Altered young J20 mice was characterized by the presence of β-CTF, but not with Aβ accumulation, in the hippocampus. Importantly, the β secretase inhibitor AZD3839-AstraZeneca significantly rescued the abnormal early electrophysiological phenotype of J20 mice. In conclusion, our data show that brain network alterations precede the canonical Aβ protein deposition and that, such alterations can be related to β-CTF fragment.
Collapse
Affiliation(s)
- Siddhartha Mondragón-Rodríguez
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- CONACYT National Council for Science and Technology, Mexico City, Mexico
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of Mexico, Querétaro, Mexico
| | - Ning Gu
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Translational Neuroscience, The Royal Mental Health Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Frederic Manseau
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Sylvain Williams
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
50
|
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: 1.8] [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
|