212 results on '"Arancio A"'
Search Results
2. Mitovesicles secreted into the extracellular space of brains with mitochondrial dysfunction impair synaptic plasticity
- Author
-
Pasquale D’Acunzo, Elentina K. Argyrousi, Jonathan M. Ungania, Yohan Kim, Steven DeRosa, Monika Pawlik, Chris N. Goulbourne, Ottavio Arancio, and Efrat Levy
- Subjects
Alzheimer’s disease ,Down syndrome ,Extracellular vesicle ,Exosome ,Long-term potentiation ,MAO-B ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Hypometabolism tied to mitochondrial dysfunction occurs in the aging brain and in neurodegenerative disorders, including in Alzheimer’s disease, in Down syndrome, and in mouse models of these conditions. We have previously shown that mitovesicles, small extracellular vesicles (EVs) of mitochondrial origin, are altered in content and abundance in multiple brain conditions characterized by mitochondrial dysfunction. However, given their recent discovery, it is yet to be explored what mitovesicles regulate and modify, both under physiological conditions and in the diseased brain. In this study, we investigated the effects of mitovesicles on synaptic function, and the molecular players involved. Methods Hippocampal slices from wild-type mice were perfused with the three known types of EVs, mitovesicles, microvesicles, or exosomes, isolated from the brain of a mouse model of Down syndrome or of a diploid control and long-term potentiation (LTP) recorded. The role of the monoamine oxidases type B (MAO-B) and type A (MAO-A) in mitovesicle-driven LTP impairments was addressed by treatment of mitovesicles with the irreversible MAO inhibitors pargyline and clorgiline prior to perfusion of the hippocampal slices. Results Mitovesicles from the brain of the Down syndrome model reduced LTP within minutes of mitovesicle addition. Mitovesicles isolated from control brains did not trigger electrophysiological effects, nor did other types of brain EVs (microvesicles and exosomes) from any genotype tested. Depleting mitovesicles of their MAO-B, but not MAO-A, activity eliminated their ability to alter LTP. Conclusions Mitovesicle impairment of LTP is a previously undescribed paracrine-like mechanism by which EVs modulate synaptic activity, demonstrating that mitovesicles are active participants in the propagation of cellular and functional homeostatic changes in the context of neurodegenerative disorders.
- Published
- 2024
- Full Text
- View/download PDF
3. PDE5 inhibitor drugs for use in dementia
- Author
-
Atticus H. Hainsworth, Ottavio Arancio, Fanny M. Elahi, Jeremy D. Isaacs, and Feixiong Cheng
- Subjects
Alzheimer's disease ,ardenafil ,clinical trials ,dementia ,drugs ,PDE5 inhibitors ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Alzheimer's disease and related dementias (ADRD) remain a major health‐care challenge with few licensed medications. Repurposing existing drugs may afford prevention and treatment. Phosphodiesterase‐5 (PDE5) is widely expressed in vascular myocytes, neurons, and glia. Potent, selective, Food and Drug Administration–approved PDE5 inhibitors are already in clinical use (sildenafil, vardenafil, tadalafil) as vasodilators in erectile dysfunction and pulmonary arterial hypertension. Animal data indicate cognitive benefits of PDE5 inhibitors. In humans, real‐world patient data suggest that sildenafil and vardenafil are associated with reduced dementia risk. While a recent clinical trial of acute tadalafil on cerebral blood flow was neutral, there may be chronic actions of PDE5 inhibition on cerebrovascular or synaptic function. We provide a perspective on the potential utility of PDE5 inhibitors for ADRD. We conclude that further prospective clinical trials with PDE5 inhibitors are warranted. The choice of drug will depend on brain penetration, tolerability in older people, half‐life, and off‐target effects. HIGHLIGHTS Potent phosphodiesterase‐5 (PDE5) inhibitors are in clinical use as vasodilators. In animals PDE5 inhibitors enhance synaptic function and cognitive ability. In humans the PDE5 inhibitor sildenafil is associated with reduced risk of Alzheimer's disease. Licensed PDE5 inhibitors have potential for repurposing in dementia. Prospective clinical trials of PDE5 inhibitors are warranted.
- Published
- 2023
- Full Text
- View/download PDF
4. The ketamine metabolite (2R,6R)‐hydroxynorketamine rescues hippocampal mRNA translation, synaptic plasticity and memory in mouse models of Alzheimer's disease.
- Author
-
Ribeiro, Felipe C., Cozachenco, Danielle, Argyrousi, Elentina K., Staniszewski, Agnieszka, Wiebe, Shane, Calixtro, Joao D., Soares‐Neto, Rubens, Al‐Chami, Aycheh, Sayegh, Fatema El, Bermudez, Sara, Arsenault, Emily, Cossenza, Marcelo, Lacaille, Jean‐Claude, Nader, Karim, Sun, Hongyu, De Felice, Fernanda G., Lourenco, Mychael V., Arancio, Ottavio, Aguilar‐Valles, Argel, and Sonenberg, Nahum
- Abstract
INTRODUCTION: Impaired brain protein synthesis, synaptic plasticity, and memory are major hallmarks of Alzheimer's disease (AD). The ketamine metabolite (2R,6R)‐hydroxynorketamine (HNK) has been shown to modulate protein synthesis, but its effects on memory in AD models remain elusive. METHODS: We investigated the effects of HNK on hippocampal protein synthesis, long‐term potentiation (LTP), and memory in AD mouse models. RESULTS: HNK activated extracellular signal‐regulated kinase 1/2 (ERK1/2), mechanistic target of rapamycin (mTOR), and p70S6 kinase 1 (S6K1)/ribosomal protein S6 signaling pathways. Treatment with HNK rescued hippocampal LTP and memory deficits in amyloid‐β oligomers (AβO)‐infused mice in an ERK1/2‐dependent manner. Treatment with HNK further corrected aberrant transcription, LTP and memory in aged APP/PS1 mice. DISCUSSION: Our findings demonstrate that HNK induces signaling and transcriptional responses that correct synaptic and memory deficits in AD mice. These results raise the prospect that HNK could serve as a therapeutic approach in AD. Highlights: The ketamine metabolite HNK activates hippocampal ERK/mTOR/S6 signaling pathways.HNK corrects hippocampal synaptic and memory defects in two mouse models of AD.Rescue of synaptic and memory impairments by HNK depends on ERK signaling.HNK corrects aberrant transcriptional signatures in APP/PS1 mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Alzheimer's disease as an autoimmune disorder of innate immunity endogenously modulated by tryptophan metabolites
- Author
-
Felix S. Meier‐Stephenson, Vanessa C. Meier‐Stephenson, Michael D. Carter, Autumn R. Meek, Yanfei Wang, Luzhe Pan, Qiangwei Chen, Sheila Jacobo, Fan Wu, Erhu Lu, Gordon A. Simms, Laural Fisher, Alaina J. McGrath, Virgil Fermo, Christopher J. Barden, Harman D.S. Clair, Todd N. Galloway, Arun Yadav, Valérie Campágna‐Slater, Mark Hadden, Mark Reed, Marcia Taylor, Brendan Kelly, Elena Diez‐Cecilia, Igri Kolaj, Clarissa Santos, Imindu Liyanage, Braden Sweeting, Paul Stafford, Robert Boudreau, G. Andrew Reid, Ryan S. Noyce, Leanne Stevens, Agnieszka Staniszewski, Hong Zhang, Mamidanna R. V. S. Murty, Pascale Lemaire, Solenne Chardonnet, Christopher D. Richardson, Valérie Gabelica, Edwin DePauw, Richard Brown, Sultan Darvesh, Ottavio Arancio, and Donald F. Weaver
- Subjects
Alzheimer's disease ,amyloid beta ,antimicrobial peptide ,arginine ,autoimmune ,cytokine ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Introduction Alzheimer's disease (AD) is characterized by neurotoxic immuno‐inflammation concomitant with cytotoxic oligomerization of amyloid beta (Aβ) and tau, culminating in concurrent, interdependent immunopathic and proteopathic pathogeneses. Methods We performed a comprehensive series of in silico, in vitro, and in vivo studies explicitly evaluating the atomistic–molecular mechanisms of cytokine‐mediated and Aβ‐mediated neurotoxicities in AD. Next, 471 new chemical entities were designed and synthesized to probe the pathways identified by these molecular mechanism studies and to provide prototypic starting points in the development of small‐molecule therapeutics for AD. Results In response to various stimuli (e.g., infection, trauma, ischemia, air pollution, depression), Aβ is released as an early responder immunopeptide triggering an innate immunity cascade in which Aβ exhibits both immunomodulatory and antimicrobial properties (whether bacteria are present, or not), resulting in a misdirected attack upon “self” neurons, arising from analogous electronegative surface topologies between neurons and bacteria, and rendering them similarly susceptible to membrane‐penetrating attack by antimicrobial peptides (AMPs) such as Aβ. After this self‐attack, the resulting necrotic (but not apoptotic) neuronal breakdown products diffuse to adjacent neurons eliciting further release of Aβ, leading to a chronic self‐perpetuating autoimmune cycle. AD thus emerges as a brain‐centric autoimmune disorder of innate immunity. Based upon the hypothesis that autoimmune processes are susceptible to endogenous regulatory processes, a subsequent comprehensive screening program of 1137 small molecules normally present in human brain identified tryptophan metabolism as a regulator of brain innate immunity and a source of potential endogenous anti‐AD molecules capable of chemical modification into multi‐site therapeutic modulators targeting AD's complex immunopathic–proteopathic pathogenesis. Discussion Conceptualizing AD as an autoimmune disease, identifying endogenous regulators of this autoimmunity, and designing small molecule drug‐like analogues of these endogenous regulators represents a novel therapeutic approach for AD.
- Published
- 2022
- Full Text
- View/download PDF
6. Alzheimer's Therapeutics Targeting Amyloid Beta 1–42 Oligomers II: Sigma-2/PGRMC1 Receptors Mediate Abeta 42 Oligomer Binding and Synaptotoxicity
- Author
-
Izzo, Nicholas J, Xu, Jinbin, Zeng, Chenbo, Kirk, Molly J, Mozzoni, Kelsie, Silky, Colleen, Rehak, Courtney, Yurko, Raymond, Look, Gary, Rishton, Gilbert, Safferstein, Hank, Cruchaga, Carlos, Goate, Alison, Cahill, Michael A, Arancio, Ottavio, Mach, Robert H, Craven, Rolf, Head, Elizabeth, LeVine, Harry, Spires-Jones, Tara L, and Catalano, Susan M
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Dementia ,Acquired Cognitive Impairment ,Aging ,Brain Disorders ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Alzheimer's Disease ,Neurosciences ,Neurodegenerative ,Aetiology ,2.1 Biological and endogenous factors ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Neurological ,Alzheimer Disease ,Amyloid beta-Peptides ,Animals ,Autoradiography ,Brain ,Cell Membrane ,Cognition ,Cognition Disorders ,Humans ,Membrane Proteins ,Mice ,Neurons ,Peptide Fragments ,Protein Binding ,Protein Structure ,Tertiary ,RNA ,Small Interfering ,Rats ,Rats ,Sprague-Dawley ,Receptors ,Progesterone ,Synapses ,General Science & Technology - Abstract
Amyloid beta (Abeta) 1-42 oligomers accumulate in brains of patients with Mild Cognitive Impairment (MCI) and disrupt synaptic plasticity processes that underlie memory formation. Synaptic binding of Abeta oligomers to several putative receptor proteins is reported to inhibit long-term potentiation, affect membrane trafficking and induce reversible spine loss in neurons, leading to impaired cognitive performance and ultimately to anterograde amnesia in the early stages of Alzheimer's disease (AD). We have identified a receptor not previously associated with AD that mediates the binding of Abeta oligomers to neurons, and describe novel therapeutic antagonists of this receptor capable of blocking Abeta toxic effects on synapses in vitro and cognitive deficits in vivo. Knockdown of sigma-2/PGRMC1 (progesterone receptor membrane component 1) protein expression in vitro using siRNA results in a highly correlated reduction in binding of exogenous Abeta oligomers to neurons of more than 90%. Expression of sigma-2/PGRMC1 is upregulated in vitro by treatment with Abeta oligomers, and is dysregulated in Alzheimer's disease patients' brain compared to age-matched, normal individuals. Specific, high affinity small molecule receptor antagonists and antibodies raised against specific regions on this receptor can displace synthetic Abeta oligomer binding to synaptic puncta in vitro and displace endogenous human AD patient oligomers from brain tissue sections in a dose-dependent manner. These receptor antagonists prevent and reverse the effects of Abeta oligomers on membrane trafficking and synapse loss in vitro and cognitive deficits in AD mouse models. These findings suggest sigma-2/PGRMC1 receptors mediate saturable oligomer binding to synaptic puncta on neurons and that brain penetrant, small molecules can displace endogenous and synthetic oligomers and improve cognitive deficits in AD models. We propose that sigma-2/PGRMC1 is a key mediator of the pathological effects of Abeta oligomers in AD and is a tractable target for small molecule disease-modifying therapeutics.
- Published
- 2014
7. Synthesis and Characterization of Click Chemical Probes for Single-Cell Resolution Detection of Epichaperomes in Neurodegenerative Disorders.
- Author
-
Bay, Sadik, Digwal, Chander S., Rodilla Martín, Ananda M., Sharma, Sahil, Stanisavljevic, Aleksandra, Rodina, Anna, Attaran, Anoosha, Roychowdhury, Tanaya, Parikh, Kamya, Toth, Eugene, Panchal, Palak, Rosiek, Eric, Pasala, Chiranjeevi, Arancio, Ottavio, Fraser, Paul E., Alldred, Melissa J., Prado, Marco A. M., Ginsberg, Stephen D., and Chiosis, Gabriela
- Subjects
NEURODEGENERATION ,ALZHEIMER'S disease ,PATHOLOGY ,PARKINSON'S disease ,DRUG discovery ,MOLECULAR probes - Abstract
Neurodegenerative disorders, including Alzheimer's disease (AD) and Parkinson's disease (PD), represent debilitating conditions with complex, poorly understood pathologies. Epichaperomes, pathologic protein assemblies nucleated on key chaperones, have emerged as critical players in the molecular dysfunction underlying these disorders. In this study, we introduce the synthesis and characterization of clickable epichaperome probes, PU-TCO, positive control, and PU-NTCO, negative control. Through comprehensive in vitro assays and cell-based investigations, we establish the specificity of the PU-TCO probe for epichaperomes. Furthermore, we demonstrate the efficacy of PU-TCO in detecting epichaperomes in brain tissue with a cellular resolution, underscoring its potential as a valuable tool for dissecting single-cell responses in neurodegenerative diseases. This clickable probe is therefore poised to address a critical need in the field, offering unprecedented precision and versatility in studying epichaperomes and opening avenues for novel insights into their role in disease pathology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. The 5HT2b Receptor in Alzheimer's Disease: Increased Levels in Patient Brains and Antagonist Attenuation of Amyloid and Tau Induced Dysfunction.
- Author
-
Acquarone, Erica, Argyrousi, Elentina K., Arancio, Ottavio, Watterson, D. Martin, and Roy, Saktimayee M.
- Subjects
ALZHEIMER'S disease ,TAU proteins ,LONG-term potentiation ,CHEMICAL biology ,AMYLOID ,ASSOCIATIVE memory (Psychology) - Abstract
BACKGROUND: Background: Neurodegenerative diseases manifest behavioral dysfunction with disease progression. Intervention with neuropsychiatric drugs is part of most multi-drug treatment paradigms. However, only a fraction of patients responds to the treatments and those responding must deal with drug-drug interactions and tolerance issues generally attributed to off-target activities. Recent efforts have focused on the identification of underexplored targets and exploration of improved outcomes by treatment with selective molecular probes. Objective: As part of ongoing efforts to identify and validate additional targets amenable to therapeutic intervention, we examined levels of the serotonin 5-HT2b receptor (5-HT2bR) in Alzheimer's disease (AD) brains and the potential of a selective 5-HT2bR antagonist to counteract synaptic plasticity and memory damage induced by AD-related proteins, amyloid-β, and tau. Methods: This work used a combination of biochemical, chemical biology, electrophysiological, and behavioral techniques. Biochemical methods included analysis of protein levels. Chemical biology methods included the use of an in vivo molecular probe MW071, a selective antagonist for the 5HT2bR. Electrophysiological methods included assessment of long-term potentiation (LTP), a type of synaptic plasticity thought to underlie memory formation. Behavioral studies investigated spatial memory and associative memory. Results: 5HT2bR levels are increased in brain specimens of AD patients compared to controls. 5HT2bR antagonist treatment rescued amyloid-β and tau oligomer-induced impairment of synaptic plasticity and memory. Conclusions: The increased levels of 5HT-2bR in AD patient brains and the attenuation of disease-related synaptic and behavioral dysfunctions by MW071 treatment suggest that the 5HT-2bR is a molecular target worth pursuing as a potential therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Mitovesicles secreted into the extracellular space of brains with mitochondrial dysfunction impair synaptic plasticity.
- Author
-
D'Acunzo, Pasquale, Argyrousi, Elentina K., Ungania, Jonathan M., Kim, Yohan, DeRosa, Steven, Pawlik, Monika, Goulbourne, Chris N., Arancio, Ottavio, and Levy, Efrat
- Subjects
EXTRACELLULAR space ,NEUROPLASTICITY ,EXTRACELLULAR vesicles ,MITOCHONDRIA ,ALZHEIMER'S disease ,THETA rhythm - Abstract
Background: Hypometabolism tied to mitochondrial dysfunction occurs in the aging brain and in neurodegenerative disorders, including in Alzheimer's disease, in Down syndrome, and in mouse models of these conditions. We have previously shown that mitovesicles, small extracellular vesicles (EVs) of mitochondrial origin, are altered in content and abundance in multiple brain conditions characterized by mitochondrial dysfunction. However, given their recent discovery, it is yet to be explored what mitovesicles regulate and modify, both under physiological conditions and in the diseased brain. In this study, we investigated the effects of mitovesicles on synaptic function, and the molecular players involved. Methods: Hippocampal slices from wild-type mice were perfused with the three known types of EVs, mitovesicles, microvesicles, or exosomes, isolated from the brain of a mouse model of Down syndrome or of a diploid control and long-term potentiation (LTP) recorded. The role of the monoamine oxidases type B (MAO-B) and type A (MAO-A) in mitovesicle-driven LTP impairments was addressed by treatment of mitovesicles with the irreversible MAO inhibitors pargyline and clorgiline prior to perfusion of the hippocampal slices. Results: Mitovesicles from the brain of the Down syndrome model reduced LTP within minutes of mitovesicle addition. Mitovesicles isolated from control brains did not trigger electrophysiological effects, nor did other types of brain EVs (microvesicles and exosomes) from any genotype tested. Depleting mitovesicles of their MAO-B, but not MAO-A, activity eliminated their ability to alter LTP. Conclusions: Mitovesicle impairment of LTP is a previously undescribed paracrine-like mechanism by which EVs modulate synaptic activity, demonstrating that mitovesicles are active participants in the propagation of cellular and functional homeostatic changes in the context of neurodegenerative disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Nitric oxide/cGMP/CREB pathway and amyloid-beta crosstalk: From physiology to Alzheimer's disease
- Author
-
Maria Rosaria Tropea, Walter Gulisano, Valeria Vacanti, Ottavio Arancio, Daniela Puzzo, and Agostino Palmeri
- Subjects
Amyloid beta-Peptides ,Nitric oxide/cGMP/CREB pathway ,Alzheimer's disease ,Nitric Oxide ,Biochemistry ,Synaptic plasticity ,Alzheimer Disease ,Memory ,Physiology (medical) ,Phosphodiesterase inhibitors ,Humans ,Amyloid-β ,Cyclic GMP ,Signal Transduction - Abstract
The nitric oxide (NO)/cGMP pathway has been extensively studied for its pivotal role in synaptic plasticity and memory processes, resulting in an increase of cAMP response element-binding (CREB) phosphorylation, and consequent synthesis of plasticity-related proteins. The NO/cGMP/CREB signaling is downregulated during aging and neurodegenerative disorders and is affected by Amyloid-β peptide (Aβ) and tau protein, whose increase and deposition is considered the key pathogenic event of Alzheimer's disease (AD). On the other hand, in physiological conditions, the crosstalk between the NO/cGMP/PKG/CREB pathway and Aβ ensures long-term potentiation and memory formation. This review summarizes the current knowledge on the interaction between the NO/cGMP/PKG/CREB pathway and Aβ in the healthy and diseased brain, offering a new perspective to shed light on AD pathophysiology. We will focus on the synaptic mechanisms underlying Aβ physiological interplay with cGMP pathway and how this balance is corrupted in AD, as high levels of Aβ interfere with NO production and cGMP molecular signaling leading to cognitive impairment. Finally, we will discuss results from preclinical and clinical studies proposing the increase of cGMP signaling as a therapeutic strategy in the treatment of AD.
- Published
- 2022
- Full Text
- View/download PDF
11. Re-Arranging the Puzzle between the Amyloid-Beta and Tau Pathology: An APP-Centric Approach.
- Author
-
Haut, Florence, Argyrousi, Elentina K., and Arancio, Ottavio
- Subjects
TAU proteins ,AMYLOID beta-protein precursor ,ALZHEIMER'S disease - Abstract
After several years of research in the field of Alzheimer's disease (AD), it is still unclear how amyloid-beta (Aβ) and Tau, two key hallmarks of the disease, mediate the neuropathogenic events that lead to AD. Current data challenge the "Amyloid Cascade Hypothesis" that has prevailed in the field of AD, stating that Aβ precedes and triggers Tau pathology that will eventually become the toxic entity in the progression of the disease. This perspective also led the field of therapeutic approaches towards the development of strategies that target Aβ or Tau. In the present review, we discuss recent literature regarding the neurotoxic role of both Aβ and Tau in AD, as well as their physiological function in the healthy brain. Consequently, we present studies suggesting that Aβ and Tau act independently of each other in mediating neurotoxicity in AD, thereafter, re-evaluating the "Amyloid Cascade Hypothesis" that places Tau pathology downstream of Aβ. More recent studies have confirmed that both Aβ and Tau could propagate the disease and induce synaptic and memory impairments via the amyloid precursor protein (APP). This finding is not only interesting from a mechanistic point of view since it provides better insights into the AD pathogenesis but also from a therapeutic point of view since it renders APP a common downstream effector for both Aβ and Tau. Subsequently, therapeutic strategies that act on APP might provide a more viable and physiologically relevant approach for targeting AD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. CRISPR/Cas9-Correctable mutation-related molecular and physiological phenotypes in iPSC-derived Alzheimer’s PSEN2 N141I neurons
- Author
-
Maitane Ortiz-Virumbrales, Cesar L. Moreno, Ilya Kruglikov, Paula Marazuela, Andrew Sproul, Samson Jacob, Matthew Zimmer, Daniel Paull, Bin Zhang, Eric E. Schadt, Michelle E. Ehrlich, Rudolph E. Tanzi, Ottavio Arancio, Scott Noggle, and Sam Gandy
- Subjects
Alzheimer’s disease ,iPSC ,BFCN ,CRISPR/Cas9 ,Electrophysiology ,Basal forebrain ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Basal forebrain cholinergic neurons (BFCNs) are believed to be one of the first cell types to be affected in all forms of AD, and their dysfunction is clinically correlated with impaired short-term memory formation and retrieval. We present an optimized in vitro protocol to generate human BFCNs from iPSCs, using cell lines from presenilin 2 (PSEN2) mutation carriers and controls. As expected, cell lines harboring the PSEN2 N141I mutation displayed an increase in the Aβ42/40 in iPSC-derived BFCNs. Neurons derived from PSEN2 N141I lines generated fewer maximum number of spikes in response to a square depolarizing current injection. The height of the first action potential at rheobase current injection was also significantly decreased in PSEN2 N141I BFCNs. CRISPR/Cas9 correction of the PSEN2 point mutation abolished the electrophysiological deficit, restoring both the maximal number of spikes and spike height to the levels recorded in controls. Increased Aβ42/40 was also normalized following CRISPR/Cas-mediated correction of the PSEN2 N141I mutation. The genome editing data confirms the robust consistency of mutation-related changes in Aβ42/40 ratio while also showing a PSEN2-mutation-related alteration in electrophysiology.
- Published
- 2017
- Full Text
- View/download PDF
13. PP2A methylation controls sensitivity and resistance to β-amyloid–induced cognitive and electrophysiological impairments
- Author
-
Nicholls, Russell E., Sontag, Jean-Marie, Zhang, Hong, Staniszewski, Agnieszka, Yan, Shijun, Kim, Carla Y., Yim, Michael, Woodruff, Caitlin M., Arning, Erland, Wasek, Brandi, Yin, Deqi, Bottiglieri, Teodoro, Sontag, Estelle, Kandel, Eric R., and Arancio, Ottavio
- Published
- 2016
14. Synaptic and memory dysfunction induced by tau oligomers is rescued by up-regulation of the nitric oxide cascade
- Author
-
Acquarone, Erica, Argyrousi, Elentina K., van den Berg, Manon, Gulisano, Walter, Fà, Mauro, Staniszewski, Agnieszka, Calcagno, Elisa, Zuccarello, Elisa, D’Adamio, Luciano, Deng, Shi-Xian, Puzzo, Daniela, Arancio, Ottavio, and Fiorito, Jole
- Published
- 2019
- Full Text
- View/download PDF
15. SUMO modulation of protein aggregation and degradation
- Author
-
Marco Feligioni, Serena Marcelli, Erin Knock, Urooba Nadeem, Ottavio Arancio, and Paul E. Fraser
- Subjects
SUMO ,neurodegeneration ,Alzheimer's disease ,Parkinson's disease ,Amyotrophic lateral sclerosis ,Huntington's disease ,tau ,amyloid ,synuclein ,superoxide dismutase ,TAR DNA-binding protein-43 ,Biology (General) ,QH301-705.5 - Abstract
Small ubiquitin-like modifier (SUMO) conjugation and binding to target proteins regulate a wide variety of cellular pathways. The functional aspects of SUMOylation include changes in protein-protein interactions, intracellular trafficking as well as protein aggregation and degradation. SUMO has also been linked to specialized cellular pathways such as neuronal development and synaptic transmission. In addition, SUMOylation is associated with neurological diseases associated with abnormal protein accumulations. SUMOylation of the amyloid and tau proteins involved in Alzheimer's disease and other tauopathies may contribute to changes in protein solubility and proteolytic processing. Similar events have been reported for α-synuclein aggregates found in Parkinson's disease, polyglutamine disorders such as Huntington's disease as well as protein aggregates found in amyotrophic lateral sclerosis (ALS). This review provides a detailed overview of the impact SUMOylation has on the etiology and pathology of these related neurological diseases.
- Published
- 2015
- Full Text
- View/download PDF
16. PDE5 inhibitor drugs for use in dementia.
- Author
-
Hainsworth, Atticus H., Arancio, Ottavio, Elahi, Fanny M., Isaacs, Jeremy D., and Cheng, Feixiong
- Subjects
PHOSPHODIESTERASE-5 inhibitors ,PHOSPHODIESTERASE inhibitors ,ALZHEIMER'S disease ,PULMONARY arterial hypertension ,CEREBRAL circulation - Abstract
Alzheimer's disease and related dementias (ADRD) remain a major health‐care challenge with few licensed medications. Repurposing existing drugs may afford prevention and treatment. Phosphodiesterase‐5 (PDE5) is widely expressed in vascular myocytes, neurons, and glia. Potent, selective, Food and Drug Administration–approved PDE5 inhibitors are already in clinical use (sildenafil, vardenafil, tadalafil) as vasodilators in erectile dysfunction and pulmonary arterial hypertension. Animal data indicate cognitive benefits of PDE5 inhibitors. In humans, real‐world patient data suggest that sildenafil and vardenafil are associated with reduced dementia risk. While a recent clinical trial of acute tadalafil on cerebral blood flow was neutral, there may be chronic actions of PDE5 inhibition on cerebrovascular or synaptic function. We provide a perspective on the potential utility of PDE5 inhibitors for ADRD. We conclude that further prospective clinical trials with PDE5 inhibitors are warranted. The choice of drug will depend on brain penetration, tolerability in older people, half‐life, and off‐target effects. HIGHLIGHTS: Potent phosphodiesterase‐5 (PDE5) inhibitors are in clinical use as vasodilators.In animals PDE5 inhibitors enhance synaptic function and cognitive ability.In humans the PDE5 inhibitor sildenafil is associated with reduced risk of Alzheimer's disease.Licensed PDE5 inhibitors have potential for repurposing in dementia.Prospective clinical trials of PDE5 inhibitors are warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Mitophagy Failure in Fibroblasts and iPSC-Derived Neurons of Alzheimer’s Disease-Associated Presenilin 1 Mutation
- Author
-
Patricia Martín-Maestro, Ricardo Gargini, Andrew A. Sproul, Esther García, Luis C. Antón, Scott Noggle, Ottavio Arancio, Jesús Avila, and Vega García-Escudero
- Subjects
Alzheimer’s disease ,presenilin 1 mutation ,mitophagy ,fibroblasts ,iPSC derived neurons ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Familial Alzheimer’s disease (FAD) is clearly related with the accumulation of amyloid-beta (Aβ) and its deleterious effect on mitochondrial function is well established. Anomalies in autophagy have also been described in these patients. In the present work, functional analyses have been performed to study mitochondrial recycling process in patient-derived fibroblasts and neurons from induced pluripotent stem cells harboring the presenilin 1 mutation A246E. Mitophagy impairment was observed due to a diminished autophagy degradation phase associated with lysosomal anomalies, thus causing the accumulation of dysfunctional mitochondria labeled by Parkin RBR E3 ubiquitin protein ligase (PARK2). The failure of mitochondrial recycling by autophagy was enhanced in the patient-derived neuronal model. Our previous studies have demonstrated similar mitophagy impairment in sporadic Alzheimer’s disease (AD); therefore, our data indicate that mitophagy deficiency should be considered a common nexus between familial and sporadic cases of the disease.
- Published
- 2017
- Full Text
- View/download PDF
18. LTP and memory impairment caused by extracellular Aβ and Tau oligomers is APP-dependent
- Author
-
Daniela Puzzo, Roberto Piacentini, Mauro Fá, Walter Gulisano, Domenica D Li Puma, Agnes Staniszewski, Hong Zhang, Maria Rosaria Tropea, Sara Cocco, Agostino Palmeri, Paul Fraser, Luciano D'Adamio, Claudio Grassi, and Ottavio Arancio
- Subjects
amyloid-beta ,tau ,APP ,synaptic plasticity ,memory ,Alzheimer's disease ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The concurrent application of subtoxic doses of soluble oligomeric forms of human amyloid-beta (oAβ) and Tau (oTau) proteins impairs memory and its electrophysiological surrogate long-term potentiation (LTP), effects that may be mediated by intra-neuronal oligomers uptake. Intrigued by these findings, we investigated whether oAβ and oTau share a common mechanism when they impair memory and LTP in mice. We found that as already shown for oAβ, also oTau can bind to amyloid precursor protein (APP). Moreover, efficient intra-neuronal uptake of oAβ and oTau requires expression of APP. Finally, the toxic effect of both extracellular oAβ and oTau on memory and LTP is dependent upon APP since APP-KO mice were resistant to oAβ- and oTau-induced defects in spatial/associative memory and LTP. Thus, APP might serve as a common therapeutic target against Alzheimer's Disease (AD) and a host of other neurodegenerative diseases characterized by abnormal levels of Aβ and/or Tau.
- Published
- 2017
- Full Text
- View/download PDF
19. Leucine Carboxyl Methyltransferase 1 Overexpression Protects Against Cognitive and Electrophysiological Impairments in Tg2576 APP Transgenic Mice
- Author
-
Ottavio Arancio, Rose Pitstick, Michael P. Kavanaugh, Russell E. Nicholls, Agnieszka Staniszewski, Madhumathi Gnanaprakash, and Hong Zhang
- Subjects
0301 basic medicine ,Genetically modified mouse ,Methyltransferase ,Transgene ,Phosphatase ,Mice, Transgenic ,Biology ,amyloid-β ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Alzheimer Disease ,Animals ,Humans ,Cognitive Dysfunction ,protein phosphatase methylesterase ,long-term potentiation ,cognitive impairment ,Amyloid beta-Peptides ,leucine carboxyl methyltransferase ,General Neuroscience ,protein phosphatase 2A ,Leucine carboxyl methyltransferase 1 ,Long-term potentiation ,General Medicine ,Protein phosphatase 2 ,Protein O-Methyltransferase ,Cell biology ,MAPT protein ,Psychiatry and Mental health ,Clinical Psychology ,Disease Models, Animal ,030104 developmental biology ,Synaptic plasticity ,Geriatrics and Gerontology ,Alzheimer’s disease ,Carboxylic Ester Hydrolases ,030217 neurology & neurosurgery ,Research Article - Abstract
Background: The serine/threonine protein phosphatase, PP2A, is thought to play a central role in the molecular pathogenesis of Alzheimer’s disease (AD), and the activity and substrate specificity of PP2A is regulated, in part, through methylation and demethylation of its catalytic subunit. Previously, we found that transgenic overexpression of the PP2A methyltransferase, LCMT-1, or the PP2A methylesterase, PME-1, altered the sensitivity of mice to impairments caused by acute exposure to synthetic oligomeric amyloid-β (Aβ). Objective: Here we sought to test the possibility that these molecules also controlled sensitivity to impairments caused by chronically elevated levels of Aβ produced in vivo. Methods: To do this, we examined the effects of transgenic LCMT-1, or PME-1 overexpression on cognitive and electrophysiological impairments caused by chronic overexpression of mutant human APP in Tg2576 mice. Results: We found that LCMT-1 overexpression prevented impairments in short-term spatial memory and synaptic plasticity in Tg2576 mice, without altering APP expression or soluble Aβ levels. While the magnitude of the effects of PME-1 overexpression in Tg2576 mice was small and potentially confounded by the emergence of non-cognitive impairments, Tg2576 mice that overexpressed PME-1 showed a trend toward earlier onset and/or increased severity of cognitive and electrophysiological impairments. Conclusion: These data suggest that the PP2A methyltransferase, LCMT-1, and the PP2A methylesterase, PME-1, may participate in the molecular pathogenesis of AD by regulating sensitivity to the pathogenic effects of chronically elevated levels of Aβ.
- Published
- 2021
20. AAV-mediated neuronal expression of an scFv antibody selective for Aβ oligomers protects synapses and rescues memory in Alzheimer models
- Author
-
Maria Clara Selles, Juliana T.S. Fortuna, Magali C. Cercato, Luis Eduardo Santos, Luciana Domett, Andre L.B. Bitencourt, Mariane Favero Carraro, Amanda S. Souza, Helena Janickova, Caroline Vieira Azevedo, Henrique Correia Campos, Jorge M. de Souza, Soniza Alves-Leon, Vania F. Prado, Marco A.M. Prado, Alberto L. Epstein, Anna Salvetti, Beatriz Monteiro Longo, Ottavio Arancio, William L. Klein, Adriano Sebollela, Fernanda G. De Felice, Diana A. Jerusalinsky, Sergio T. Ferreira, New York University School of Medicine (NYU Grossman School of Medicine), Universidade Federal do Rio de Janeiro (UFRJ), Universidad de Buenos Aires [Buenos Aires] (UBA), Universidade de São Paulo = University of São Paulo (USP), University of Western Ontario (UWO), Universidade Federal de São Paulo, Hospital Universitário Clementino Fraga Filho [Rio de Janeiro] (HUCFF / UFRJ), Handicap neuromusculaire : Physiopathologie, Biothérapie et Pharmacologies appliquées (END-ICAP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Columbia University [New York], Northwestern University [Evanston], Queen's University [Kingston, Canada], 16/820,269, Northwestern University, NU, Canadian Institutes of Health Research, IRSC: PJT 159781, PJT 162431, Alzheimer Society, Fundação de Amparo à Pesquisa do Estado de São Paulo, FAPESP: 2014/25681-3, Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, CAPES, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Conselho Nacional de Desenvolvimento Científico e Tecnológico, CNPq, Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro, FAPERJ, Instituto Nacional de Ciência e Tecnologia de Neurociência Translacional, INCT-INNT, International Society for Neurochemistry, ISN, S.T.F. and F.G.D.F. were supported by grants from the National Council for Scientific and Technological Development (CNPq/Brazil), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ/Brazil), and National Institute of Translational Neuroscience (Brazil). S.T.F. and D.A.J. were jointly supported by a binational research grant from CNPq/CONICET. A. Sebollela was supported by the São Paulo Research Foundation (FAPESP) grant 2014/25681-3 . M.A.M.P. and V.F.P. received support from the Alzheimer’s Society of Canada and the Canadian Institutes of Health Research (CIHR, PJT 162431 and PJT 159781 ). A.L.B.B. received a predoctoral fellowship from CNPq. M.C.S. received predoctoral fellowships from CNPq and FAPERJ and travel grants from the International Society for Neurochemistry , Company of Biologists , and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior . We thank Drs. Moses Chao and Mauricio M. Oliveira for critical reading of the manuscript and discussions., S.T.F. and F.G.D.F. were supported by grants from the National Council for Scientific and Technological Development (CNPq/Brazil), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ/Brazil), and National Institute of Translational Neuroscience (Brazil). S.T.F. and D.A.J. were jointly supported by a binational research grant from CNPq/CONICET. A. Sebollela was supported by the São Paulo Research Foundation (FAPESP) grant 2014/25681-3. M.A.M.P. and V.F.P. received support from the Alzheimer's Society of Canada and the Canadian Institutes of Health Research (CIHR, PJT 162431 and PJT 159781). A.L.B.B. received a predoctoral fellowship from CNPq. M.C.S. received predoctoral fellowships from CNPq and FAPERJ and travel grants from the International Society for Neurochemistry, Company of Biologists, and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior. We thank Drs. Moses Chao and Mauricio M. Oliveira for critical reading of the manuscript and discussions. M.C.S. D.A.J. and S.T.F. designed the study. A. Sebollela, W.L.K. and S.T.F. developed the recombinant NUsc1 scFv. D.A.J. A.L.E. A. Salvetti, and S.T.F. conceived constructs and vector development. M.C.S. J.T.S.F. M.C.C. L.E.S. L.D. A.L.B.B. M.F.C. A.S.S. H.J. C.V.A. and H.C.C. performed research. M.C.S. J.T.S.F. M.C.C. L.E.S. L.D. A.L.B.B. O.A. F.G.D.F. and S.T.F analyzed data. J.M.S. S.A.L. V.F.P. M.A.M.P. A.L.E. A. Salvetti, A. Sebollela, and W.L.K. contributed reagents, materials, and analysis tools. M.C.S. J.T.S.F. L.E.S. A.L.E. A. Salvetti, A. Sebollela, B.M.L, O.A. W.L.K. F.G.D.F. D.A.J. and S.T.F. analyzed and discussed the results. M.C.S. W.L.K. and S.T.F. wrote the manuscript with contributions from other authors. A patent application covering the use of AAV-NUsc1 in Alzheimer's disease has been filed with the USPTO (16/820,269, and pending) by Northwestern University with S.T.F. W.L.K. D.A.J. A. Sebollela, and M.C.S. as named inventors.
- Subjects
memory ,NUsc1 ,Pharmacology ,AβOs ,[SDV]Life Sciences [q-bio] ,Drug Discovery ,Genetics ,Molecular Medicine ,AAV ,Alzheimer's disease ,immuno-gene therapy ,Molecular Biology ,scFv - Abstract
International audience; The accumulation of soluble oligomers of the amyloid-β peptide (AβOs) in the brain has been implicated in synapse failure and memory impairment in Alzheimer's disease. Here, we initially show that treatment with NUsc1, a single-chain variable-fragment antibody (scFv) that selectively targets a subpopulation of AβOs and shows minimal reactivity to Aβ monomers and fibrils, prevents the inhibition of long-term potentiation in hippocampal slices and memory impairment induced by AβOs in mice. As a therapeutic approach for intracerebral antibody delivery, we developed an adeno-associated virus vector to drive neuronal expression of NUsc1 (AAV-NUsc1) within the brain. Transduction by AAV-NUsc1 induced NUsc1 expression and secretion in adult human brain slices and inhibited AβO binding to neurons and AβO-induced loss of dendritic spines in primary rat hippocampal cultures. Treatment of mice with AAV-NUsc1 prevented memory impairment induced by AβOs and, remarkably, reversed memory deficits in aged APPswe/PS1ΔE9 Alzheimer's disease model mice. These results support the feasibility of immunotherapy using viral vector-mediated gene delivery of NUsc1 or other AβO-specific single-chain antibodies as a potential therapeutic approach in Alzheimer's disease.
- Published
- 2022
- Full Text
- View/download PDF
21. Synaptojanin 1-Linked Phosphoinositide Dyshomeostasis and Cognitive Deficits in Mouse Models of Down's Syndrome
- Author
-
Voronov, Sergey V., Frere, Samuel G., Giovedi, Silvia, Pollina, Elizabeth A., Borel, Christelle, Zhang, Hong, Schmidt, Cecilia, Akeson, Ellen C., Wenk, Markus R., Cimasoni, Laurent, Arancio, Ottavio, Davisson, Muriel T., Antonarakis, Stylianos E., Gardiner, Katheleen, De Camilli, Pietro, and Di Paolo, Gilbert
- Published
- 2008
- Full Text
- View/download PDF
22. A transgenic rat that develops Alzheimer's disease-like amyloid pathology, deficits in synaptic plasticity and cognitive impairment
- Author
-
Li Liu, Ian J. Orozco, Emmanuel Planel, Yi Wen, Alexis Bretteville, Pavan Krishnamurthy, Lili Wang, Mathieu Herman, Helen Figueroa, W. Haung Yu, Ottavio Arancio, and Karen Duff
- Subjects
Alzheimer's disease ,Transgenic rat ,β-Amyloid peptide ,Amyloid plaques ,Synaptic plasticity ,Long-term potentiation ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
In the last decade, multiple lines of transgenic APP overexpressing mice have been created that recapitulate certain aspects of Alzheimer's disease (AD). However, none of the previously reported transgenic APP overexpressing rat models developed AD-like β-amyloid (Aβ) deposits, or age-related learning and memory deficits. In the present study, we have characterized a transgenic rat model overexpressing transgenes with three, familial AD mutations (two in APP and one in PS1) that were developed by Flood et al. [Flood, D.G., et al., Aβ deposition in a transgenic rat model of Alzheimer's disease. Society for Neuroscience 2003, Washington, DC, 2003]. From the age of 9 months, these rats develop Aβ deposits in both diffuse and compact forms, with the latter being closely associated with activated microglia and reactive astrocytes. Impaired long-term potentiation (LTP) was revealed by electrophysiological recordings performed on hippocampal slices from rats at 7 months of age, which is 2 months before the appearance of amyloid plaques. The deficit in LTP was accompanied by impaired spatial learning and memory in the Morris water maze, which became more pronounced in transgenic rats of 13 months of age. For Tg rats of both ages, there was a trend for cognitive impairment to correlate with total Aβ42 levels in the hippocampus. The rat model therefore recapitulates AD-like amyloid pathology and cognitive impairment. The advantage of the rat model over the available mouse models is that rats provide better opportunities for advanced studies, such as serial CSF sampling, electrophysiology, neuroimaging, cell-based transplant manipulations, and complex behavioral testing.
- Published
- 2008
- Full Text
- View/download PDF
23. SUMO and Alzheimer’s Disease
- Author
-
Lee, Linda, Sakurai, Mikako, Matsuzaki, Shinsuke, Arancio, Ottavio, and Fraser, Paul
- Published
- 2013
- Full Text
- View/download PDF
24. Microglial large extracellular vesicles propagate early synaptic dysfunction in Alzheimer's disease.
- Author
-
Gabrielli, Martina, Prada, Ilaria, Joshi, Pooja, Falcicchia, Chiara, D'Arrigo, Giulia, Rutigliano, Grazia, Battocchio, Elisabetta, Zenatelli, Rossella, Tozzi, Francesca, Radeghieri, Annalisa, Arancio, Ottavio, Origlia, Nicola, and Verderio, Claudia
- Abstract
Synaptic dysfunction is an early mechanism in Alzheimer's disease that involves progressively larger areas of the brain over time. However, how it starts and propagates is unknown. Here we show that amyloid-β released by microglia in association with large extracellular vesicles (Aβ-EVs) alters dendritic spine morphology in vitro, at the site of neuron interaction, and impairs synaptic plasticity both in vitro and in vivo in the entorhinal cortex-dentate gyrus circuitry. One hour after Aβ-EV injection into the mouse entorhinal cortex, long-term potentiation was impaired in the entorhinal cortex but not in the dentate gyrus, its main target region, while 24 h later it was also impaired in the dentate gyrus, revealing a spreading of long-term potentiation deficit between the two regions. Similar results were obtained upon injection of extracellular vesicles carrying Aβ naturally secreted by CHO7PA2 cells, while neither Aβ42 alone nor inflammatory extracellular vesicles devoid of Aβ were able to propagate long-term potentiation impairment. Using optical tweezers combined to time-lapse imaging to study Aβ-EV-neuron interaction, we show that Aβ-EVs move anterogradely at the axon surface and that their motion can be blocked through annexin-V coating. Importantly, when Aβ-EV motility was inhibited, no propagation of long-term potentiation deficit occurred along the entorhinal-hippocampal circuit, implicating large extracellular vesicle motion at the neuron surface in the spreading of long-term potentiation impairment. Our data indicate the involvement of large microglial extracellular vesicles in the rise and propagation of early synaptic dysfunction in Alzheimer's disease and suggest a new mechanism controlling the diffusion of large extracellular vesicles and their pathogenic signals in the brain parenchyma, paving the way for novel therapeutic strategies to delay the disease. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Involvement of p38 MAPK in Synaptic Function and Dysfunction
- Author
-
Nicola Origlia, Ottavio Arancio, Chiara Falcicchia, Daniel Martin Watterson, and Francesca Tozzi
- Subjects
Gene isoform ,p38 mitogen-activated protein kinases ,Review ,Biology ,p38 Mitogen-Activated Protein Kinases ,Catalysis ,neuroinflammation ,lcsh:Chemistry ,Inorganic Chemistry ,Small Molecule Libraries ,Neuroplasticity ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Protein Kinase Inhibitors ,Spectroscopy ,Neuroinflammation ,Inflammation ,synaptic plasticity ,β-amyloid ,Organic Chemistry ,Neurodegeneration ,Brain ,Long-term potentiation ,General Medicine ,p38-MAPK α inhibitor ,medicine.disease ,Pathophysiology ,Computer Science Applications ,lcsh:Biology (General) ,lcsh:QD1-999 ,Synaptic plasticity ,Synapses ,Tau ,Neuroscience ,Alzheimer’s disease - Abstract
Many studies have revealed a central role of p38 MAPK in neuronal plasticity and the regulation of long-term changes in synaptic efficacy, such as long-term potentiation (LTP) and long-term depression (LTD). However, p38 MAPK is classically known as a responsive element to stress stimuli, including neuroinflammation. Specific to the pathophysiology of Alzheimer’s disease (AD), several studies have shown that the p38 MAPK cascade is activated either in response to the Aβ peptide or in the presence of tauopathies. Here, we describe the role of p38 MAPK in the regulation of synaptic plasticity and its implication in an animal model of neurodegeneration. In particular, recent evidence suggests the p38 MAPK α isoform as a potential neurotherapeutic target, and specific inhibitors have been developed and have proven to be effective in ameliorating synaptic and memory deficits in AD mouse models.
- Published
- 2020
26. Tau is not necessary for amyloid-β-induced synaptic and memory impairments
- Author
-
Elentina K. Argyrousi, Erica Acquarone, Mauro Fa, Domenica Donatella Li Puma, Ottavio Arancio, Claudio Grassi, Hong Zhang, Elisa Calcagno, Daniela Puzzo, Luciano D'Adamio, Nicholas M. Kanaan, Elisa Zuccarello, Paul E. Fraser, and Agnieszka Staniszewski
- Subjects
0301 basic medicine ,Amyloid β ,Settore BIO/09 - FISIOLOGIA ,Tau protein ,Long-Term Potentiation ,tau Proteins ,Basal synaptic transmission ,Synaptic Transmission ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Memory ,Alzheimer Disease ,mental disorders ,Animals ,Mice, Knockout ,Amyloid beta-Peptides ,biology ,Mechanism (biology) ,Behavioral methods ,General Medicine ,Alzheimer's disease ,3. Good health ,Biochemistry of Alzheimer's disease ,Electrophysiology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Synaptic plasticity ,Synapses ,biology.protein ,Alzheimer’s disease ,Neuroscience ,Research Article - Abstract
The amyloid hypothesis posits that the amyloid-beta (Aβ) protein precedes and requires microtubule-associated protein tau in a sort of trigger-bullet mechanism leading to Alzheimer's disease (AD) pathology. This sequence of events has become dogmatic in the AD field and is used to explain clinical trial failures due to a late start of the intervention when Aβ already activated tau. Here, using a multidisciplinary approach combining molecular biological, biochemical, histopathological, electrophysiological, and behavioral methods, we demonstrated that tau suppression did not protect against Aβ-induced damage of long-term synaptic plasticity and memory, or from amyloid deposition. Tau suppression could even unravel a defect in basal synaptic transmission in a mouse model of amyloid deposition. Similarly, tau suppression did not protect against exogenous oligomeric tau-induced impairment of long-term synaptic plasticity and memory. The protective effect of tau suppression was, in turn, confined to short-term plasticity and memory. Taken together, our data suggest that therapies downstream of Aβ and tau together are more suitable to combat AD than therapies against one or the other alone.
- Published
- 2020
27. A role for tau in learning, memory and synaptic plasticity
- Author
-
Fabrizio Biundo, Ottavio Arancio, Dolores Del Prete, Luciano D'Adamio, and Hong Zhang
- Subjects
0301 basic medicine ,Male ,Central nervous system ,Short-term memory ,lcsh:Medicine ,tau Proteins ,Biology ,Hippocampus ,Article ,Pathogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Alzheimer Disease ,Memory ,mental disorders ,medicine ,Dementia ,Animals ,Learning ,Learning memory ,Phosphorylation ,lcsh:Science ,Mice, Knockout ,Neurons ,Physiological function ,Memory Disorders ,Multidisciplinary ,Neuronal Plasticity ,lcsh:R ,Brain ,Neurofibrillary Tangles ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Synaptic plasticity ,lcsh:Q ,Alzheimer's disease ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Tau plays a pivotal role in the pathogenesis of neurodegenerative disorders: mutations in the gene encoding for tau (MAPT) are linked to Fronto-temporal Dementia (FTD) and hyper-phosphorylated aggregates of tau forming neurofibrillary tangles (NFTs) that constitute a pathological hallmark of Alzheimer disease (AD) and FTD. Accordingly, tau is a favored therapeutic target for the treatment of these diseases. Given the criticality of tau to dementia’s pathogenesis and therapy, it is important to understand the physiological function of tau in the central nervous system. Analysis of Mapt knock out (Mapt−/−) mice has yielded inconsistent results. Some studies have shown that tau deletion does not alter memory while others have described synaptic plasticity and memory alterations in Mapt−/− mice. To help clarifying these contrasting results, we analyzed a distinct Mapt−/− model on a B6129PF3/J genetic background. We found that tau deletion leads to aging-dependent short-term memory deficits, hyperactivity and synaptic plasticity defects. In contrast, Mapt+/− mice only showed a mild short memory deficit in the novel object recognition task. Thus, while tau is important for normal neuronal functions underlying learning and memory, partial reduction of tau expression may have fractional deleterious effects.
- Published
- 2018
- Full Text
- View/download PDF
28. Cell cultures from animal models of Alzheimer’s disease as a tool for faster screening and testing of drug efficacy
- Author
-
Trinchese, Fabrizio, Liu, Shumin, Ninan, Ipe, Puzzo, Daniela, Jacob, Joel P., and Arancio, Ottavio
- Published
- 2004
- Full Text
- View/download PDF
29. Calpain inhibitors, a treatment for alzheimer’s disease: Position paper
- Author
-
Battaglia, Fortunato, Trinchese, Fabrizio, Liu, Shumin, Walter, Sean, Nixon, Ralph A., and Arancio, Ottavio
- Published
- 2003
- Full Text
- View/download PDF
30. Usefulness of Behavioral and Electrophysiological Studies in Transgenic Models of Alzheimer's Disease
- Author
-
Sant'Angelo, Antonino, Trinchese, Fabrizio, and Arancio, Ottavio
- Published
- 2003
- Full Text
- View/download PDF
31. Calpain inhibitors: A treatment for alzheimer’s disease
- Author
-
Di Rosa, Gabriella, Odrlijn, Tatjana, Nixon, Ralph A., and Arancio, Ottavio
- Published
- 2002
- Full Text
- View/download PDF
32. Genetic deletion of α7 nicotinic acetylcholine receptors induces an age-dependent Alzheimer’s disease-like pathology
- Author
-
Marcello Melone, Domenica Donatella Li Puma, Fiorenzo Conti, Ottavio Arancio, Maria Rosaria Tropea, Daniela Puzzo, Claudio Grassi, and Walter Gulisano
- Subjects
Pathology ,medicine.medical_specialty ,Settore BIO/09 - FISIOLOGIA ,Tau protein ,Hyperphosphorylation ,Alpha7 nicotinic acetylcholine receptor ,Receptors, Nicotinic ,Hippocampus ,Synaptic plasticity ,Synapse ,Mice ,Alzheimer Disease ,Memory ,Amyloid precursor protein ,medicine ,Animals ,Acetylcholine receptor ,Amyloid beta-Peptides ,Glycogen Synthase Kinase 3 beta ,biology ,General Neuroscience ,Alzheimer's disease ,Peptide Fragments ,Nicotinic agonist ,biology.protein ,Cholinergic ,Amyloid-beta peptide ,Neuroscience - Abstract
The accumulation of amyloid-beta peptide (Aβ) and the failure of cholinergic transmission are key players in Alzheimer's disease (AD). However, in the healthy brain, Aβ contributes to synaptic plasticity and memory acting through α7 subtype nicotinic acetylcholine receptors (α7nAChRs). Here, we hypothesized that the α7nAChR deletion blocks Aβ physiological function and promotes a compensatory increase in Aβ levels that, in turn, triggers an AD-like pathology. To validate this hypothesis, we studied the age-dependent phenotype of α7 knock out mice. We found that α7nAChR deletion caused an impairment of hippocampal synaptic plasticity and memory at 12 months of age, paralleled by an increase of Amyloid Precursor Protein expression and Aβ levels. This was accompanied by other classical AD features such as a hyperphosphorylation of tau at residues Ser 199, Ser 396, Thr 205, a decrease of GSK-3β at Ser 9, the presence of paired helical filaments and neurofibrillary tangles, neuronal loss and an increase of GFAP-positive astrocytes. Our findings suggest that α7nAChR malfunction might precede Aβ and tau pathology, offering a different perspective to interpret the failure of anti-Aβ therapies against AD and to find novel therapeutical approaches aimed at restoring α7nAChRs-mediated Aβ function at the synapse.
- Published
- 2021
- Full Text
- View/download PDF
33. Post-translational remodeling of ryanodine receptor induces calcium leak leading to Alzheimer’s disease-like pathologies and cognitive deficits
- Author
-
Alain Lacampagne, Andrew R. Marks, Steven Reiken, Clark A. Briggs, Xiaoping Liu, Ottavio Arancio, Andrew F. Teich, Shreaya Chakroborty, Charlotte Bauer, Michael L. Shelanski, Nathalie Saint, Inger Lauritzen, Fabrice Duprat, Grace E. Stutzmann, Mounia Chami, Frédéric Checler, Ran Zalk, Renaud Bussiere, Albano C. Meli, Physiologie & médecine expérimentale du Cœur et des Muscles [U 1046] (PhyMedExp), Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Columbia University [New York], Department of Physiology & Cellular Biophysics, Institut de pharmacologie moléculaire et cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Chicago Medical School [Rosalind Franklin University], Rosalind Franklin University, Department of Neuroscience Rosalind Franklin University, Rosalind Franklin University-Chicago Medical Scchool, Università degli Studi di Ferrara (UniFE), and Columbia University College of Physicians and Surgeons
- Subjects
Male ,0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Ryanodine receptor 2 ,Mice ,0302 clinical medicine ,Phosphorylation ,ComputingMilieux_MISCELLANEOUS ,biology ,Ryanodine receptor ,Nitrosylation ,PKA-dependent phosphorylation ,musculoskeletal system ,Sarcoplasmic Reticulum ,cardiovascular system ,Female ,Alzheimer's disease ,Signal transduction ,tissues ,medicine.medical_specialty ,Amyloid beta ,Mice, Transgenic ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,Internal medicine ,medicine ,Animals ,Humans ,Calcium Signaling ,Maze Learning ,Endoplasmic reticulum ,Recognition, Psychology ,Ryanodine Receptor Calcium Release Channel ,medicine.disease ,Cyclic AMP-Dependent Protein Kinases ,030104 developmental biology ,Endocrinology ,Oxidative stress ,Synaptic plasticity ,biology.protein ,Calcium ,Neurology (clinical) ,Cognition Disorders ,Protein Processing, Post-Translational ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The mechanisms underlying ryanodine receptor (RyR) dysfunction associated with Alzheimer disease (AD) are still not well understood. Here, we show that neuronal RyR2 channels undergo post-translational remodeling (PKA phosphorylation, oxidation, and nitrosylation) in brains of AD patients, and in two murine models of AD (3 × Tg-AD, APP +/− /PS1 +/−). RyR2 is depleted of calstabin2 (KFBP12.6) in the channel complex, resulting in endoplasmic reticular (ER) calcium (Ca2+) leak. RyR-mediated ER Ca2+ leak activates Ca2+-dependent signaling pathways, contributing to AD pathogenesis. Pharmacological (using a novel RyR stabilizing drug Rycal) or genetic rescue of the RyR2-mediated intracellular Ca2+ leak improved synaptic plasticity, normalized behavioral and cognitive functions and reduced Aβ load. Genetically altered mice with congenitally leaky RyR2 exhibited premature and severe defects in synaptic plasticity, behavior and cognitive function. These data provide a mechanism underlying leaky RyR2 channels, which could be considered as potential AD therapeutic targets.
- Published
- 2017
- Full Text
- View/download PDF
34. Synaptic and memory dysfunction induced by tau oligomers is rescued by up-regulation of the nitric oxide cascade
- Author
-
Agnieszka Staniszewski, Erica Acquarone, Elisa Zuccarello, Jole Fiorito, Luciano D'Adamio, Shi Xian Deng, Elentina K. Argyrousi, Walter Gulisano, Mauro Fa, Manon Van Den Berg, Elisa Calcagno, Ottavio Arancio, Daniela Puzzo, RS: MHeNs - R3 - Neuroscience, Promovendi MHN, Psychiatrie & Neuropsychologie, UM Sports, RS: GROW - R1 - Prevention, RS: FPN NPPP II, and Section Psychopharmacology
- Subjects
Male ,0301 basic medicine ,Tau oligomers ,lcsh:Geriatrics ,lcsh:RC346-429 ,0302 clinical medicine ,Soluble guanylyl cyclase ,DEPENDENT PROTEIN-KINASE ,Neurons ,Neuronal Plasticity ,Arc (protein) ,biology ,Chemistry ,CREB ,PAIRED HELICAL FILAMENTS ,Long-term potentiation ,MOUSE MODEL ,Alzheimer's disease ,Cell biology ,ALZHEIMERS-DISEASE ,Female ,LONG-TERM POTENTIATION ,Alzheimer’s disease ,Research Article ,Memory Dysfunction ,SELECTIVE PDE5 INHIBITOR ,Memory ,Nitric oxide ,PDE5 ,Protein kinase G ,tau Proteins ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,OBJECT MEMORY ,Alzheimer Disease ,Animals ,Memory impairment ,Molecular Biology ,lcsh:Neurology. Diseases of the nervous system ,Memory Disorders ,PHOSPHODIESTERASE 5 INHIBITOR ,Amyloid beta-Peptides ,ADENYLYL-CYCLASE ,ELEMENT-BINDING PROTEIN ,Mice, Inbred C57BL ,lcsh:RC952-954.6 ,030104 developmental biology ,Synaptic plasticity ,biology.protein ,Neurology (clinical) ,cGMP-dependent protein kinase ,030217 neurology & neurosurgery - Abstract
Background Soluble aggregates of oligomeric forms of tau protein (oTau) have been associated with impairment of synaptic plasticity and memory in Alzheimer’s disease. However, the molecular mechanisms underlying the synaptic and memory dysfunction induced by elevation of oTau are still unknown. Methods This work used a combination of biochemical, electrophysiological and behavioral techniques. Biochemical methods included analysis of phosphorylation of the cAMP-responsive element binding (CREB) protein, a transcriptional factor involved in memory, histone acetylation, and expression immediate early genes c-Fos and Arc. Electrophysiological methods included assessment of long-term potentiation (LTP), a type of synaptic plasticity thought to underlie memory formation. Behavioral studies investigated both short-term spatial memory and associative memory. These phenomena were examined following oTau elevation. Results Levels of phospho-CREB, histone 3 acetylation at lysine 27, and immediate early genes c-Fos and Arc, were found to be reduced after oTau elevation during memory formation. These findings led us to explore whether up-regulation of various components of the nitric oxide (NO) signaling pathway impinging onto CREB is capable of rescuing oTau-induced impairment of plasticity, memory, and CREB phosphorylation. The increase of NO levels protected against oTau-induced impairment of LTP through activation of soluble guanylyl cyclase. Similarly, the elevation of cGMP levels and stimulation of the cGMP-dependent protein kinases (PKG) re-established normal LTP after exposure to oTau. Pharmacological inhibition of cGMP degradation through inhibition of phosphodiesterase 5 (PDE5), rescued oTau-induced LTP reduction. These findings could be extrapolated to memory because PKG activation and PDE5 inhibition rescued oTau-induced memory impairment. Finally, PDE5 inhibition re-established normal elevation of CREB phosphorylation and cGMP levels after memory induction in the presence of oTau. Conclusions Up-regulation of CREB activation through agents acting on the NO cascade might be beneficial against tau-induced synaptic and memory dysfunctions. Electronic supplementary material The online version of this article (10.1186/s13024-019-0326-4) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
35. The penalty of stress ‐ Epichaperomes negatively reshaping the brain in neurodegenerative disorders.
- Author
-
Ginsberg, Stephen D., Joshi, Suhasini, Sharma, Sahil, Guzman, Gianny, Wang, Tai, Arancio, Ottavio, and Chiosis, Gabriela
- Subjects
NEURODEGENERATION ,CENTRAL nervous system ,PSYCHOLOGICAL stress ,ALZHEIMER'S disease ,LARGE-scale brain networks - Abstract
Adaptation to acute and chronic stress and/or persistent stressors is a subject of wide interest in central nervous system disorders. In this context, stress is an effector of change in organismal homeostasis and the response is generated when the brain perceives a potential threat. Herein, we discuss a nuanced and granular view whereby a wide variety of genotoxic and environmental stressors, including aging, genetic risk factors, environmental exposures, and age‐ and lifestyle‐related changes, act as direct insults to cellular, as opposed to organismal, homeostasis. These two concepts of how stressors impact the central nervous system are not mutually exclusive. We discuss how maladaptive stressor‐induced changes in protein connectivity through epichaperomes, disease‐associated pathologic scaffolds composed of tightly bound chaperones, co‐chaperones, and other factors, impact intracellular protein functionality altering phenotypes, that in turn disrupt and remodel brain networks ranging from intercellular to brain connectome levels. We provide an evidence‐based view on how these maladaptive changes ranging from stressor to phenotype provide unique precision medicine opportunities for diagnostic and therapeutic development, especially in the context of neurodegenerative disorders including Alzheimer's disease where treatment options are currently limited. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
36. Beta‐amyloid 1‐42 monomers, but not oligomers, produce <scp>PHF</scp> ‐like conformation of Tau protein
- Author
-
Elena Tamagno, Massimo Tabaton, Ottavio Arancio, Patrizio Odetti, Laura Colombo, Roberta Borghi, Raluca Zamfir, Michela Guglielmotto, Giusi Manassero, Mario Salmona, and George Perry
- Subjects
0301 basic medicine ,PHF ,Aging ,Amyloid ,Protein Conformation ,Tau protein ,Hyperphosphorylation ,tau Proteins ,Protein aggregation ,tau protein ,Protein Aggregates ,03 medical and health sciences ,0302 clinical medicine ,Protein structure ,Alzheimer Disease ,mental disorders ,medicine ,Animals ,Humans ,Alzheimer's disease ,beta-amyloid ,hTau mice ,MAPK ,Cell Biology ,Phosphorylation ,beta‐amyloid ,Mice, Knockout ,Amyloid beta-Peptides ,biology ,Kinase ,Beta amyloid ,Original Articles ,medicine.disease ,Peptide Fragments ,Enzyme Activation ,Alternative Splicing ,030104 developmental biology ,Solubility ,Biochemistry ,Disease Progression ,biology.protein ,Biophysics ,Original Article ,Alzheimer Disease, Tau Protein, Beta amyloid ,Protein Multimerization ,030217 neurology & neurosurgery - Abstract
Summary The mechanistic relationship between amyloid β1‐42 (Aβ1‐42) and the alteration of Tau protein are debated. We investigated the effect of Aβ1‐42 monomers and oligomers on Tau, using mice expressing wild‐type human Tau that do not spontaneously develop Tau pathology. After intraventricular injection of Aβ1‐42, mice were sacrificed after 3 h or 4 days. The short‐lasting treatment with Aβ monomers, but not oligomers, showed a conformational PHF‐like change of Tau, together with hyperphosphorylation. The same treatment induced increase in concentration of GSK3 and MAP kinases. The inhibition of the kinases rescued the Tau changes. Aβ monomers increased the levels of total Tau, through the inhibition of proteasomal degradation. Aβ oligomers reproduced all the aforementioned alterations only after 4 days of treatment. It is known that Aβ1‐42 monomers foster synaptic activity. Our results suggest that Aβ monomers physiologically favor Tau activity and dendritic sprouting, whereas their excess causes Tau pathology. Moreover, our study indicates that anti‐Aβ therapies should be targeted to Aβ1‐42 monomers too.
- Published
- 2016
- Full Text
- View/download PDF
37. Sub-efficacious doses of phosphodiesterase 4 and 5 inhibitors improve memory in a mouse model of Alzheimer's disease
- Author
-
Maria Rosaria Tropea, Walter Gulisano, Ottavio Arancio, Daniela Puzzo, and Agostino Palmeri
- Subjects
0301 basic medicine ,Cyclopropanes ,Male ,Phosphodiesterase Inhibitors ,Aminopyridines ,Disease ,Pharmacology ,Transgenic ,Mice ,Amyloid beta-Protein Precursor ,Random Allocation ,0302 clinical medicine ,Transgenic models ,Nootropic Agents ,Phosphodiesterase ,Cognition ,Alzheimer's disease ,Type 5 ,Benzamides ,Female ,Type 4 ,medicine.drug ,Cyclic Nucleotide Phosphodiesterases ,Cyclic nucleotides ,Mice, Transgenic ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Vardenafil Dihydrochloride ,Alzheimer Disease ,Memory ,medicine ,Animals ,Humans ,Roflumilast ,Cyclic Nucleotide Phosphodiesterases, Type 5 ,business.industry ,Animal ,medicine.disease ,Cyclic Nucleotide Phosphodiesterases, Type 4 ,Disease Models, Animal ,030104 developmental biology ,Vardenafil ,Synaptic plasticity ,Disease Models ,business ,Phosphodiesterase inhibitors ,030217 neurology & neurosurgery - Abstract
Cyclic nucleotides cAMP and cGMP cooperate to ensure memory acquisition and consolidation. Increasing their levels by phosphodiesterase inhibitors (PDE-Is) enhanced cognitive functions and rescued memory loss in different models of aging and Alzheimer's disease (AD). However, side effects due to the high doses used limited their application in humans. Based on previous studies suggesting that combinations of sub-efficacious doses of cAMP- and cGMP-specific PDE-Is improved synaptic plasticity and memory in physiological conditions, here we aimed to study whether this treatment was effective to counteract the AD phenotype in APPswe mice. We found that a 3-week chronic treatment with a combination of sub-efficacious doses of the cAMP-specific PDE4-I roflumilast (0.01 mg/kg) and the cGMP-specific PDE5-I vardenafil (0.1 mg/kg) improved recognition, spatial and contextual fear memory. Importantly, the cognitive enhancement persisted for 2 months beyond administration. This long-lasting action, and the possibility to minimize side effects due to the low doses used, might open feasible therapeutic strategies against AD.
- Published
- 2018
38. Increased neuronal PreP activity reduces Aβ accumulation, attenuates neuroinflammation and improves mitochondrial and synaptic function in Alzheimer disease's mouse model
- Author
-
Heng Du, Du Fang, Molly Rabinowitz, Shiqiang Yan, John Xi Chen, Ottavio Arancio, Jhansi Rani Vangavaragu, Yongfu Wang, Zhihua Zhang, Shirley ShiDu Yan, Guy M. McKhann, Shijun Yan, Gang Hu, Elzbieta Glaser, Alexander A. Sosunov, Long Wu, Lan Guo, Qinru Sun, and Changjia Zhong
- Subjects
Transgene ,Gene Expression ,Mice, Transgenic ,Mitochondrion ,Biology ,medicine.disease_cause ,Protein Aggregation, Pathological ,Proinflammatory cytokine ,Mice ,Cognition ,Alzheimer Disease ,In vivo ,Genetics ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Genetics (clinical) ,Neuroinflammation ,Neurons ,Amyloid beta-Peptides ,Behavior, Animal ,Serine Endopeptidases ,Long-term potentiation ,Articles ,General Medicine ,medicine.disease ,Mitochondria ,Cell biology ,Disease Models, Animal ,Oxidative Stress ,Proteolysis ,Synapses ,Inflammation Mediators ,Alzheimer's disease ,Oxidative stress - Abstract
Accumulation of amyloid-β (Aβ) in synaptic mitochondria is associated with mitochondrial and synaptic injury. The underlying mechanisms and strategies to eliminate Aβ and rescue mitochondrial and synaptic defects remain elusive. Presequence protease (PreP), a mitochondrial peptidasome, is a novel mitochondrial Aβ degrading enzyme. Here, we demonstrate for the first time that increased expression of active human PreP in cortical neurons attenuates Alzheimer disease's (AD)-like mitochondrial amyloid pathology and synaptic mitochondrial dysfunction, and suppresses mitochondrial oxidative stress. Notably, PreP-overexpressed AD mice show significant reduction in the production of proinflammatory mediators. Accordingly, increased neuronal PreP expression improves learning and memory and synaptic function in vivo AD mice, and alleviates Aβ-mediated reduction of long-term potentiation (LTP). Our results provide in vivo evidence that PreP may play an important role in maintaining mitochondrial integrity and function by clearance and degradation of mitochondrial Aβ along with the improvement in synaptic and behavioral function in AD mouse model. Thus, enhancing PreP activity/expression may be a new therapeutic avenue for treatment of AD.
- Published
- 2015
- Full Text
- View/download PDF
39. Targeting Human Central Nervous System Protein Kinases: An Isoform Selective p38αMAPK Inhibitor That Attenuates Disease Progression in Alzheimer’s Disease Mouse Models
- Author
-
Jeffrey C. Pelletier, Faisal Saeed, Valerie Grum-Tokars, Agnieszka Staniszewski, Wayne F. Anderson, George Minasov, Scott J. Webster, D. Martin Watterson, Saktimayee M. Roy, James P. Schavocky, Andrew F. Teich, Adam D. Bachstetter, Ottavio Arancio, and Linda J. Van Eldik
- Subjects
Male ,Gene isoform ,Physiology ,medicine.drug_class ,Cognitive Neuroscience ,Drug Evaluation, Preclinical ,Mice, Transgenic ,Signal transduction ,Biology ,Pharmacology ,Biochemistry ,Tyrosine-kinase inhibitor ,Cell Line ,Mitogen-Activated Protein Kinase 14 ,Rats, Sprague-Dawley ,cognitive dysfunction ,Alzheimer Disease ,medicine ,Animals ,Humans ,crystallography ,Protein kinase A ,Protein Kinase Inhibitors ,Neuropharmacology ,Spatial Memory ,Memory Disorders ,Dose-Response Relationship, Drug ,Molecular Structure ,Kinase ,Association Learning ,Brain ,protein kinase ,Cell Biology ,General Medicine ,Protein kinase inhibitor ,medicine.disease ,3. Good health ,Pyridazines ,Disease Models, Animal ,Neuroprotective Agents ,Synapses ,Disease Progression ,Microsomes, Liver ,pharmacology ,Alzheimer's disease ,chemical synthesis ,Research Article - Abstract
The first kinase inhibitor drug approval in 2001 initiated a remarkable decade of tyrosine kinase inhibitor drugs for oncology indications, but a void exists for serine/threonine protein kinase inhibitor drugs and central nervous system indications. Stress kinases are of special interest in neurological and neuropsychiatric disorders due to their involvement in synaptic dysfunction and complex disease susceptibility. Clinical and preclinical evidence implicates the stress related kinase p38αMAPK as a potential neurotherapeutic target, but isoform selective p38αMAPK inhibitor candidates are lacking and the mixed kinase inhibitor drugs that are promising in peripheral tissue disease indications have limitations for neurologic indications. Therefore, pursuit of the neurotherapeutic hypothesis requires kinase isoform selective inhibitors with appropriate neuropharmacology features. Synaptic dysfunction disorders offer a potential for enhanced pharmacological efficacy due to stress-induced activation of p38αMAPK in both neurons and glia, the interacting cellular components of the synaptic pathophysiological axis, to be modulated. We report a novel isoform selective p38αMAPK inhibitor, MW01-18-150SRM (=MW150), that is efficacious in suppression of hippocampal-dependent associative and spatial memory deficits in two distinct synaptic dysfunction mouse models. A synthetic scheme for biocompatible product and positive outcomes from pharmacological screens are presented. The high-resolution crystallographic structure of the p38αMAPK/MW150 complex documents active site binding, reveals a potential low energy conformation of the bound inhibitor, and suggests a structural explanation for MW150's exquisite target selectivity. As far as we are aware, MW150 is without precedent as an isoform selective p38MAPK inhibitor or as a kinase inhibitor capable of modulating in vivo stress related behavior.
- Published
- 2015
- Full Text
- View/download PDF
40. Synaptic Therapy in Alzheimer’s Disease: A CREB-centric Approach
- Author
-
Mauro Fa, Andrew F. Teich, Jole Fiorito, Rosa Purgatorio, Ottavio Arancio, Russell E. Nicholls, and Daniela Puzzo
- Subjects
Review ,Neurotransmission ,CREB ,Synaptic Transmission ,memory ,chemistry.chemical_compound ,Alzheimer Disease ,Pathology ,medicine ,Cyclic AMP Response Element-Binding Protein ,Animals ,Humans ,Pharmacology (medical) ,Cyclic adenosine monophosphate ,Pharmacology ,Histone Acetyltransferases ,biology ,Kinase ,FOS: Clinical medicine ,Neurosciences ,Alzheimer's disease ,medicine.disease ,Histone ,chemistry ,Synapses ,biology.protein ,Medicine ,Mental health ,Neurology (clinical) ,Neuroscience - Abstract
Therapeutic attempts to cure Alzheimer's disease (AD) have failed, and new strategies are desperately needed. Motivated by this reality, many laboratories (including our own) have focused on synaptic dysfunction in AD because synaptic changes are highly correlated with the severity of clinical dementia. In particular, memory formation is accompanied by altered synaptic strength, and this phenomenon (and its dysfunction in AD) has been a recent focus for many laboratories. The molecule cyclic adenosine monophosphate response element-binding protein (CREB) is at a central converging point of pathways and mechanisms activated during the processes of synaptic strengthening and memory formation, as CREB phosphorylation leads to transcription of memory-associated genes. Disruption of these mechanisms in AD results in a reduction of CREB activation with accompanying memory impairment. Thus, it is likely that strategies aimed at these mechanisms will lead to future therapies for AD. In this review, we will summarize literature that investigates 5 possible therapeutic pathways for rescuing synaptic dysfunction in AD: 4 enzymatic pathways that lead to CREB phosphorylation (the cyclic adenosine monophosphate cascade, the serine/threonine kinases extracellular regulated kinases 1 and 2, the nitric oxide cascade, and the calpains), as well as histone acetyltransferases and histone deacetylases (2 enzymes that regulate the histone acetylation necessary for gene transcription).
- Published
- 2015
- Full Text
- View/download PDF
41. Leucine Carboxyl Methyltransferase 1 Overexpression Protects Against Cognitive and Electrophysiological Impairments in Tg2576 APP Transgenic Mice.
- Author
-
Gnanaprakash, Madhumathi, Staniszewski, Agnieszka, Zhang, Hong, Pitstick, Rose, Kavanaugh, Michael P, Arancio, Ottavio, and Nicholls, Russell E
- Abstract
Background: The serine/threonine protein phosphatase, PP2A, is thought to play a central role in the molecular pathogenesis of Alzheimer's disease (AD), and the activity and substrate specificity of PP2A is regulated, in part, through methylation and demethylation of its catalytic subunit. Previously, we found that transgenic overexpression of the PP2A methyltransferase, LCMT-1, or the PP2A methylesterase, PME-1, altered the sensitivity of mice to impairments caused by acute exposure to synthetic oligomeric amyloid-β (Aβ).Objective: Here we sought to test the possibility that these molecules also controlled sensitivity to impairments caused by chronically elevated levels of Aβ produced in vivo.Methods: To do this, we examined the effects of transgenic LCMT-1, or PME-1 overexpression on cognitive and electrophysiological impairments caused by chronic overexpression of mutant human APP in Tg2576 mice.Results: We found that LCMT-1 overexpression prevented impairments in short-term spatial memory and synaptic plasticity in Tg2576 mice, without altering APP expression or soluble Aβ levels. While the magnitude of the effects of PME-1 overexpression in Tg2576 mice was small and potentially confounded by the emergence of non-cognitive impairments, Tg2576 mice that overexpressed PME-1 showed a trend toward earlier onset and/or increased severity of cognitive and electrophysiological impairments.Conclusion: These data suggest that the PP2A methyltransferase, LCMT-1, and the PP2A methylesterase, PME-1, may participate in the molecular pathogenesis of AD by regulating sensitivity to the pathogenic effects of chronically elevated levels of Aβ. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
42. Abolishing Tau cleavage by caspases at Aspartate421 causes memory/synaptic plasticity deficits and pre-pathological Tau alterations
- Author
-
Francesca Vitale, Cristina d'Abramo, Luciano D'Adamio, Dolores Del Prete, Ottavio Arancio, H. Zhang, Luca Giliberto, Fabrizio Biundo, and M D Tambini
- Subjects
0301 basic medicine ,biology ,Chemistry ,Transgene ,Neurodegeneration ,medicine.disease ,Pathogenesis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,030104 developmental biology ,0302 clinical medicine ,Synaptic plasticity ,Forebrain ,Neuroplasticity ,mental disorders ,biology.protein ,medicine ,Original Article ,Alzheimer's disease ,Neuroscience ,030217 neurology & neurosurgery ,Biological Psychiatry ,Caspase - Abstract
TAU mutations are genetically linked to fronto-temporal dementia (FTD) and hyper-phosphorylated aggregates of Tau form neurofibrillary tangles (NFTs) that constitute a pathological hallmark of Alzheimer disease (AD) and FTD. These observations indicate that Tau has a pivotal role in the pathogenesis of neurodegenerative disorders. Tau is cleaved by caspases at Aspartate421, to form a Tau metabolite known as δTau; δTau is increased in AD, due to the hyper-activation of caspases in AD brains. δTau is considered a critical toxic moiety underlying neurodegeneration, which initiates and facilitates NFT formation. As Tau is a therapeutic target in neurodegeneration, it is important to rigorously determine whether δTau is a toxic Tau species that should be pharmacologically attacked. To directly address these questions, we have generated a knock-in (KI) mouse called TauDN—that expresses a Tau mutant that cannot be cleaved by caspases. TauDN mice present short-term memory deficits and synaptic plasticity defects. Moreover, mice carrying two mutant Tau alleles show increased total insoluble hyper-phosphorylated Tau in the forebrain. These data are in contrast with the concept that δTau is a critical toxic moiety underlying neurodegeneration, and suggest that cleavage of Tau by caspases represents a negative feedback mechanism aimed to eliminate toxic Tau species. Alternatively, it is possible that either a reduction or an increase in δTau leads to synaptic dysfunction, memory impairments and Tau pathology. Both possibilities will have to be considered when targeting caspase cleavage of Tau in AD therapy.
- Published
- 2017
43. Dual Mechanism of Toxicity for Extracellular Injection of Tau Oligomers versus Monomers in Human Tau Mice
- Author
-
Olena Butenko, Giusi Manassero, Valeria Vasciaveo, Ottavio Arancio, Massimo Tabaton, Elena Tamagno, Michela Guglielmotto, and Debora Monteleone
- Subjects
0301 basic medicine ,Peptide ,law.invention ,Mice ,0302 clinical medicine ,law ,Phosphorylation ,bcl-2-Associated X Protein ,chemistry.chemical_classification ,biology ,Chemistry ,Kinase ,Calpain ,General Neuroscience ,Neurodegeneration ,General Medicine ,Alzheimer's disease ,Recombinant Proteins ,Clinical Psychology ,Biochemistry ,hTau mice ,paired helical filaments ,tau protein ,Geriatrics and Gerontology ,Psychiatry and Mental Health ,Proto-Oncogene Proteins c-bcl-2 ,Toxicity ,Recombinant DNA ,Neurotoxicity Syndromes ,Signal Transduction ,Tau protein ,Green Fluorescent Proteins ,Mice, Transgenic ,tau Proteins ,Cleavage (embryo) ,Article ,03 medical and health sciences ,mental disorders ,medicine ,Extracellular ,Animals ,Humans ,Injections, Intraventricular ,Extracellular Fluid ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,Biophysics ,030217 neurology & neurosurgery - Abstract
The mechanism of tau toxicity is still unclear. Here we report that recombinant tau oligomers and monomers, intraventricularly injected in mice with a pure human tau background, foster tau pathology through different mechanisms. Oligomeric forms of tau alter the conformation of tau in a paired helical filament-like manner. This effect occurs without tau hyperphosphorylation as well as activation of specific kinases, suggesting that oligomers of tau induce tau assembly through a nucleation effect. Monomers, in turn, induce neurodegeneration through a calpain-mediated tau cleavage that leads to accumulation of a 17 kDa neurotoxic peptide and induction of apoptotic cell death.
- Published
- 2017
44. LTP and memory impairment caused by extracellular Aβ and Tau oligomers is APP-dependent
- Author
-
Agostino Palmeri, Walter Gulisano, Mauro Fa, Agnes Staniszewski, Sara Cocco, Daniela Puzzo, Luciano D'Adamio, Domenica Donatella Li Puma, Roberto Piacentini, Paul E. Fraser, Ottavio Arancio, Hong Zhang, Maria Rosaria Tropea, and Claudio Grassi
- Subjects
0301 basic medicine ,Amyloid beta ,QH301-705.5 ,Settore BIO/09 - FISIOLOGIA ,Science ,Plasma protein binding ,urologic and male genital diseases ,General Biochemistry, Genetics and Molecular Biology ,neuroscience ,memory ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,Extracellular ,medicine ,Amyloid precursor protein ,Memory impairment ,tau ,Biology (General) ,mouse ,synaptic plasticity ,General Immunology and Microbiology ,biology ,Chemistry ,General Neuroscience ,Long-term potentiation ,General Medicine ,Alzheimer's disease ,medicine.disease ,female genital diseases and pregnancy complications ,humanities ,amyloid-beta ,3. Good health ,030104 developmental biology ,Synaptic plasticity ,biology.protein ,Medicine ,APP ,Neuroscience ,030217 neurology & neurosurgery ,Research Article - Abstract
The concurrent application of subtoxic doses of soluble oligomeric forms of human amyloid-beta (oAβ) and Tau (oTau) proteins impairs memory and its electrophysiological surrogate long-term potentiation (LTP), effects that may be mediated by intra-neuronal oligomers uptake. Intrigued by these findings, we investigated whether oAβ and oTau share a common mechanism when they impair memory and LTP in mice. We found that as already shown for oAβ, also oTau can bind to amyloid precursor protein (APP). Moreover, efficient intra-neuronal uptake of oAβ and oTau requires expression of APP. Finally, the toxic effect of both extracellular oAβ and oTau on memory and LTP is dependent upon APP since APP-KO mice were resistant to oAβ- and oTau-induced defects in spatial/associative memory and LTP. Thus, APP might serve as a common therapeutic target against Alzheimer's Disease (AD) and a host of other neurodegenerative diseases characterized by abnormal levels of Aβ and/or Tau. DOI: http://dx.doi.org/10.7554/eLife.26991.001
- Published
- 2017
45. Memory-enhancing effects of GEBR-32a, a new PDE4D inhibitor holding promise for the treatment of Alzheimer’s disease
- Author
-
Carla Villa, Jos Prickaerts, Chiara Brullo, Claudia Rebosio, Matilde Balbi, Roberta Ricciarelli, Elisa Calcagno, Britt T. J. van Hagen, Olga Bruno, Ottavio Arancio, Elentina K. Argyrousi, Ernesto Fedele, Hong Zhang, Maria Adelaide Pronzato, RS: MHeNs - R3 - Neuroscience, Psychiatrie & Neuropsychologie, and Promovendi MHN
- Subjects
PHOSPHODIESTERASE 4D ,0301 basic medicine ,IMPROVES MEMORY ,Central nervous system ,Intracellular Space ,Hippocampus ,Mice, Transgenic ,Hippocampal formation ,Article ,LATE-PHASE ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,COGNITION ENHANCEMENT ,Alzheimer Disease ,Memory ,In vivo ,Cyclic AMP ,medicine ,Animals ,Humans ,Cyclic adenosine monophosphate ,PROTEIN-KINASE-A ,Cells, Cultured ,SPATIAL MEMORY ,Multidisciplinary ,Molecular Structure ,business.industry ,ADENYLYL-CYCLASE ,Phosphodiesterase ,Long-term potentiation ,HIPPOCAMPAL SYNAPTIC PLASTICITY ,medicine.disease ,Recombinant Proteins ,Cyclic Nucleotide Phosphodiesterases, Type 4 ,Isoenzymes ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,RECOGNITION MEMORY ,Phosphodiesterase 4 Inhibitors ,LONG-TERM POTENTIATION ,Alzheimer's disease ,business ,Neuroscience ,030217 neurology & neurosurgery ,DNA Damage - Abstract
Memory loss characterizes several neurodegenerative disorders, including Alzheimer’s disease (AD). Inhibition of type 4 phosphodiesterase (PDE4) and elevation of cyclic adenosine monophosphate (cAMP) has emerged as a promising therapeutic approach to treat cognitive deficits. However, PDE4 exists in several isoforms and pan inhibitors cannot be used in humans due to severe emesis. Here, we present GEBR-32a, a new PDE4D full inhibitor that has been characterized both in vitro and in vivo using biochemical, electrophysiological and behavioural analyses. GEBR-32a efficiently enhances cAMP in neuronal cultures and hippocampal slices. In vivo pharmacokinetic analysis shows that GEBR-32a is rapidly distributed within the central nervous system with a very favourable brain/blood ratio. Specific behavioural tests (object location and Y-maze continuous alternation tasks) demonstrate that this PDE4D inhibitor is able to enhance memory in AD transgenic mice and concomitantly rescues their hippocampal long-term potentiation deficit. Of great relevance, our preliminary toxicological analysis indicates that GEBR-32a is not cytotoxic and genotoxic, and does not seem to possess emetic-like side effects. In conclusion, GEBR-32a could represent a very promising cognitive-enhancing drug with a great potential for the treatment of Alzheimer’s disease.
- Published
- 2017
- Full Text
- View/download PDF
46. SUMO1 impact on Alzheimer disease pathology in an amyloid-depositing mouse model
- Author
-
Taiichi Katayama, Kanayo Satoh, Hong Zhang, Agnieszka Staniszewski, Kyung Han, Hironori Takamura, Erin Knock, Grace Rooke, Ottavio Arancio, Paul E. Fraser, and Shinsuke Matsuzaki
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,Pathology ,medicine.medical_specialty ,Amyloid ,Dendritic spine ,SUMO-1 Protein ,SUMO protein ,Mice, Transgenic ,Plaque, Amyloid ,Article ,Learning and memory ,lcsh:RC321-571 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Alzheimer Disease ,mental disorders ,Amyloid precursor protein ,medicine ,Animals ,Humans ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Amyloid beta-Peptides ,biology ,Microglia ,Chemistry ,Brain ,Neurofibrillary tangle ,medicine.disease ,Electrophysiology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Alzheimer disease transgenic mouse models ,SUMO ,biology.protein ,Female ,Alzheimer's disease ,030217 neurology & neurosurgery - Abstract
Small ubiquitin-related modifiers (SUMOs) conjugated or bound to target proteins can affect protein trafficking, processing and solubility. SUMOylation has been suggested to play a role in the amyloid plaque and neurofibrillary tangle pathology of Alzheimer disease (AD) and related neurodegenerative diseases. The current study examines the impact of SUMO1 on processing of the amyloid precursor protein (APP) leading to the production and deposition of the amyloid-β (Aβ) peptide. An in vivo model of these pathways was developed by the generation of double transgenic mice over-expressing human SUMO1 and a mutant APP. The SUMO1-APP transgenics displayed normal APP processing but, at later ages, exhibited increased insoluble Aβ and plaque density accompanied by increased dendritic spine loss, more pronounced synaptic and cognitive deficits. These findings suggest a potential impairment in Aβ clearance as opposed to increased amyloid production. Examination of microglia indicated a reduction in the SUMO1-APP transgenics which is a possible mechanism for the SUMO1-mediated increase in amyloid load. These findings suggest an indirect activity of SUMO1 possibly in the removal of Aβ plaques rather than a direct impact on amyloid generation.
- Published
- 2017
47. CRISPR/Cas9-Correctable mutation-related molecular and physiological phenotypes in iPSC-derived Alzheimer’s PSEN2N141I neurons
- Author
-
Maitane Ortiz-Virumbrales, Samson T. Jacob, Michelle E. Ehrlich, Scott Noggle, Cesar L. Moreno, Andrew Sproul, Matthew Zimmer, Ottavio Arancio, I. A. Kruglikov, Paula Marazuela, Eric E. Schadt, Daniel Paull, Bin Zhang, Sam Gandy, and Rudolph E. Tanzi
- Subjects
0301 basic medicine ,Male ,Action Potentials ,BFCN ,lcsh:RC346-429 ,0302 clinical medicine ,Neural Stem Cells ,CRISPR ,Cholinergic ,Gene Editing ,PSEN2 ,iPSC ,Cell Death ,Cholinergic Neurons ,Electrophysiology ,Rheobase ,Mutation (genetic algorithm) ,Female ,Alzheimer’s disease ,Adult ,Heterozygote ,Basal Forebrain ,Neurogenesis ,Induced Pluripotent Stem Cells ,Biology ,Presenilin ,Pathology and Forensic Medicine ,Cell Line ,Basal forebrain ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,Presenilin-2 ,Humans ,RNA, Messenger ,Cholinergic neuron ,CRISPR/Cas9 ,lcsh:Neurology. Diseases of the nervous system ,Adaptor Proteins, Signal Transducing ,Amyloid beta-Peptides ,Point mutation ,Research ,Peptide Fragments ,030104 developmental biology ,Mutation ,Neurology (clinical) ,CRISPR-Cas Systems ,Apoptosis Regulatory Proteins ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Basal forebrain cholinergic neurons (BFCNs) are believed to be one of the first cell types to be affected in all forms of AD, and their dysfunction is clinically correlated with impaired short-term memory formation and retrieval. We present an optimized in vitro protocol to generate human BFCNs from iPSCs, using cell lines from presenilin 2 (PSEN2) mutation carriers and controls. As expected, cell lines harboring the PSEN2 N141I mutation displayed an increase in the Aβ42/40 in iPSC-derived BFCNs. Neurons derived from PSEN2 N141I lines generated fewer maximum number of spikes in response to a square depolarizing current injection. The height of the first action potential at rheobase current injection was also significantly decreased in PSEN2 N141I BFCNs. CRISPR/Cas9 correction of the PSEN2 point mutation abolished the electrophysiological deficit, restoring both the maximal number of spikes and spike height to the levels recorded in controls. Increased Aβ42/40 was also normalized following CRISPR/Cas-mediated correction of the PSEN2 N141I mutation. The genome editing data confirms the robust consistency of mutation-related changes in Aβ42/40 ratio while also showing a PSEN2-mutation-related alteration in electrophysiology.
- Published
- 2017
48. Eicosanoyl-5-hydroxytryptamide (EHT) prevents Alzheimer's disease-related cognitive and electrophysiological impairments in mice exposed to elevated concentrations of oligomeric beta-amyloid
- Author
-
Scott L. Melideo, Agnieszka Staniszewski, Jeffry B. Stock, Michael Voronkov, Ottavio Arancio, Russell E. Nicholls, Maxwell Stock, Eduardo Perez, Hong Zhang, Kesava Asam, Adolfo Mazzeo, and Kristen L. Huber
- Subjects
0301 basic medicine ,Male ,Physiology ,Long-Term Potentiation ,Hippocampus ,lcsh:Medicine ,Water maze ,Pharmacology ,Alzheimer's Disease ,Coffee ,Biochemistry ,Mice ,0302 clinical medicine ,Cognition ,Behavioral Conditioning ,Conditioning, Psychological ,Medicine and Health Sciences ,Medicine ,Phosphorylation ,Post-Translational Modification ,lcsh:Science ,Cognitive Impairment ,Multidisciplinary ,Neuronal Plasticity ,Animal Behavior ,Cognitive Neurology ,Chemical Reactions ,Long-term potentiation ,Neurodegenerative Diseases ,Fear ,3. Good health ,Electrophysiology ,Chemistry ,Neurology ,Physical Sciences ,Female ,Research Article ,Serotonin ,Amyloid ,Transgene ,Cognitive Neuroscience ,Phosphatase ,Methylation ,03 medical and health sciences ,Alzheimer Disease ,Neuroplasticity ,Mental Health and Psychiatry ,Animals ,Maze Learning ,Nutrition ,Behavior ,Amyloid beta-Peptides ,business.industry ,lcsh:R ,Biology and Life Sciences ,Proteins ,Protein phosphatase 2 ,Diet ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Solubility ,Cognitive Science ,Dementia ,lcsh:Q ,Nervous System Diseases ,business ,Cognition Disorders ,Zoology ,Fear Conditioning ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Soluble forms of oligomeric beta-amyloid (Aβ) are thought to play a central role in Alzheimer’s disease (AD). Transgenic manipulation of methylation of the serine/threonine protein phosphatase, PP2A, was recently shown to alter the sensitivity of mice to AD-related impairments resulting from acute exposure to elevated levels of Aβ. In addition, eicosanoyl-5-hydroxytryptamide (EHT), a naturally occurring component from coffee beans that modulates PP2A methylation, was shown to confer therapeutic benefits in rodent models of AD and Parkinson’s disease. Here, we tested the hypothesis that EHT protects animals from the pathological effects of exposure to elevated levels of soluble oligomeric Aβ. We treated mice with EHT-containing food at two different doses and assessed the sensitivity of these animals to Aβ-induced behavioral and electrophysiological impairments. We found that EHT administration protected animals from Aβ-induced cognitive impairments in both a radial-arm water maze and contextual fear conditioning task. We also found that both chronic and acute EHT administration prevented Aβ-induced impairments in long-term potentiation. These data add to the accumulating evidence suggesting that interventions with pharmacological agents, such as EHT, that target PP2A activity may be therapeutically beneficial for AD and other neurological conditions.
- Published
- 2017
49. Behavioral assays with mouse models of Alzheimer's disease: Practical considerations and guidelines
- Author
-
Agostino Palmeri, Ottavio Arancio, Giorgio Calabrese, Linda Lee, and Daniela Puzzo
- Subjects
Pathology ,medicine.medical_specialty ,Morris water navigation task ,Guidelines as Topic ,Water maze ,Disease ,Biochemistry ,Article ,Mice ,Alzheimer Disease ,Alzheimer’s disease ,behavior ,mouse models ,medicine ,Animals ,Cognitive skill ,Pharmacology ,Behavior, Animal ,Drug discovery ,Cognition ,medicine.disease ,Disease etiology ,Disease Models, Animal ,Alzheimer's disease ,Psychology ,Neuroscience - Abstract
In Alzheimer’s disease (AD) basic research and drug discovery, mouse models are essential resources for uncovering biological mechanisms, validating molecular targets and screening potential compounds. Both transgenic and non-genetically modified mouse models enable access to different types of AD-like pathology in vivo. Although there is a wealth of genetic and biochemical studies on proposed AD pathogenic pathways, as a disease that centrally features cognitive failure, the ultimate readout for any interventions should be measures of learning and memory. This is particularly important given the lack of knowledge on disease etiology – assessment by cognitive assays offers the advantage of targeting relevant memory systems without requiring assumptions about pathogenesis. A multitude of behavioral assays are available for assessing cognitive functioning in mouse models, including ones specific for hippocampal-dependent learning and memory. Here we review the basics of available transgenic and non-transgenic AD mouse models and detail three well-established behavioral tasks commonly used for testing hippocampal-dependent cognition in mice – contextual fear conditioning, radial arm water maze and Morris water maze. In particular, we discuss the practical considerations, requirements and caveats of these behavioral testing paradigms.
- Published
- 2014
- Full Text
- View/download PDF
50. Amyloid-β Peptides Disrupt Interactions Between VAMP-2 and SNAP-25 in Neuronal Cells as Determined by FRET/FLIM.
- Author
-
Sharda, Nidhi, Pengo, Thomas, Wang, Zengtao, Kandimalla, Karunya K., and Arancio, Ottavio
- Subjects
SNARE proteins ,NEUROPEPTIDES ,PEPTIDES ,FLUORESCENCE resonance energy transfer ,SYNAPTIC vesicles ,SYNAPTOPHYSIN ,AMYLOID - Abstract
Background: Synaptic dysfunction prevalent in Alzheimer's disease (AD) brain is closely associated with increased accumulation of amyloid-β (Aβ) peptides in the brain parenchyma. It is widely believed that Aβ peptides trigger synaptic dysfunction by interfering with the synaptic vesicular fusion and the release of neurotransmitters, primarily facilitated by the SNARE protein complexes formed by VAMP-2, SNAP-25, and syntaxin-1. However, Aβ interactions with SNARE proteins to ultimately disrupt synaptic vesicular fusion are not well understood.Objective: Our objective is to elucidate mechanisms by which Aβ peptides perturb SNARE complexes.Methods: Intensity (qualitative) and lifetime (quantitative) based measurements involving Forster (fluorescence) resonance energy transfer (FRET) followed by fluorescence lifetime imaging microscopy (FLIM) were employed to investigate the effect of Aβ peptides on dynamic interactions between VAMP-2, labeled with cerulean (Cer) at the N-terminus (FRET donor), and SNAP-25 labeled with citrine (Cit) on the N-terminus (FRET acceptor). The FRET and FLIM interactions at the exocytosis locations on the pre-synaptic membrane were recorded under spontaneous and high potassium evoked conditions. Moreover, cellular accumulation of fluorescein labeled Aβ (F-Aβ) peptides and their co-localization with Cer-VAMP2 was investigated by confocal microscopy.Results: The F-Aβ40 and F-Aβ42 are internalized by differentiated N2A cells, where they colocalize with Cer-VAMP2. Both Aβ40 and Aβ42 decrease interactions between the N-termini of Cer-VAMP2 and Cit-SNAP25 in N2A cells, as determined by FRET/FLIM.Conclusion: By perturbing the N-terminal interactions between VAMP-2 and SNAP-25, Aβ40 and Aβ42, can directly interfere with the SNARE complex formation, which is critical for the docking and fusion of synaptic vesicles. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.