1
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Unveiling the Therapeutic Potential of Kelulut (Stingless Bee) Honey in Alzheimer's Disease: Findings from a Rat Model Study. Antioxidants (Basel) 2024; 13:926. [PMID: 39199172 PMCID: PMC11351951 DOI: 10.3390/antiox13080926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
Alzheimer's disease (AD) poses a major worldwide health challenge because of its profound impact on cognitive abilities and overall well-being. Despite extensive research and numerous clinical trials, therapeutic options remain limited. Our study aimed to investigate the potential of Kelulut honey (KH) as a novel therapeutic agent for addressing the multifactorial pathology of AD. We tried to evaluate the disease-attenuating and neuroprotective potential of KH in the intrahippocampally induced AD rat model by utilizing histochemistry and enzyme-linked immunosorbent assay (ELISA) studies. A total of 26 male Sprague Dawley rats weighing ~280-380 g were randomly divided into three groups: Control, AD-induced (Aβ), and AD-induced and treated with KH (Aβ+KH). The latter two groups underwent stereotaxic surgery, where 6.25 µg of amyloid β1-42 peptides were injected intrahippocampally. One-week post-surgery, KH was administered to the treatment group at a dose of 1 g/kg body weight for a period of four weeks, after which the rats went through behavior tests. After completion of behavior analysis, the rats were sacrificed, and the brains were processed for histochemistry and ELISA studies. The open field test analysis demonstrated that KH improved the locomotion of Aβ+KH compared to Aβ (p = 0.0013). In comparison, the Morris water maze did not show any nootropic effects on cognition with a paradoxical increase in time spent in the target quadrant by the Aβ group (p = 0.029). Histochemical staining showed markedly increased Congo-red-stained amyloid plaques, which were significantly reduced in dentate gyrus of Aβ+KH compared to Aβ (p < 0.05). Moreover, significantly higher apoptosis was seen in the Aβ group compared to Aβ+KH (p < 0.01) and control groups (p < 0.001). Furthermore, the ELISA studies deduced more phosphorylated tau in the diseased group compared to Aβ+KH (p = 0.038) and controls (p = 0.016). These findings suggest that KH consumption for twenty-eight days has the potential to attenuate the pathological burden of disease while exerting neuroprotective effects in rodent models of AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| |
Collapse
|
2
|
Awogbindin IO, Ikeji CN, Adedara IA, Farombi EO. Neurotoxicity of furan in juvenile Wistar rats involves behavioral defects, microgliosis, astrogliosis and oxidative stress. Food Chem Toxicol 2023:113934. [PMID: 37423315 DOI: 10.1016/j.fct.2023.113934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/21/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Evidence suggests that furan, a widespread environmental and food contaminant, causes liver toxicity and cancer, but its implications in the brain are not well defined. We measured behavioral, glial, and biochemical responses in male juvenile rats exposed orally to 2.5, 5 and 10 mg/kg furan and vitamin E after 28 days. Furan-mediated hyperactivity peaked at 5 mg/kg and did not exacerbate at 10 mg/kg. Enhanced motor defect was also observed at 10 mg/kg. Furan-treated rats elicited inquisitive exploration but showed impaired working memory. Without compromising the blood-brain barrier, furan induced glial reactivity with enhanced phagocytic activity, characterized by parenchyma-wide microglial aggregation and proliferation, which switched from hyper-ramified to rod-like morphology with increasing doses. Furan altered the glutathione-S-transferase-driven enzymatic and non-enzymatic antioxidant defence systems differentially and dose-dependently across brain regions. Redox homeostasis was most perturbed in the striatum and least disrupted in hippocampus/cerebellum. Vitamin E supplementation attenuated exploratory hyperactivity and glial reactivity but did not affect impaired working memory and oxidative imbalance. Overall, sub-chronic exposure of juvenile rats to furan triggered glial reactivity and behavioral defects suggesting the brain's vulnerability during juvenile development to furan toxicity. It remains to be determined whether environmentally relevant furan concentrations interfere with critical brain developmental milestones.
Collapse
Affiliation(s)
- Ifeoluwa O Awogbindin
- Molecular Drug Metabolism and Toxicology Research Laboratories, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Cynthia N Ikeji
- Molecular Drug Metabolism and Toxicology Research Laboratories, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Isaac A Adedara
- Molecular Drug Metabolism and Toxicology Research Laboratories, College of Medicine, University of Ibadan, Ibadan, Nigeria; Department of Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Ebenezer O Farombi
- Molecular Drug Metabolism and Toxicology Research Laboratories, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
3
|
Spatial Disorientation Under Dark Conditions Across Development in an Alzheimer's Disease Mouse Model. Neuroscience 2023; 511:53-69. [PMID: 36587866 DOI: 10.1016/j.neuroscience.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is associated with hippocampal neuropathology and cognitive impairments, including wandering behavior or becoming lost in a familiar environment. Wandering behavior is severe and manifests early in life for people with specific genetic mutations. Genetic mouse models of AD have been developed to characterize the onset and progression of behavioral deficits that represent human behaviors, such as wandering, to test the efficacy of therapeutics. It is not clear if current assessments of mouse models capture the onset of AD or a snapshot of its progression. Sequential analysis of open field behavior provides a robust, quick test to dissociate navigation cues that contribute to spatial disorientation, a feature of wandering. Despite potential utility in evaluating this feature of AD, little work has been reported using animal models of dementia in this task. Thus, we examined the use of different sources of information to maintain spatial orientation at two prodromal ages in female transgenic CRND8 AD (n = 17) and Control mice (n = 16). These mice exhibit amyloid plaques, a hallmark neuropathological feature of AD, that are associated with cognitive dysfunction at ∼three months of age. Spatial disorientation was observed at two months and more severely at four months under dark conditions, but performance was spared when visual environmental cues were available. This study provides documentation of impaired self-movement cue processing in AD mice, establishing the dark open field as a behavioral tool to characterize spatial disorientation associated with AD. These findings may accelerate future assessments of novel therapeutic interventions for neurological disorders.
Collapse
|
4
|
Zhang L, Wong LR, Wong P, Shen W, Yang S, Huang L, Lim YA, Ho PCL. Chronic treatment with baicalein alleviates behavioural disorders and improves cerebral blood flow via reverting metabolic abnormalities in a J20 transgenic mouse model of Alzheimer's disease. Brain Behav Immun Health 2023; 28:100599. [PMID: 36817510 PMCID: PMC9931920 DOI: 10.1016/j.bbih.2023.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/21/2023] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
Baicalein (BE) has both antioxidant and anti-inflammatory effects. It has also been reported able to improve cerebral blood circulation in brain ischemic injury. However, its chronic efficacy and metabolomics in Alzheimer's disease (AD) remain unknown. In this study, BE at 80 mg/kg was administrated through the oral route in J20 AD transgenic mice aged from aged 4 months to aged 10 months. Metabolic- and neurobehavioural phenotyping was done before and after 6 months' treatment to evaluate the drug efficacy and the relevant mechanisms. Meanwhile, molecular docking was used to study the binding affinity of BE and poly (ADP-ribose) polymerase-1 (PARP-1) which is related to neuronal injury. The open field test showed that BE could suppress hyperactivity in J20 mice and increase the frequency of the target quadrant crossing in the Morris Water Maze test. More importantly, BE restored cerebral blood flow back to the normal level after the chronic treatment. A 1H NMR-based metabolomics study showed that BE treatment could restore the tricarboxylic acid cycle in plasma. And such a treatment could suppress oxidative stress, inhibit neuroinflammation, alleviate mitochondrial dysfunction, improve neurotransmission, and restore amino homeostasis via starch and sucrose metabolism and glycolipid metabolism in the cortex and hippocampus, which could affect the behavioural and cerebral blood flow. These findings showed that BE is a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Li Zhang
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 117583, Singapore,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Ling Rong Wong
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Peiyan Wong
- Neuroscience Phenotyping Core, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Wanxiang Shen
- Department of Chemistry, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Shili Yang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yun-An Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Paul Chi-Lui Ho
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 117583, Singapore,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore,Monash University Malaysia, School of Pharmacy, Subang Jaya, 47500, Selangor, Malaysia,Corresponding author. Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
| |
Collapse
|
5
|
Löscher W, Stafstrom CE. Epilepsy and its neurobehavioral comorbidities: Insights gained from animal models. Epilepsia 2023; 64:54-91. [PMID: 36197310 DOI: 10.1111/epi.17433] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023]
Abstract
It is well established that epilepsy is associated with numerous neurobehavioral comorbidities, with a bidirectional relationship; people with epilepsy have an increased incidence of depression, anxiety, learning and memory difficulties, and numerous other psychosocial challenges, and the occurrence of epilepsy is higher in individuals with those comorbidities. Although the cause-and-effect relationship is uncertain, a fuller understanding of the mechanisms of comorbidities within the epilepsies could lead to improved therapeutics. Here, we review recent data on epilepsy and its neurobehavioral comorbidities, discussing mainly rodent models, which have been studied most extensively, and emphasize that clinically relevant information can be gained from preclinical models. Furthermore, we explore the numerous potential factors that may confound the interpretation of emerging data from animal models, such as the specific seizure induction method (e.g., chemical, electrical, traumatic, genetic), the role of species and strain, environmental factors (e.g., laboratory environment, handling, epigenetics), and the behavioral assays that are chosen to evaluate the various aspects of neural behavior and cognition. Overall, the interplay between epilepsy and its neurobehavioral comorbidities is undoubtedly multifactorial, involving brain structural changes, network-level differences, molecular signaling abnormalities, and other factors. Animal models are well poised to help dissect the shared pathophysiological mechanisms, neurological sequelae, and biomarkers of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Barker-Haliski M, Nishi T, White HS. Soticlestat, a novel cholesterol 24-hydroxylase inhibitor, modifies acute seizure burden and chronic epilepsy-related behavioral deficits following Theiler's virus infection in mice. Neuropharmacology 2023; 222:109310. [PMID: 36341806 DOI: 10.1016/j.neuropharm.2022.109310] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Temporal lobe epilepsy is the most common form of acquired epilepsy and can arise due to multiple inciting events, including central nervous system (CNS) infection. CNS infection with the Theiler's murine encephalomyelitis virus (TMEV) in male C57Bl/6J mice leads to acute, drug-resistant handling-induced seizures. Cholesterol 24-hydroxylase (CH24H) is a brain-specific enzyme that converts cholesterol into 24S-hydroxycholesterol; the primary mechanism of cholesterol catabolism in the brain. The novel CH24H inhibitor, soticlestat (SOT; or TAK-935), demonstrates the potential to restore excitatory/inhibitory balance in multiple preclinical models of hyperexcitability. This study thus sought to characterize the anticonvulsant potential of SOT in the TMEV model. Treatment with SOT (30 mg/kg, p.o.; n = 30) 0-7 days post-infection (DPI) reduced overall seizure burden and severity. SOT administration significantly delayed onset of infection-induced Racine stage 5 seizures, from 8.6 ± 0.6 (VEH-treated) to 10.8 ± 0.8 (SOT-treated) observation sessions. Infected mice were then allowed 36 days treatment-free recovery before assessing impact of earlier drug administration on epilepsy-related cognitive and behavioral comorbidities, including a non-habituated open field (OF) task. Total OF distance traveled was significantly less in SOT-treated mice compared to VEH-treated mice, suggesting attenuated TMEV-induced spatial memory deficits, or reduced chronic hyperexcitability. Mice with history of SOT treatment also spent significantly more time and traveled farther in the OF center, indicative of reduced epilepsy-induced anxiety-like behavior. These studies suggest that SOT is a mechanistically novel agent for symptomatic seizure control. Moreover, acute SOT administration during an epileptogenic insult may attenuate the resulting long-term behavioral comorbidities of epilepsy.
Collapse
Affiliation(s)
| | - Toshiya Nishi
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Japan; Takeda Pharmaceutical Company Limited, Cambridge, MA, 02139, USA.
| | - H Steve White
- University of Washington, Department of Pharmacy, Seattle, WA, USA
| |
Collapse
|
7
|
Grünblatt E, Homolak J, Babic Perhoc A, Davor V, Knezovic A, Osmanovic Barilar J, Riederer P, Walitza S, Tackenberg C, Salkovic-Petrisic M. From attention-deficit hyperactivity disorder to sporadic Alzheimer's disease-Wnt/mTOR pathways hypothesis. Front Neurosci 2023; 17:1104985. [PMID: 36875654 PMCID: PMC9978448 DOI: 10.3389/fnins.2023.1104985] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder with the majority of patients classified as sporadic AD (sAD), in which etiopathogenesis remains unresolved. Though sAD is argued to be a polygenic disorder, apolipoprotein E (APOE) ε4, was found three decades ago to pose the strongest genetic risk for sAD. Currently, the only clinically approved disease-modifying drugs for AD are aducanumab (Aduhelm) and lecanemab (Leqembi). All other AD treatment options are purely symptomatic with modest benefits. Similarly, attention-deficit hyperactivity disorder (ADHD), is one of the most common neurodevelopmental mental disorders in children and adolescents, acknowledged to persist in adulthood in over 60% of the patients. Moreover, for ADHD whose etiopathogenesis is not completely understood, a large proportion of patients respond well to treatment (first-line psychostimulants, e.g., methylphenidate/MPH), however, no disease-modifying therapy exists. Interestingly, cognitive impairments, executive, and memory deficits seem to be common in ADHD, but also in early stages of mild cognitive impairment (MCI), and dementia, including sAD. Therefore, one of many hypotheses is that ADHD and sAD might have similar origins or that they intercalate with one another, as shown recently that ADHD may be considered a risk factor for sAD. Intriguingly, several overlaps have been shown between the two disorders, e.g., inflammatory activation, oxidative stress, glucose and insulin pathways, wingless-INT/mammalian target of rapamycin (Wnt/mTOR) signaling, and altered lipid metabolism. Indeed, Wnt/mTOR activities were found to be modified by MPH in several ADHD studies. Wnt/mTOR was also found to play a role in sAD and in animal models of the disorder. Moreover, MPH treatment in the MCI phase was shown to be successful for apathy including some improvement in cognition, according to a recent meta-analysis. In several AD animal models, ADHD-like behavioral phenotypes have been observed indicating a possible interconnection between ADHD and AD. In this concept paper, we will discuss the various evidence in human and animal models supporting the hypothesis in which ADHD might increase the risk for sAD, with common involvement of the Wnt/mTOR-pathway leading to lifespan alteration at the neuronal levels.
Collapse
Affiliation(s)
- Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and the Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Jan Homolak
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Babic Perhoc
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Virag Davor
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Jelena Osmanovic Barilar
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Peter Riederer
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany.,Department and Research Unit of Psychiatry, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and the Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Christian Tackenberg
- Neuroscience Center Zurich, University of Zurich and the Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Switzerland
| | - Melita Salkovic-Petrisic
- Department of Pharmacology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
8
|
Alzahrani JM, Murray KO, Gambino BJ, Garcia CK, Sheikh LH, Cusack KJ, Laitano O, Clanton TL. Neuromotor deficits and altered physiological responses to repeated exertional heat stroke exposures in mice. Am J Physiol Regul Integr Comp Physiol 2022; 323:R951-R961. [PMID: 36279505 PMCID: PMC9722251 DOI: 10.1152/ajpregu.00152.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022]
Abstract
Exertional heat stroke (EHS) is a life-threatening illness that can lead to negative health outcomes. Using a "severe" preclinical mouse model of EHS, we tested the hypotheses that one EHS exposure results in altered susceptibility to a subsequent EHS and reduced neuromotor performance. Female C57BL/6 mice underwent two protocols, 2 wk apart, either an EHS trial (EHS) or a sham exercise control trial (EXC). For EHS, mice ran in a forced running wheel at 37.5°C/40% relative humidity until loss of consciousness, followed by a slow cooling protocol (2 h recovery at 37.5°C). EXC mice exercised equally but in ∼22°C. Mice were randomized into three groups: 1) EXC-EXC (two consecutive EXC, n = 6, 2) EHS-EXC (EHS followed by EXC, n = 5), and 3) EHS-EHS (repeated EHS, n = 9). Mice underwent noninvasive neuromotor and behavioral tests during recovery and isolated soleus force measurements at the end of recovery. At the first EHS, mice reached average peak core temperatures (Tc,max) of 42.4°C, (46% mortality). On the second EHS, average Tc,max was reduced by ∼0.7°C (P < 0.05; mortality 18%). After the first EHS, both EHS-EX and EHS-EHS showed significant reductions in maximum strength (24 h and 1 wk post). After the second EHS, strength, horizontal rotation, hindlimb tone, suspended hindlimb splay, trunk curl, and provoked biting continued to decline in the EHS-EHS group. In conclusion, exposure to a second EHS after 2 wk leads to increased exercise times in the heat, symptom limitation at a lower Tc,max, and greater deficits in neuromotor and behavioral function during recovery.
Collapse
Affiliation(s)
- Jamal M Alzahrani
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Kevin O Murray
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Bryce J Gambino
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Christian K Garcia
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Laila H Sheikh
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Kevin J Cusack
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Thomas L Clanton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| |
Collapse
|
9
|
Shen Y, Qian L, Luo H, Li X, Ruan Y, Fan R, Si Z, Chen Y, Li L, Liu Y. The Significance of NLRP Inflammasome in Neuropsychiatric Disorders. Brain Sci 2022; 12:brainsci12081057. [PMID: 36009120 PMCID: PMC9406040 DOI: 10.3390/brainsci12081057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 12/02/2022] Open
Abstract
The NLRP inflammasome is a multi-protein complex which mainly consists of the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain. Its activation is linked to microglial-mediated neuroinflammation and partial neuronal degeneration. Many neuropsychiatric illnesses have increased inflammatory responses as both a primary cause and a defining feature. The NLRP inflammasome inhibition delays the progression and alleviates the deteriorating effects of neuroinflammation on several neuropsychiatric disorders. Evidence on the central effects of the NLRP inflammasome potentially provides the scientific base of a promising drug target for the treatment of neuropsychiatric disorders. This review elucidates the classification, composition, and functions of the NLRP inflammasomes. It also explores the underlying mechanisms of NLRP inflammasome activation and its divergent role in neuropsychiatric disorders, including Alzheimer’s disease, Huntington’s disease, Parkinson’s disease, depression, drug use disorders, and anxiety. Furthermore, we explore the treatment potential of the NLRP inflammasome inhibitors against these disorders.
Collapse
Affiliation(s)
- Yao Shen
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
| | - Liyin Qian
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
| | - Hu Luo
- Department of Psychology, Faculty of Teacher Education, Ningbo University, Ningbo 315021, China
| | - Xiaofang Li
- Department of Psychology, Faculty of Teacher Education, Ningbo University, Ningbo 315021, China
| | - Yuer Ruan
- Department of Psychology, Faculty of Teacher Education, Ningbo University, Ningbo 315021, China
| | - Runyue Fan
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
- Ningbo Yinzhou District Center for Disease Control and Prevention, Ningbo 315199, China
| | - Zizhen Si
- Department of Physiological Pharmacology, School of Medicine, Ningbo University, Ningbo 315021, China
- Department of Pharmacology, Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
| | - Yunpeng Chen
- Department of Public Health, School of Medicine, Ningbo University, Ningbo 315021, China
| | - Longhui Li
- Ningbo Kangning Hospital, Ningbo 315201, China
| | - Yu Liu
- Department of Physiological Pharmacology, School of Medicine, Ningbo University, Ningbo 315021, China
- Correspondence:
| |
Collapse
|
10
|
The effects of locomotion on sensory-evoked haemodynamic responses in the cortex of awake mice. Sci Rep 2022; 12:6236. [PMID: 35422473 PMCID: PMC9010417 DOI: 10.1038/s41598-022-10195-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/04/2022] [Indexed: 12/22/2022] Open
Abstract
Investigating neurovascular coupling in awake rodents is becoming ever more popular due, in part, to our increasing knowledge of the profound impacts that anaesthesia can have upon brain physiology. Although awake imaging brings with it many advantages, we still do not fully understand how voluntary locomotion during imaging affects sensory-evoked haemodynamic responses. In this study we investigated how evoked haemodynamic responses can be affected by the amount and timing of locomotion. Using an awake imaging set up, we used 2D-Optical Imaging Spectroscopy (2D-OIS) to measure changes in cerebral haemodynamics within the sensory cortex of the brain during either 2 s whisker stimulation or spontaneous (no whisker stimulation) experiments, whilst animals could walk on a spherical treadmill. We show that locomotion alters haemodynamic responses. The amount and timing of locomotion relative to whisker stimulation is important, and can significantly impact sensory-evoked haemodynamic responses. If locomotion occurred before or during whisker stimulation, the amplitude of the stimulus-evoked haemodynamic response was significantly altered. Therefore, monitoring of locomotion during awake imaging is necessary to ensure that conclusions based on comparisons of evoked haemodynamic responses (e.g., between control and disease groups) are not confounded by the effects of locomotion.
Collapse
|
11
|
Guo C, Wen D, Zhang Y, Mustaklem R, Mustaklem B, Zhou M, Ma T, Ma YY. Amyloid-β oligomers in the nucleus accumbens decrease motivation via insertion of calcium-permeable AMPA receptors. Mol Psychiatry 2022; 27:2146-2157. [PMID: 35105968 PMCID: PMC9133055 DOI: 10.1038/s41380-022-01459-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/31/2021] [Accepted: 01/18/2022] [Indexed: 12/17/2022]
Abstract
It is essential to identify the neuronal mechanisms of Alzheimer's Disease (AD)-associated neuropsychiatric symptoms, e.g., apathy, before improving the life quality of AD patients. Here, we focused on the nucleus accumbens (NAc), a critical brain region processing motivation, also known to display AD-associated pathological changes in human cases. We found that the synaptic calcium permeable (CP)-AMPA receptors (AMPARs), which are normally absent in the NAc, can be revealed by acute exposure to Aβ oligomers (AβOs), and play a critical role in the emergence of synaptic loss and motivation deficits. Blockade of NAc CP-AMPARs can effectively prevent AβO-induced downsizing and pruning of spines and silencing of excitatory synaptic transmission. We conclude that AβO-triggered synaptic insertion of CP-AMPARs is a key mechanism mediating synaptic degeneration in AD, and preserving synaptic integrity may prevent or delay the onset of AD-associated psychiatric symptoms.
Collapse
Affiliation(s)
- Changyong Guo
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Di Wen
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yihong Zhang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Richie Mustaklem
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Basil Mustaklem
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Miou Zhou
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Tao Ma
- Department of Internal Medicine-Gerontology and Geriatric Medicine; Department of Physiology and Pharmacology; Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Yao-Ying Ma
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
12
|
Salehi C, Seiiedy M, Soraya H, Fazli F, Ghasemnejad-Berenji M. Pretreatment with bisoprolol and vitamin E alone or in combination provides neuroprotection against cerebral ischemia/reperfusion injury in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:685-695. [PMID: 33106920 DOI: 10.1007/s00210-020-02007-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/19/2020] [Indexed: 01/26/2023]
Abstract
Global cerebral ischemia/reperfusion (I/R) induces selective neuronal injury in the hippocampus, leading to severe impairment in behavior, learning, and memory functions. This study aimed to evaluate the neuroprotective effects of bisoprolol (biso) and vitamin E (vit E) treatment alone or in combination on cerebral ischemia/reperfusion (I/R) injury. A total of 30 male rats were divided randomly into five groups (n = 6), sham, I/R, I/R + biso, I/R + vit E, and I/R + biso+vit E. Cerebral I/R group underwent global ischemia by bilateral common carotid artery occlusion for 20 min. Treatment groups received drugs once daily intraperitoneally for 7 days before the I/R induction. Locomotive and cognitive behaviors were utilized by open-field and Morris water maze tests. After behavioral testing, the brain was removed and processed to evaluate cerebral infarct size, histopathologic changes, myeloperoxidase (MPO) activity, and malondialdehyde (MDA) level. In I/R group tissue MDA and MPO levels and cerebral infarct size were significantly increased in comparison with the sham group. Furthermore, significant deficits were observed in locomotion and spatial memory after I/R. The areas of cerebral infarction, MPO, and MDA levels in biso, vit E, and combination group were significantly reduced compared with I/R group. Histopathological analysis demonstrated a significant reduction in leukocyte infiltration in all treated groups with the most profound reduction in the combination group. According to the behavioral tests, administration of biso and/or vit E protected locomotive ability and improved spatial memory after cerebral I/R. Our findings show that biso and vit E have beneficial effects against the I/R injury and due to their synergistic effects when administered in combination, may have a more pronounced protective effect on the cerebral I/R injury.
Collapse
Affiliation(s)
- Chiman Salehi
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, PO Box 5715799313, Urmia, Iran
| | - Monireh Seiiedy
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, PO Box 5715799313, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, PO Box 5715799313, Urmia, Iran.
| | - Farzaneh Fazli
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, PO Box 5715799313, Urmia, Iran
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, PO Box 5715799313, Urmia, Iran.
| |
Collapse
|
13
|
Liu M, Beckett TL, Thomason LAM, Dorr A, Stefanovic B, McLaurin J. Covert strokes prior to Alzheimer's disease onset accelerate peri-lesional pathology but not cognitive deficits in an inducible APP mouse model. Brain Res 2021; 1754:147233. [PMID: 33412147 DOI: 10.1016/j.brainres.2020.147233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022]
Abstract
It is estimated that up to 1 in 3 healthy middle-aged adults will have had a covert stroke during their lifetime. Furthermore, post-stroke, survivors are more than twice as likely to develop dementia. In the present study, we aimed to model the impact of focal subclinical ischemia prior to the onset of AD pathogenesis in a preclinical model. We utilized endothelin-1 to induce ischemia in an iducible transgenic mouse model of Alzheimer's disease, APPsi:tTA, allowing for temporal control of APP gene expression. We induced the focal subclinical ischemic events in the absence of APP expression, thus prior to AD onset. T2 structural magnetic resonance imaging confirmed the volume and location of focal subclinical ischemic lesions to the medial prefrontal cortex. Following recovery from surgery and 7 weeks of APP expression, we found that two subclinical ischemic lesions resulted in a significant localized increase in amyloid load and in microglial activation proximal to the lesion. However, no differences were found in astrogliosis. A battery of behaviour tests was conducted, in which no significant differences were detected in activities of daily living and cognitive function between stroked and sham cohorts. Overall, our results demonstrated that APP expression was the sole driving force behind behavioural deficits. In conclusion, our results suggest that a history of two subclinical strokes prior to AD onset does not worsen early disease trajectory in a mouse model.
Collapse
Affiliation(s)
- Mingzhe Liu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.
| | - Tina L Beckett
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Adrienne Dorr
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Bojana Stefanovic
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - JoAnne McLaurin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
14
|
Antunes MS, Ladd FVL, Ladd AABL, Moreira AL, Boeira SP, Cattelan Souza L. Hesperidin protects against behavioral alterations and loss of dopaminergic neurons in 6-OHDA-lesioned mice: the role of mitochondrial dysfunction and apoptosis. Metab Brain Dis 2021; 36:153-167. [PMID: 33057922 DOI: 10.1007/s11011-020-00618-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/14/2020] [Indexed: 01/01/2023]
Abstract
Hesperidin is a flavonoid glycoside that is frequently found in citrus fruits. Our group have demonstrated that hesperidin has neuroprotective effect in 6-hydroxydopamine (6-OHDA) model of Parkinson's disease (PD), mainly by antioxidant mechanisms. Although the pathophysiology of PD remains uncertain, a large body of evidence has demonstrated that mitochondrial dysfunction and apoptosis play a critical role in dopaminergic nigrostriatal degeneration. However, the ability of hesperidin in modulating these mechanisms has not yet been investigated. In the present study, we examined the potential of a 28-day hesperidin treatment (50 mg/kg/day, p.o.) in preventing behavioral alterations induced by 6-OHDA injection via regulating mitochondrial dysfunction, apoptosis and dopaminergic neurons in the substantia nigra pars compacta (SNpc) in C57BL/6 mice. Our results demonstrated that hesperidin treatment improved motor, olfactory and spatial memory impairments elicited by 6-OHDA injection. Moreover, hesperidin treatment attenuated the loss of dopaminergic neurons (TH+ cells) in the SNpc and the depletion of dopamine (DA) and its metabolities 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA) in the striatum of 6-OHDA-lesioned mice. Hesperidin also protected against the inhibition of mitochondrial respiratory chain complex-I, -IV and V, the decrease of Na + -K + -ATPase activity and the increase of caspase-3 and -9 activity in the striatum. Taken together, our findings indicate that hesperidin mitigates the degeneration of dopaminergic neurons in the SNpc by preventing mitochondrial dysfunction and modulating apoptotic pathways in the striatum of 6-OHDA-treated mice, thus improving behavioral alterations. These results provide new insights on neuroprotective mechanisms of hesperidin in a relevant preclinical model of PD.
Collapse
Affiliation(s)
- Michelle S Antunes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui, RS, CEP 97650-000, Brazil
| | - Fernando Vagner Lobo Ladd
- Department of Morphology/Laboratory of Neuroanatom, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Aliny Antunes Barbosa Lobo Ladd
- Laboratory of Stochastic Stereology and Chemical Anatomy, Department of Surgery, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Amanda Lopez Moreira
- Laboratory of Stochastic Stereology and Chemical Anatomy, Department of Surgery, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Silvana Peterini Boeira
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui, RS, CEP 97650-000, Brazil
| | - Leandro Cattelan Souza
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui, RS, CEP 97650-000, Brazil.
| |
Collapse
|
15
|
Cognitive impairment appears progressive in the mdx mouse. Neuromuscul Disord 2020; 30:368-388. [PMID: 32360405 PMCID: PMC7306157 DOI: 10.1016/j.nmd.2020.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 11/22/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive muscle wasting disease caused by mutations in the DMD gene, which encodes the large cytoskeletal protein dystrophin. Approximately one-third of DMD patient's exhibit cognitive problems yet it is unknown if cognitive impairments worsen with age. The mdx mouse model is deficient in dystrophin demonstrates cognitive abnormalities, but no studies have investigated this longitudinally. We assessed the consequences of dystrophin deficiency on brain morphology and cognition in male mdx mice. We utilised non-invasive methods to monitor CNS pathology with an aim to identify changes longitudinally (between 4 and 18 months old) which could be used as outcome measures. MRI identified a total brain volume (TBV) increase in control mice with ageing (p < 0.05); but the mdx mice TBV increased significantly more (p < 0.01). Voxel-based morphometry (VBM) identified decreases in grey matter volume, particularly in the hippocampus of the mdx brain, most noticeable from 12 months onwards, as were enlarged lateral ventricles in mdx mice. The caudate putamen of older mdx mice showed increases in T2- relaxometry which may be considered as evidence of increased water content. Hippocampal spatial learning and memory was decreased in mdx mice, particularly long-term memory, which progressively worsened with age. The novel object recognition (NOR) task highlighted elevated anxiety-related behaviour in older mdx mice. Our studies suggest that dystrophin deficiency causes a progressive cognitive impairment in mice (compared to ageing control mice), becoming evident at late disease stages, and may explain why progressive CNS symptoms are not obvious in DMD patients.
Collapse
|
16
|
Shepherd A, Zhang TD, Zeleznikow-Johnston AM, Hannan AJ, Burrows EL. Transgenic Mouse Models as Tools for Understanding How Increased Cognitive and Physical Stimulation Can Improve Cognition in Alzheimer's Disease. Brain Plast 2018; 4:127-150. [PMID: 30564551 PMCID: PMC6296266 DOI: 10.3233/bpl-180076] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cognitive decline appears as a core feature of dementia, of which the most prevalent form, Alzheimer's disease (AD) affects more than 45 million people worldwide. There is no cure, and therapeutic options remain limited. A number of modifiable lifestyle factors have been identified that contribute to cognitive decline in dementia. Sedentary lifestyle has emerged as a major modifier and accordingly, boosting mental and physical activity may represent a method to prevent decline in dementia. Beneficial effects of increased physical activity on cognition have been reported in healthy adults, showing potential to harness exercise and cognitive stimulation as a therapy in dementia. 'Brain training' (cognitive stimulation) has also been investigated as an intervention protecting against cognitive decline with normal aging. Consequently, the utility of exercise regimes and/or cognitive stimulation to improve cognition in dementia in clinical populations has been a major area of study. However, these therapies are in their infancy and efficacy is unclear. Investigations utilising animal models, where dose and timing of treatment can be tightly controlled, have provided many mechanistic insights. Genetically engineered mouse models are powerful tools to investigate mechanisms underlying cognitive decline, and also how environmental manipulations can alter both cognitive outcomes and pathology. A myriad of effects following physical activity and housing in enriched environments have been reported in transgenic mice expressing Alzheimer's disease-associated mutations. In this review, we comprehensively evaluate all studies applying environmental enrichment and/or increased physical exercise to transgenic mouse models of Alzheimer's disease. It is unclear whether interventions must be applied before first onset of cognitive deficits to be effective. In order to determine the importance of timing of interventions, we specifically scrutinised studies exposing transgenic mice to exercise and environmental enrichment before and after first report of cognitive impairment. We discuss the strengths and weaknesses of these preclinical studies and suggest approaches for enhancing rigor and using mechanistic insights to inform future therapeutic interventions.
Collapse
Affiliation(s)
- Amy Shepherd
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Tracy D Zhang
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Ariel M Zeleznikow-Johnston
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| | - Emma L Burrows
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
17
|
Sakakibara Y, Sekiya M, Saito T, Saido TC, Iijima KM. Cognitive and emotional alterations in App knock-in mouse models of Aβ amyloidosis. BMC Neurosci 2018; 19:46. [PMID: 30055565 PMCID: PMC6064053 DOI: 10.1186/s12868-018-0446-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/21/2018] [Indexed: 12/21/2022] Open
Abstract
Background Alzheimer’s disease (AD), the most common cause of dementia, is characterized by the progressive deposition of amyloid-β (Aβ) peptides and neurofibrillary tangles. Mouse models of Aβ amyloidosis generated by knock-in (KI) of a humanized Aβ sequence provide distinct advantages over traditional transgenic models that rely on overexpression of amyloid precursor protein (APP). In App-KI mice, three familial AD-associated mutations were introduced into the endogenous mouse App locus to recapitulate Aβ pathology observed in AD: the Swedish (NL) mutation, which elevates total Aβ production; the Beyreuther/Iberian (F) mutation, which increases the Aβ42/Aβ40 ratio; and the Arctic (G) mutation, which promotes Aβ aggregation. AppNL-G-F mice harbor all three mutations and develop progressive Aβ amyloidosis and neuroinflammatory response in broader brain areas, whereas AppNL mice carrying only the Swedish mutation exhibit no overt AD-related pathological changes. To identify behavioral alterations associated with Aβ pathology, we assessed emotional and cognitive domains of AppNL-G-F and AppNL mice at different time points, using the elevated plus maze, contextual fear conditioning, and Barnes maze tasks. Results Assessments of emotional domains revealed that, in comparison with wild-type (WT) C57BL/6J mice, AppNL-G-F/NL-G-F mice exhibited anxiolytic-like behavior that was detectable from 6 months of age. By contrast, AppNL/NL mice exhibited anxiogenic-like behavior from 15 months of age. In the contextual fear conditioning task, both AppNL/NL and AppNL-G-F/NL-G-F mice exhibited intact learning and memory up to 15–18 months of age, whereas AppNL-G-F/NL-G-F mice exhibited hyper-reactivity to painful stimuli. In the Barnes maze task, AppNL-G-F/NL-G-F mice exhibited a subtle decline in spatial learning ability at 8 months of age, but retained normal memory functions. Conclusion AppNL/NL and AppNL-G-F/NL-G-F mice exhibit behavioral changes associated with non-cognitive, emotional domains before the onset of definitive cognitive deficits. Our observations consistently indicate that AppNL-G-F/NL-G-F mice represent a model for preclinical AD. These mice are useful for the study of AD prevention rather than treatment after neurodegeneration. Electronic supplementary material The online version of this article (10.1186/s12868-018-0446-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yasufumi Sakakibara
- Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
| | - Michiko Sekiya
- Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Koichi M Iijima
- Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan. .,Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| |
Collapse
|
18
|
Klakotskaia D, Agca C, Richardson RA, Stopa EG, Schachtman TR, Agca Y. Memory deficiency, cerebral amyloid angiopathy, and amyloid-β plaques in APP+PS1 double transgenic rat model of Alzheimer's disease. PLoS One 2018; 13:e0195469. [PMID: 29641600 PMCID: PMC5895023 DOI: 10.1371/journal.pone.0195469] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/22/2018] [Indexed: 01/31/2023] Open
Abstract
Transgenic rat models of Alzheimer's disease were used to examine differences in memory and brain histology. Double transgenic female rats (APP+PS1) over-expressing human amyloid precursor protein (APP) and presenilin 1 (PS1) and single transgenic rats (APP21) over-expressing human APP were compared with wild type Fischer rats (WT). The Barnes maze assessed learning and memory and showed that both APP21 and APP+PS1 rats made significantly more errors than the WT rats during the acquisition phase, signifying slower learning. Additionally, the APP+PS1 rats made significantly more errors following a retention interval, indicating impaired memory compared to both the APP21 and WT rats. Immunohistochemistry using an antibody against amyloid-β (Aβ) showed extensive and mostly diffuse Aβ plaques in the hippocampus and dense plaques that contained tau in the cortex of the brains of the APP+PS1 rats. Furthermore, the APP+PS1 rats also showed vascular changes, including cerebral amyloid angiopathy with extensive Aβ deposits in cortical and leptomeningeal blood vessel walls and venous collagenosis. In addition to the Aβ accumulation observed in arterial, venous, and capillary walls, APP+PS1 rats also displayed enlarged blood vessels and perivascular space. Overall, the brain histopathology and behavioral assessment showed that the APP+PS1 rats demonstrated behavioral characteristics and vascular changes similar to those commonly observed in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Cansu Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Rachel A. Richardson
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Edward G. Stopa
- Department of Pathology, Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Todd R. Schachtman
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Yuksel Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
19
|
Litofsky NS, Miller DC, Chen Z, Simonyi A, Klakotskaia D, Giritharan A, Feng Q, McConnell D, Cui J, Gu Z. Anaemia worsens early functional outcome after traumatic brain injury: a preliminary study. Brain Inj 2018; 32:342-349. [DOI: 10.1080/02699052.2018.1425913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- N Scott Litofsky
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Douglas C Miller
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| | - Zhenzhou Chen
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| | - Agnes Simonyi
- Department of Biochemistry, University of Missouri School of Medicine, Columbia, MO, USA
| | - Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Andrew Giritharan
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| | - Qi Feng
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Diane McConnell
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, MO, USA
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
20
|
Hinrich AJ, Jodelka FM, Chang JL, Brutman D, Bruno AM, Briggs CA, James BD, Stutzmann GE, Bennett DA, Miller SA, Rigo F, Marr RA, Hastings ML. Therapeutic correction of ApoER2 splicing in Alzheimer's disease mice using antisense oligonucleotides. EMBO Mol Med 2017; 8:328-45. [PMID: 26902204 PMCID: PMC4818756 DOI: 10.15252/emmm.201505846] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Apolipoprotein E receptor 2 (ApoER2) is an apolipoprotein E receptor involved in long‐term potentiation, learning, and memory. Given its role in cognition and its association with the Alzheimer's disease (AD) risk gene, apoE, ApoER2 has been proposed to be involved in AD, though a role for the receptor in the disease is not clear. ApoER2 signaling requires amino acids encoded by alternatively spliced exon 19. Here, we report that the balance of ApoER2 exon 19 splicing is deregulated in postmortem brain tissue from AD patients and in a transgenic mouse model of AD. To test the role of deregulated ApoER2 splicing in AD, we designed an antisense oligonucleotide (ASO) that increases exon 19 splicing. Treatment of AD mice with a single dose of ASO corrected ApoER2 splicing for up to 6 months and improved synaptic function and learning and memory. These results reveal an association between ApoER2 isoform expression and AD, and provide preclinical evidence for the utility of ASOs as a therapeutic approach to mitigate Alzheimer's disease symptoms by improving ApoER2 exon 19 splicing.
Collapse
Affiliation(s)
- Anthony J Hinrich
- Department of Cell Biology and Anatomy, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Francine M Jodelka
- Department of Cell Biology and Anatomy, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Jennifer L Chang
- Department of Cell Biology and Anatomy, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Daniella Brutman
- Department of Biology, Lake Forest College, Lake Forest, IL, USA
| | - Angela M Bruno
- Department of Neuroscience, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Clark A Briggs
- Department of Neuroscience, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Bryan D James
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Grace E Stutzmann
- Department of Neuroscience, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Steven A Miller
- Department of Psychology, College of Health Professions Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Robert A Marr
- Department of Neuroscience, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Michelle L Hastings
- Department of Cell Biology and Anatomy, Chicago Medical School Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
21
|
Depletion of coagulation factor XII ameliorates brain pathology and cognitive impairment in Alzheimer disease mice. Blood 2017; 129:2547-2556. [PMID: 28242605 DOI: 10.1182/blood-2016-11-753202] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/21/2017] [Indexed: 12/21/2022] Open
Abstract
Vascular abnormalities and inflammation are found in many Alzheimer disease (AD) patients, but whether these changes play a causative role in AD is not clear. The factor XII (FXII) -initiated contact system can trigger both vascular pathology and inflammation and is activated in AD patients and AD mice. We have investigated the role of the contact system in AD pathogenesis. Cleavage of high-molecular-weight kininogen (HK), a marker for activation of the inflammatory arm of the contact system, is increased in a mouse model of AD, and this cleavage is temporally correlated with the onset of brain inflammation. Depletion of FXII in AD mice inhibited HK cleavage in plasma and reduced neuroinflammation, fibrinogen deposition, and neurodegeneration in the brain. Moreover, FXII-depleted AD mice showed better cognitive function than untreated AD mice. These results indicate that FXII-mediated contact system activation contributes to AD pathogenesis, and therefore this system may offer novel targets for AD treatment.
Collapse
|
22
|
Olver TD, McDonald MW, Klakotskaia D, Richardson RA, Jasperse JL, Melling CWJ, Schachtman TR, Yang HT, Emter CA, Laughlin MH. A chronic physical activity treatment in obese rats normalizes the contributions of ET-1 and NO to insulin-mediated posterior cerebral artery vasodilation. J Appl Physiol (1985) 2017; 122:1040-1050. [PMID: 28183819 DOI: 10.1152/japplphysiol.00811.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/11/2017] [Accepted: 02/02/2017] [Indexed: 12/31/2022] Open
Abstract
This study tested the hypotheses that obesity-induced decrements in insulin-stimulated cerebrovascular vasodilation would be normalized with acute endothelin-1a receptor antagonism and that treatment with a physical activity intervention restores vasoreactivity to insulin through augmented nitric oxide synthase (NOS)-dependent dilation. Otsuka Long-Evans Tokushima Fatty rats were divided into the following groups: 20 wk old food controlled (CON-20); 20 wk old free food access (model of obesity, OB-20); 40 wk old food controlled (CON-40); 40 wk old free food access (OB-40); and 40 wk old free food access+RUN (RUN-40; wheel-running access from 20 to 40 wk). Rats underwent Barnes maze testing and a euglycemic hyperinsulinemic clamp (EHC). In the 40-wk cohort, cerebellum and hippocampus blood flow (BF) were examined (microsphere infusion). Vasomotor responses (pressurized myography) to insulin were assessed in untreated, endothelin-1a receptor antagonism, and NOS inhibition conditions in posterior cerebral arteries. Insulin-stimulated vasodilation was attenuated in the OB vs. CON and RUN groups (P ≤ 0.04). Dilation to insulin was normalized with endothelin-1a receptor antagonism in the OB groups (between groups, P ≥ 0.56), and insulin-stimulated NOS-mediated dilation was greater in the RUN-40 vs. OB-40 group (P < 0.01). At 40 wk of age, cerebellum BF decreased during EHC in the OB-40 group (P = 0.02) but not CON or RUN groups (P ≥ 0.36). Barnes maze testing revealed increased entry errors and latencies in the RUN-40 vs. CON and OB groups (P < 0.01). These findings indicate that obesity-induced impairments in vasoreactivity to insulin involve increased endothelin-1 and decreased nitric oxide signaling. Chronic spontaneous physical activity, initiated after disease onset, reversed impaired vasodilation to insulin and decreased Barnes maze performance, possibly because of increased exploratory behavior.NEW & NOTEWORTHY The new and noteworthy findings are that 1) in rodents, obesity-related deficits in insulin-mediated vasodilation are associated with increased influence of insulin-stimulated ET-1 and depressed influence of insulin-stimulated NOS and 2) a physical activity intervention, initiated after the onset of disease, restores insulin-mediated vasodilation, likely by normalizing insulin-stimulated ET-1 and NOS balance. These data demonstrate that the treatment effects of chronic exercise on insulin-mediated vasodilation extend beyond active skeletal muscle vasculature and include the cerebrovasculature.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri;
| | - Matthew W McDonald
- Department of Kinesiology, University of Western Ontario, London, Ontario, Canada
| | - Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri; and
| | - Rachel A Richardson
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri; and
| | | | - C W James Melling
- Department of Kinesiology, University of Western Ontario, London, Ontario, Canada
| | - Todd R Schachtman
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri; and
| | - Hsiao T Yang
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
23
|
Translational Assays for Assessment of Cognition in Rodent Models of Alzheimer’s Disease and Dementia. J Mol Neurosci 2016; 60:371-382. [DOI: 10.1007/s12031-016-0837-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023]
|
24
|
Chung YC, Kruyer A, Yao Y, Feierman E, Richards A, Strickland S, Norris EH. Hyperhomocysteinemia exacerbates Alzheimer's disease pathology by way of the β-amyloid fibrinogen interaction. J Thromb Haemost 2016; 14:1442-52. [PMID: 27090576 PMCID: PMC4949110 DOI: 10.1111/jth.13340] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/31/2016] [Indexed: 01/02/2023]
Abstract
UNLABELLED Essentials Evidence suggests a comorbidity between hyperhomocysteinemia (HHC) and Alzheimer's disease (AD). Homocysteine (HC) could affect the β-amyloid (Aβ)-fibrinogen interaction in AD pathology. AD patients with concomitant HHC have increased fibrin and Aβ deposits in their brains. HC contributes to AD pathology via the Aβ-fibrinogen interaction. SUMMARY Background Accumulating clinical evidence suggests that hyperhomocysteinemia (HHC) is correlated with Alzheimer's disease (AD) and vascular dementia. Objective This study was carried out to elucidate the specific role of elevated homocysteine (HC) levels in AD pathophysiology. Methods Immunohistochemistry was used to examine β-amyloid (Aβ) deposition along blood vessels, also known as cerebral amyloid angiopathy (CAA), fibrin(ogen) deposition, and their correlation to each other in the brains of AD patients with and without HHC. To study AD-HHC co-morbidity in detail, an AD mouse model was administered a high methionine diet for several months. Parenchymal Aβ plaques, CAA-positive vessels and fibrin deposits were then assessed by immunohistochemistry at different stages of AD progression. Memory deficits were evaluated with contextual fear conditioning and the Barnes maze. Additionally, the effect of HC and its metabolite, homocysteine thiolactone (HCTL), on the Aβ-fibrinogen interaction was analyzed by pull-down, ELISA and fibrin clot formation and fibrinolysis assays in vitro. Results We found increased fibrin(ogen) levels and Aβ deposits in the blood vessels and brain parenchyma of AD patients with HHC. We demonstrate that HC and HCTL enhance the interaction between fibrinogen and Aβ, promote the formation of tighter fibrin clots and delay clot fibrinolysis. Additionally, we show that diet-induced HHC in an AD mouse model leads to severe CAA and parenchymal Aβ deposition, as well as significant impairments in learning and memory. Conclusions These findings suggest that elevated levels of plasma HC/HCTL contribute to AD pathology via the Aβ-fibrin(ogen) interaction.
Collapse
Affiliation(s)
- Young Cheul Chung
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Anna Kruyer
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Yao Yao
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Emily Feierman
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Allison Richards
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
25
|
Granger MW, Franko B, Taylor MW, Messier C, George-Hyslop PS, Bennett SA. A TgCRND8 Mouse Model of Alzheimer’s Disease Exhibits Sexual Dimorphisms in Behavioral Indices of Cognitive Reserve. J Alzheimers Dis 2016; 51:757-73. [DOI: 10.3233/jad-150587] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Matthew W. Granger
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Bettina Franko
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew W. Taylor
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Claude Messier
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
| | - Peter St. George-Hyslop
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK and Tanz Centre for Research in Neurodegenerative Diseases University of Toronto, Toronto, ON, Canada
| | - Steffany A.L. Bennett
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
26
|
Audrain M, Fol R, Dutar P, Potier B, Billard JM, Flament J, Alves S, Burlot MA, Dufayet-Chaffaud G, Bemelmans AP, Valette J, Hantraye P, Déglon N, Cartier N, Braudeau J. Alzheimer's disease-like APP processing in wild-type mice identifies synaptic defects as initial steps of disease progression. Mol Neurodegener 2016; 11:5. [PMID: 26759118 PMCID: PMC4709894 DOI: 10.1186/s13024-016-0070-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/06/2016] [Indexed: 12/15/2022] Open
Abstract
Background Alzheimer’s disease (AD) is the most frequent form of dementia in the elderly and no effective treatment is currently available. The mechanisms triggering AD onset and progression are still imperfectly dissected. We aimed at deciphering the modifications occurring in vivo during the very early stages of AD, before the development of amyloid deposits, neurofibrillary tangles, neuronal death and inflammation. Most current AD models based on Amyloid Precursor Protein (APP) overproduction beginning from in utero, to rapidly reproduce the histological and behavioral features of the disease within a few months, are not appropriate to study the early steps of AD development. As a means to mimic in vivo amyloid APP processing closer to the human situation in AD, we used an adeno-associated virus (AAV)-based transfer of human mutant APP and Presenilin 1 (PS1) genes to the hippocampi of two-month-old C57Bl/6 J mice to express human APP, without significant overexpression and to specifically induce its amyloid processing. Results The human APP, βCTF and Aβ42/40 ratio were similar to those in hippocampal tissues from AD patients. Three months after injection the murine Tau protein was hyperphosphorylated and rapid synaptic failure occurred characterized by decreased levels of both PSD-95 and metabolites related to neuromodulation, on proton magnetic resonance spectroscopy (1H-MRS). Astrocytic GLT-1 transporter levels were lower and the tonic glutamatergic current was stronger on electrophysiological recordings of CA1 hippocampal region, revealing the overstimulation of extrasynaptic N-methyl D-aspartate receptor (NMDAR) which precedes the loss of long-term potentiation (LTP). These modifications were associated with early behavioral impairments in the Open-field, Y-maze and Morris Mater Maze tasks. Conclusions Altogether, this demonstrates that an AD-like APP processing, yielding to levels of APP, βCTF and Aβ42/Aβ40 ratio similar to those observed in AD patients, are sufficient to rapidly trigger early steps of the amyloidogenic and Tau pathways in vivo. With this strategy, we identified a sequence of early events likely to account for disease onset and described a model that may facilitate efforts to decipher the factors triggering AD and to evaluate early neuroprotective strategies. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0070-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mickael Audrain
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France.,Université Paris Descartes, Paris, France.,CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France
| | - Romain Fol
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France.,Université Paris Descartes, Paris, France.,CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France
| | - Patrick Dutar
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Brigitte Potier
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Marie Billard
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Julien Flament
- CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France.,INSERM UMS27, Fontenay-aux-Roses 92265, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France
| | - Sandro Alves
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France.,CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France
| | - Marie-Anne Burlot
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France.,Université Paris Descartes, Paris, France.,CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France
| | - Gaelle Dufayet-Chaffaud
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France.,CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France
| | - Alexis-Pierre Bemelmans
- CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France.,CNRS UMR9199, Fontenay-aux-Roses 92265, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France
| | - Julien Valette
- CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France.,CNRS UMR9199, Fontenay-aux-Roses 92265, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France
| | - Philippe Hantraye
- CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France.,INSERM UMS27, Fontenay-aux-Roses 92265, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France.,CNRS UMR9199, Fontenay-aux-Roses 92265, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France
| | - Nicole Déglon
- Department of Clinical Neurosciences, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital, Lausanne, Switzerland.,Neuroscience Research Center, Laboratory of Cellular and Molecular Neurotherapies, Lausanne University Hospital, Lausanne, Switzerland
| | - Nathalie Cartier
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France. .,CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France.
| | - Jérome Braudeau
- INSERM UMR1169, Université Paris-Sud, Université Paris-Saclay, Orsay, 94100, France.,CEA, DSV, I2BM, MIRCen, Fontenay-aux-Roses, 92265, France
| |
Collapse
|
27
|
Lee M, Chen Z, Tomlinson BN, Gooyit M, Hesek D, Juárez MR, Nizam R, Boggess B, Lastochkin E, Schroeder VA, Wolter WR, Suckow MA, Cui J, Mobashery S, Gu Z, Chang M. Water-Soluble MMP-9 Inhibitor Reduces Lesion Volume after Severe Traumatic Brain Injury. ACS Chem Neurosci 2015; 6:1658-64. [PMID: 26241578 DOI: 10.1021/acschemneuro.5b00140] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
SB-3CT is a potent and selective inhibitor of matrix metalloproteinase (MMP)-2 and -9, which has shown efficacy in an animal model of severe traumatic brain injury (TBI). However, SB-3CT is poorly water-soluble and is metabolized primarily to p-hydroxy SB-3CT (2), a more potent inhibitor than SB-3CT. We synthesized the O-phosphate prodrug (3) of compound 2 to enhance its water solubility by more than 2000-fold. The prodrug 3 was a poor MMP inhibitor, but readily hydrolyzed to the active 2 in human blood. Pharmacokinetics and brain distribution studies in mice showed that 2 crossed the blood-brain barrier (BBB) and achieved therapeutic concentrations in the brain. The prodrug 3/compound 2 was evaluated in a mouse model of severe TBI and found to significantly decrease the brain lesion volume and improve neurological outcomes. MMP-9 inhibition by a water-soluble thiirane inhibitor is a promising therapy for treatment of TBI.
Collapse
Affiliation(s)
| | - Zhenzhou Chen
- Department of Pathology and Anatomical Sciences and Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Brittany N. Tomlinson
- Department of Pathology and Anatomical Sciences and Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | | | | | - María Raquel Juárez
- Department of Pathology and Anatomical Sciences and Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Rasheeq Nizam
- Department of Pathology and Anatomical Sciences and Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | | | | | | | | | | | - Jiancun Cui
- Department of Pathology and Anatomical Sciences and Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | | | - Zezong Gu
- Department of Pathology and Anatomical Sciences and Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | | |
Collapse
|
28
|
Salgado-Puga K, Prado-Alcalá RA, Peña-Ortega F. Amyloid β Enhances Typical Rodent Behavior While It Impairs Contextual Memory Consolidation. Behav Neurol 2015; 2015:526912. [PMID: 26229236 PMCID: PMC4502279 DOI: 10.1155/2015/526912] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/04/2015] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is associated with an early hippocampal dysfunction, which is likely induced by an increase in soluble amyloid beta peptide (Aβ). This hippocampal failure contributes to the initial memory deficits observed both in patients and in AD animal models and possibly to the deterioration in activities of daily living (ADL). One typical rodent behavior that has been proposed as a hippocampus-dependent assessment model of ADL in mice and rats is burrowing. Despite the fact that AD transgenic mice show some evidence of reduced burrowing, it has not been yet determined whether or not Aβ can affect this typical rodent behavior and whether this alteration correlates with the well-known Aβ-induced memory impairment. Thus, the purpose of this study was to test whether or not Aβ affects burrowing while inducing hippocampus-dependent memory impairment. Surprisingly, our results show that intrahippocampal application of Aβ increases burrowing while inducing memory impairment. We consider that this Aβ-induced increase in burrowing might be associated with a mild anxiety state, which was revealed by increased freezing behavior in the open field, and conclude that Aβ-induced hippocampal dysfunction is reflected in the impairment of ADL and memory, through mechanisms yet to be determined.
Collapse
Affiliation(s)
- Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, 76230 Juriquilla, Querétaro, QRO, Mexico
| | - Roberto A. Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, 76230 Juriquilla, Querétaro, QRO, Mexico
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, 76230 Juriquilla, Querétaro, QRO, Mexico
| |
Collapse
|
29
|
Barker-Haliski ML, Friedman D, French JA, White HS. Disease Modification in Epilepsy: From Animal Models to Clinical Applications. Drugs 2015; 75:749-67. [DOI: 10.1007/s40265-015-0395-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Quarta E, Bravi R, Scambi I, Mariotti R, Minciacchi D. Increased anxiety-like behavior and selective learning impairments are concomitant to loss of hippocampal interneurons in the presymptomatic SOD1(G93A) ALS mouse model. J Comp Neurol 2015; 523:1622-38. [DOI: 10.1002/cne.23759] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Eros Quarta
- Department of Experimental and Clinical Medicine, Physiological Sciences Section; University of Florence; Florence I-50134 Italy
| | - Riccardo Bravi
- Department of Experimental and Clinical Medicine, Physiological Sciences Section; University of Florence; Florence I-50134 Italy
| | - Ilaria Scambi
- Department of Neurological and Movement Sciences; University of Verona; Verona I-37134 Italy
| | - Raffaella Mariotti
- Department of Neurological and Movement Sciences; University of Verona; Verona I-37134 Italy
| | - Diego Minciacchi
- Department of Experimental and Clinical Medicine, Physiological Sciences Section; University of Florence; Florence I-50134 Italy
| |
Collapse
|
31
|
Memory formation and retention are affected in adult miR-132/212 knockout mice. Behav Brain Res 2015; 287:15-26. [PMID: 25813747 DOI: 10.1016/j.bbr.2015.03.032] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/11/2015] [Accepted: 03/16/2015] [Indexed: 01/16/2023]
Abstract
The miR-132/212 family is thought to play an important role in neural function and plasticity, while its misregulation has been observed in various neurodegenerative disorders. In this study, we analyzed 6-month-old miR-132/212 knockout mice in a battery of cognitive and non-cognitive behavioral tests. No significant changes were observed in reflexes and basic sensorimotor functions as determined by the SHIRPA primary screen. Accordingly, miR-132/212 knockout mice did not differ from wild-type controls in general locomotor activity in an open-field test. Furthermore, no significant changes of anxiety were measured in an elevated plus maze task. However, the mutant mice showed retention phase defects in a novel object recognition test and in the T-water maze. Moreover, the learning and probe phases in the Barnes maze were clearly altered in knockout mice when compared to controls. Finally, changes in BDNF, CREB, and MeCP2 were identified in the miR-132/212-deficient mice, providing a potential mechanism for promoting memory loss. Taken together, these results further strengthen the role of miR-132/212 in memory formation and retention, and shed light on the potential consequences of its deregulation in neurodegenerative diseases.
Collapse
|
32
|
Barker-Haliski ML, Dahle EJ, Heck TD, Pruess TH, Vanegas F, Wilcox KS, White HS. Evaluating an etiologically relevant platform for therapy development for temporal lobe epilepsy: effects of carbamazepine and valproic acid on acute seizures and chronic behavioral comorbidities in the Theiler's murine encephalomyelitis virus mouse model. J Pharmacol Exp Ther 2015; 353:318-29. [PMID: 25755209 DOI: 10.1124/jpet.114.222513] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Central nervous system infections can underlie the development of epilepsy, and Theiler's murine encephalomyelitis virus (TMEV) infection in C57BL/6J mice provides a novel model of infection-induced epilepsy. Approximately 50-65% of infected mice develop acute, handling-induced seizures during the infection. Brains display acute neuropathology, and a high number of mice develop spontaneous, recurrent seizures and behavioral comorbidities weeks later. This study characterized the utility of this model for drug testing by assessing whether antiseizure drug treatment during the acute infection period attenuates handling-induced seizures, and whether such treatment modifies associated comorbidities. Male C57BL/6J mice infected with TMEV received twice-daily valproic acid (VPA; 200 mg/kg), carbamazepine (CBZ; 20 mg/kg), or vehicle during the infection (days 0-7). Mice were assessed twice daily during the infection period for handling-induced seizures. Relative to vehicle-treated mice, more CBZ-treated mice presented with acute seizures; VPA conferred no change. In mice displaying seizures, VPA, but not CBZ, reduced seizure burden. Animals were then randomly assigned to acute and long-term follow-up. VPA was associated with significant elevations in acute (day 8) glial fibrillary acidic protein (astrocytes) immunoreactivity, but did not affect NeuN (neurons) immunoreactivity. Additionally, VPA-treated mice showed improved motor performance 15 days postinfection (DPI). At 36 DPI, CBZ-treated mice traveled significantly less distance through the center of an open field, indicative of anxiety-like behavior. CBZ-treated mice also presented with significant astrogliosis 36 DPI. Neither CBZ nor VPA prevented long-term reductions in NeuN immunoreactivity. The TMEV model thus provides an etiologically relevant platform to evaluate potential treatments for acute seizures and disease modification.
Collapse
Affiliation(s)
- Melissa L Barker-Haliski
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - E Jill Dahle
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - Taylor D Heck
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - Timothy H Pruess
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - Fabiola Vanegas
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - Karen S Wilcox
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - H Steve White
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
33
|
Rial D, Castro AA, Machado N, Garção P, Gonçalves FQ, Silva HB, Tomé ÂR, Köfalvi A, Corti O, Raisman-Vozari R, Cunha RA, Prediger RD. Behavioral phenotyping of Parkin-deficient mice: looking for early preclinical features of Parkinson's disease. PLoS One 2014; 9:e114216. [PMID: 25486126 PMCID: PMC4259468 DOI: 10.1371/journal.pone.0114216] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/04/2014] [Indexed: 01/24/2023] Open
Abstract
There is considerable evidence showing that the neurodegenerative processes that lead to sporadic Parkinson's disease (PD) begin many years before the appearance of the characteristic motor symptoms. Neuropsychiatric, sensorial and cognitive deficits are recognized as early non-motor manifestations of PD, and are not attenuated by the current anti-parkinsonian therapy. Although loss-of-function mutations in the parkin gene cause early-onset familial PD, Parkin-deficient mice do not display spontaneous degeneration of the nigrostriatal pathway or enhanced vulnerability to dopaminergic neurotoxins such as 6-OHDA and MPTP. Here, we employed adult homozygous C57BL/6 mice with parkin gene deletion on exon 3 (parkin−/−) to further investigate the relevance of Parkin in the regulation of non-motor features, namely olfactory, emotional, cognitive and hippocampal synaptic plasticity. Parkin−/− mice displayed normal performance on behavioral tests evaluating olfaction (olfactory discrimination), anxiety (elevated plus-maze), depressive-like behavior (forced swimming and tail suspension) and motor function (rotarod, grasping strength and pole). However, parkin−/− mice displayed a poor performance in the open field habituation, object location and modified Y-maze tasks suggestive of procedural and short-term spatial memory deficits. These behavioral impairments were accompanied by impaired hippocampal long-term potentiation (LTP). These findings indicate that the genetic deletion of parkin causes deficiencies in hippocampal synaptic plasticity, resulting in memory deficits with no major olfactory, emotional or motor impairments. Therefore, parkin−/− mice may represent a promising animal model to study the early stages of PD and for testing new therapeutic strategies to restore learning and memory and synaptic plasticity impairments in PD.
Collapse
Affiliation(s)
- Daniel Rial
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, UFSC, Florianópolis, 88049-900, SC, Brazil
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Adalberto A. Castro
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, UFSC, Florianópolis, 88049-900, SC, Brazil
| | - Nuno Machado
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Pedro Garção
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Francisco Q. Gonçalves
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Henrique B. Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Ângelo R. Tomé
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Attila Köfalvi
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Olga Corti
- CNRS UMR 7225, Hôpital de la Salpêtrière—Bâtiment, ICM (Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière), CRICM, Thérapeutique Expérimentale de la Neurodégénérescence, Université Pierre et Marie Curie, UPMC, 75651, Paris, France
| | - Rita Raisman-Vozari
- CNRS UMR 7225, Hôpital de la Salpêtrière—Bâtiment, ICM (Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière), CRICM, Thérapeutique Expérimentale de la Neurodégénérescence, Université Pierre et Marie Curie, UPMC, 75651, Paris, France
| | - Rodrigo A. Cunha
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3005-504, Coimbra, Portugal
| | - Rui D. Prediger
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, UFSC, Florianópolis, 88049-900, SC, Brazil
- * E-mail:
| |
Collapse
|
34
|
Lysenko LV, Kim J, Henry C, Tyrtyshnaia A, Kohnz RA, Madamba F, Simon GM, Kleschevnikova NE, Nomura DK, Ezekowitz R.AB, Kleschevnikov AM. Monoacylglycerol lipase inhibitor JZL184 improves behavior and neural properties in Ts65Dn mice, a model of down syndrome. PLoS One 2014; 9:e114521. [PMID: 25474204 PMCID: PMC4256450 DOI: 10.1371/journal.pone.0114521] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/10/2014] [Indexed: 12/13/2022] Open
Abstract
Genetic alterations or pharmacological treatments affecting endocannabinoid signaling have profound effects on synaptic and neuronal properties and, under certain conditions, may improve higher brain functions. Down syndrome (DS), a developmental disorder caused by triplication of chromosome 21, is characterized by deficient cognition and inevitable development of the Alzheimer disease (AD) type pathology during aging. Here we used JZL184, a selective inhibitor of monoacylglycerol lipase (MAGL), to examine the effects of chronic MAGL inhibition on the behavioral, biochemical, and synaptic properties of aged Ts65Dn mice, a genetic model of DS. In both Ts65Dn mice and their normosomic (2N) controls, JZL184-treatment increased brain levels of 2-arachidonoylglycerol (2-AG) and decreased levels of its metabolites such as arachidonic acid, prostaglandins PGD2, PGE2, PGFα, and PGJ2. Enhanced spontaneous locomotor activity of Ts65Dn mice was reduced by the JZL184-treatement to the levels observed in 2N animals. Deficient long-term memory was also improved, while short-term and working types of memory were unaffected. Furthermore, reduced hippocampal long-term potentiation (LTP) was increased in the JZL184-treated Ts65Dn mice to the levels observed in 2N mice. Interestingly, changes in synaptic plasticity and behavior were not observed in the JZL184-treated 2N mice suggesting that the treatment specifically attenuated the defects in the trisomic animals. The JZL184-treatment also reduced the levels of Aβ40 and Aβ42, but had no effect on the levels of full length APP and BACE1 in both Ts65Dn and 2N mice. These data show that chronic MAGL inhibition improves the behavior and brain functions in a DS model suggesting that pharmacological targeting of MAGL may be considered as a perspective new approach for improving cognition in DS.
Collapse
Affiliation(s)
- Larisa V. Lysenko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | - Jeesun Kim
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | - Cassandra Henry
- Abide Therapeutics, Inc., San Diego, CA, United States of America
| | - Anna Tyrtyshnaia
- School of Biomedicine, Far Eastern Federal University, Sukhanova 8, Vladivostok, Russian Federation
| | - Rebecca A. Kohnz
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Francisco Madamba
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | - Gabriel M. Simon
- Abide Therapeutics, Inc., San Diego, CA, United States of America
| | - Natalia E. Kleschevnikova
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
| | - Daniel K. Nomura
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | | | - Alexander M. Kleschevnikov
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States of America
- * E-mail:
| |
Collapse
|
35
|
Abstract
Key neuropathological hallmarks of Alzheimer's disease (AD) are elevated levels of amyloid β-peptide (Aβ) species generated via amyloid precursor protein (APP) endoproteolysis and cleavage by the rate-limiting β-site enzyme 1 (BACE1). Because rodents do not develop amyloid pathologies, we here investigated whether AD-like endophenotypes can be created in mice by expression of human bace1. To avoid pitfalls of existing models, we introduced hbace1 via knock-in under the control of the CaMKII α promoter into the safe HPRT locus. We report amyloidogenic processing of murine APP in the hBACE1 mice (termed PLB4), resulting in the formation of toxic APP metabolites that accumulate intra- and extraneuronally in hippocampus and cortex. Pronounced accumulation of Aβ*56 and Aβ hexamers in the absence of plaque deposition was detected in brain tissue from symptomatic PLB4 mice. Heightened levels of inflammation (gliosis) also appeared in several AD-related brain regions (dentate gyrus, hippocampal area CA1, piriform and parietal cortices) at 6 and 12 months of age. Behaviorally, deficits in habituation to a novel environment and semantic-like memory (social transmission of food preference) were detected from 3 to 4 months of age. Impairments in spatial learning strategies in long-term reference (water maze) and working memory (Y-maze) tasks presented at 6 months, and were distinct from reductions in locomotor activity and anxiety. Overall, our data indicate for the first time that targeted, subtle forebrain-specific expression through single gene knock-in of hBACE1 is sufficient to generate AD-relevant cognitive impairments amid corresponding histopathologies, confirming human BACE as the key parameter in amyloid pathogenesis.
Collapse
|
36
|
Bronfman ZZ, Ginsburg S, Jablonka E. Shaping the learning curve: epigenetic dynamics in neural plasticity. Front Integr Neurosci 2014; 8:55. [PMID: 25071483 PMCID: PMC4083220 DOI: 10.3389/fnint.2014.00055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 06/18/2014] [Indexed: 12/22/2022] Open
Abstract
A key characteristic of learning and neural plasticity is state-dependent acquisition dynamics reflected by the non-linear learning curve that links increase in learning with practice. Here we propose that the manner by which epigenetic states of individual cells change during learning contributes to the shape of the neural and behavioral learning curve. We base our suggestion on recent studies showing that epigenetic mechanisms such as DNA methylation, histone acetylation, and RNA-mediated gene regulation are intimately involved in the establishment and maintenance of long-term neural plasticity, reflecting specific learning-histories and influencing future learning. Our model, which is the first to suggest a dynamic molecular account of the shape of the learning curve, leads to several testable predictions regarding the link between epigenetic dynamics at the promoter, gene-network, and neural-network levels. This perspective opens up new avenues for therapeutic interventions in neurological pathologies.
Collapse
Affiliation(s)
- Zohar Z Bronfman
- The Cohn Institute for the History and Philosophy of Science and Ideas, Faculty of Humanities, Tel-Aviv University Tel-Aviv, Israel ; School of Psychology, Faculty of Social Science, Tel-Aviv University Tel-Aviv, Israel
| | - Simona Ginsburg
- Natural Science Department, The Open University of Israel Raanana, Israel
| | - Eva Jablonka
- The Cohn Institute for the History and Philosophy of Science and Ideas, Faculty of Humanities, Tel-Aviv University Tel-Aviv, Israel
| |
Collapse
|
37
|
Schneider F, Baldauf K, Wetzel W, Reymann KG. Behavioral and EEG changes in male 5xFAD mice. Physiol Behav 2014; 135:25-33. [PMID: 24907698 DOI: 10.1016/j.physbeh.2014.05.041] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/23/2014] [Accepted: 05/28/2014] [Indexed: 11/18/2022]
Abstract
Transgenic animal models of Alzheimer's disease (AD) are widely used to investigate mechanisms of pathophysiology and cognitive dysfunctions. A model with a very early development of parenchymal plaque load at the age of 2months is the 5xFAD mouse (Tg6799, Oakley et al. 2006). These 5xFAD mice over-express both human amyloid precursor protein (APP) and human presenilin 1 (PS1). Mice from this line have a high APP expression correlating with a high burden and an accelerated accumulation of the 42 amino acid species of amyloid-β (Aβ). The aim of this study was the behavioral and functional investigations of 5xFAD males because in most studies females of this strain were characterized. In comparison to literature of transgenic 5xFAD females, transgenic 5xFAD males showed decreased anxiety in the elevated plus maze, reduced locomotion and exploration in the open field and disturbances in learning performance in the Morris water maze starting at 9months of age. Electroencephalogram (EEG) recordings on 6month old transgenic mice revealed a decrease of delta, theta, alpha, beta and gamma frequency bands whereas the subdelta frequency was increased. EEG recordings during sleep showed a reduction of rapid eye movement sleep in relation to the amount of total sleep. Thus, 5xFAD males develop early functional disturbances and subsequently behavioral deficits and therefore they are a good mouse model for studying Alzheimer's disease.
Collapse
Affiliation(s)
- F Schneider
- German Centre for Neurodegenerative Diseases (DZNE), D-39120 Magdeburg, Germany.
| | - K Baldauf
- German Centre for Neurodegenerative Diseases (DZNE), D-39120 Magdeburg, Germany.
| | - W Wetzel
- Leibniz Institute for Neurobiology, Brenneckestr. 6, D-39118 Magdeburg, Germany.
| | - K G Reymann
- German Centre for Neurodegenerative Diseases (DZNE), D-39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, D-39118 Magdeburg, Germany.
| |
Collapse
|
38
|
Ahn HJ, Glickman JF, Poon KL, Zamolodchikov D, Jno-Charles OC, Norris EH, Strickland S. A novel Aβ-fibrinogen interaction inhibitor rescues altered thrombosis and cognitive decline in Alzheimer's disease mice. ACTA ACUST UNITED AC 2014; 211:1049-62. [PMID: 24821909 PMCID: PMC4042638 DOI: 10.1084/jem.20131751] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Pharmacological disruption of the interaction between fibrinogen and β-amyloid reduces vascular amyloid deposition and improves cognition in a mouse model of Alzheimer’s disease. Many Alzheimer’s disease (AD) patients suffer from cerebrovascular abnormalities such as altered cerebral blood flow and cerebral microinfarcts. Recently, fibrinogen has been identified as a strong cerebrovascular risk factor in AD, as it specifically binds to β-amyloid (Aβ), thereby altering fibrin clot structure and delaying clot degradation. To determine if the Aβ–fibrinogen interaction could be targeted as a potential new treatment for AD, we designed a high-throughput screen and identified RU-505 as an effective inhibitor of the Aβ–fibrinogen interaction. RU-505 restored Aβ-induced altered fibrin clot formation and degradation in vitro and inhibited vessel occlusion in AD transgenic mice. Furthermore, long-term treatment of RU-505 significantly reduced vascular amyloid deposition and microgliosis in the cortex and improved cognitive impairment in mouse models of AD. Our studies suggest that inhibitors targeting the Aβ–fibrinogen interaction show promise as therapy for treating AD.
Collapse
Affiliation(s)
- Hyung Jin Ahn
- Laboratory of Neurobiology and Genetics and High Throughput Screening Resource Center, The Rockefeller University, New York, NY 10065
| | - J Fraser Glickman
- Laboratory of Neurobiology and Genetics and High Throughput Screening Resource Center, The Rockefeller University, New York, NY 10065
| | - Ka Lai Poon
- Laboratory of Neurobiology and Genetics and High Throughput Screening Resource Center, The Rockefeller University, New York, NY 10065
| | - Daria Zamolodchikov
- Laboratory of Neurobiology and Genetics and High Throughput Screening Resource Center, The Rockefeller University, New York, NY 10065
| | - Odella C Jno-Charles
- Laboratory of Neurobiology and Genetics and High Throughput Screening Resource Center, The Rockefeller University, New York, NY 10065
| | - Erin H Norris
- Laboratory of Neurobiology and Genetics and High Throughput Screening Resource Center, The Rockefeller University, New York, NY 10065
| | - Sidney Strickland
- Laboratory of Neurobiology and Genetics and High Throughput Screening Resource Center, The Rockefeller University, New York, NY 10065
| |
Collapse
|
39
|
Bernstein KE, Koronyo Y, Salumbides BC, Sheyn J, Pelissier L, Lopes DHJ, Shah KH, Bernstein EA, Fuchs DT, Yu JJY, Pham M, Black KL, Shen XZ, Fuchs S, Koronyo-Hamaoui M. Angiotensin-converting enzyme overexpression in myelomonocytes prevents Alzheimer's-like cognitive decline. J Clin Invest 2014; 124:1000-12. [PMID: 24487585 DOI: 10.1172/jci66541] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 11/18/2013] [Indexed: 11/17/2022] Open
Abstract
Cognitive decline in patients with Alzheimer's disease (AD) is associated with elevated brain levels of amyloid β protein (Aβ), particularly neurotoxic Aβ(1-42). Angiotensin-converting enzyme (ACE) can degrade Aβ(1-42), and ACE overexpression in myelomonocytic cells enhances their immune function. To examine the effect of targeted ACE overexpression on AD, we crossed ACE(10/10) mice, which overexpress ACE in myelomonocytes using the c-fms promoter, with the transgenic APP(SWE)/PS1(ΔE9) mouse model of AD (AD⁺). Evaluation of brain tissue from these AD⁺ACE(10/10) mice at 7 and 13 months revealed that levels of both soluble and insoluble brain Aβ(1-42) were reduced compared with those in AD⁺ mice. Furthermore, both plaque burden and astrogliosis were drastically reduced. Administration of the ACE inhibitor ramipril increased Aβ levels in AD⁺ACE(10/10) mice compared with the levels induced by the ACE-independent vasodilator hydralazine. Overall, AD⁺ACE(10/10) mice had less brain-infiltrating cells, consistent with reduced AD-associated pathology, though ACE-overexpressing macrophages were abundant around and engulfing Aβ plaques. At 11 and 12 months of age, the AD⁺ACE(10/WT) and AD⁺ACE(10/10) mice were virtually equivalent to non-AD mice in cognitive ability, as assessed by maze-based behavioral tests. Our data demonstrate that an enhanced immune response, coupled with increased myelomonocytic expression of catalytically active ACE, prevents cognitive decline in a murine model of AD.
Collapse
|
40
|
Rosenfeld CS, Ferguson SA. Barnes maze testing strategies with small and large rodent models. J Vis Exp 2014:e51194. [PMID: 24637673 DOI: 10.3791/51194] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Spatial learning and memory of laboratory rodents is often assessed via navigational ability in mazes, most popular of which are the water and dry-land (Barnes) mazes. Improved performance over sessions or trials is thought to reflect learning and memory of the escape cage/platform location. Considered less stressful than water mazes, the Barnes maze is a relatively simple design of a circular platform top with several holes equally spaced around the perimeter edge. All but one of the holes are false-bottomed or blind-ending, while one leads to an escape cage. Mildly aversive stimuli (e.g. bright overhead lights) provide motivation to locate the escape cage. Latency to locate the escape cage can be measured during the session; however, additional endpoints typically require video recording. From those video recordings, use of automated tracking software can generate a variety of endpoints that are similar to those produced in water mazes (e.g. distance traveled, velocity/speed, time spent in the correct quadrant, time spent moving/resting, and confirmation of latency). Type of search strategy (i.e. random, serial, or direct) can be categorized as well. Barnes maze construction and testing methodologies can differ for small rodents, such as mice, and large rodents, such as rats. For example, while extra-maze cues are effective for rats, smaller wild rodents may require intra-maze cues with a visual barrier around the maze. Appropriate stimuli must be identified which motivate the rodent to locate the escape cage. Both Barnes and water mazes can be time consuming as 4-7 test trials are typically required to detect improved learning and memory performance (e.g. shorter latencies or path lengths to locate the escape platform or cage) and/or differences between experimental groups. Even so, the Barnes maze is a widely employed behavioral assessment measuring spatial navigational abilities and their potential disruption by genetic, neurobehavioral manipulations, or drug/ toxicant exposure.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Biomedical Sciences and Bond Life Sciences Center, University of Missouri;
| | - Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration;
| |
Collapse
|
41
|
Dao AT, Zagaar MA, Levine AT, Salim S, Eriksen JL, Alkadhi KA. Treadmill exercise prevents learning and memory impairment in Alzheimer's disease-like pathology. Curr Alzheimer Res 2014; 10:507-15. [PMID: 23627709 DOI: 10.2174/1567205011310050006] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by progressive memory loss. In contrast, accumulating evidence suggests a neuroprotective role of regular exercise in aging associated memory impairment. In this study, we investigated the ability of regular exercise to prevent impairments of short-term memory (STM) and early long-term potentiation (E-LTP) in area CA1 of the hippocampus in a rat model of AD (i.c.v. infusion of 250 pmol/day Aβ1-42 peptides). We utilized behavioral assessment, in vivo electrophysiological recording, and immunoblotting in 4 groups of adult Wistar rats: control, treadmill exercise (Ex), β-amyloid-infused (Aβ), and amyloid-infused/treadmill exercised (Ex/Aβ). Our findings indicated that Aβ rats made significantly more errors in the radial arm water maze (RAWM) compared to all other groups and exhibited suppressed E-LTP in area CA1, which correlated with deleterious alterations in the levels of memory and E-LTP-related signaling molecules including calcineurin (PP2B), brain derivedneurotrophic factor (BDNF) and phosphorylated CaMKII (p-CaMKII). Compared to controls, Ex and Ex/Aβ rats showed a similar behavioral performance and a normal E-LTP with no detrimental changes in the levels of PP2B, BDNF, and p- CaMKII. We conclude that treadmill exercise maybe able to prevent cognitive impairment associated with AD pathology.
Collapse
Affiliation(s)
- An T Dao
- Department of PPS, College of Pharmacy, University of Houston, Houston, TX 77204-5037, USA
| | - Munder A Zagaar
- Texas Southern University Department of Pharmacy Practice and Clinical Health Sciences Houston, TX 77004
| | | | | | | | | |
Collapse
|
42
|
Attar A, Liu T, Chan WTC, Hayes J, Nejad M, Lei K, Bitan G. A shortened Barnes maze protocol reveals memory deficits at 4-months of age in the triple-transgenic mouse model of Alzheimer's disease. PLoS One 2013; 8:e80355. [PMID: 24236177 PMCID: PMC3827415 DOI: 10.1371/journal.pone.0080355] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/08/2013] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disease that manifests as memory loss, cognitive dysfunction, and dementia. Animal models of Alzheimer's disease have been instrumental in understanding the underlying pathological mechanism and in evaluation of potential therapies. The triple transgenic (3 × Tg) mouse model of AD is unique because it recapitulates both pathologic hallmarks of Alzheimer's disease--amyloid plaques and neurofibrillary tangles. The earliest cognitive deficits in this model have been shown at 6-m of age by most groups, necessitating aging of the mice to this age before initiating evaluation of the cognitive effects of therapies. To assess cognitive deficits in the 3 × Tg mice, originally we employed a typical Barnes maze protocol of 15 training trials, but found no significant deficits in aged mice. Therefore, we shortened the protocol to include only 5 training trials to increase difficulty. We found cognitive deficits using this protocol using mainly measures from the probe day, rather than the training trials. This also decreased the effort involved with data analysis. We compared 3 × Tg and wild-type mice at 4-m- and 15-m of age using both the original, long training, and the short training paradigms. We found that differences in learning between 3 × Tg and wild-type mice disappeared after the 4(th) training trial. Measures of learning and memory on the probe day showed significant differences between 3 × Tg and wild-type mice following the short, 5-training trial protocol but not the long, 15-training trial protocol. Importantly, we detected cognitive dysfunction already at 4-m of age in 3 × Tg mice using the short Barnes-maze protocol. The ability to test learning and memory in 4-m old 3 × Tg mice using a shortened Barnes maze protocol offers considerable time and cost savings and provides support for the utilization of this model at pre-pathology stages for therapeutic studies.
Collapse
Affiliation(s)
- Aida Attar
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tingyu Liu
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Wai-Ting Coco Chan
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jane Hayes
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Mona Nejad
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - KaiChyuan Lei
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Gal Bitan
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
43
|
Lok K, Zhao H, Shen H, Wang Z, Gao X, Zhao W, Yin M. Characterization of the APP/PS1 mouse model of Alzheimer's disease in senescence accelerated background. Neurosci Lett 2013; 557 Pt B:84-9. [PMID: 24176881 DOI: 10.1016/j.neulet.2013.10.051] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 10/10/2013] [Accepted: 10/18/2013] [Indexed: 11/26/2022]
Abstract
The APP/PS1 mouse model of Alzheimer's disease (AD) exhibits remarkable elevation of β-amyloid production associated with certain behavioral abnormalities, while the senescence accelerated mouse prone 8 (SAMP8) is characterized by early and age-related deterioration of learning and memory. In order to obtain an AD model that develops earlier pathological changes and cognitive impairment, we generated SAMP8-APP/PS1, a novel senescence accelerated APP/PS1 transgenic mouse model of AD. Standard histological staining and immunohistochemistry using an amyloid beta (Aβ) antibody showed an age, genotype and strain-dependent progression of amyloid deposition and neuron loss in the cerebral cortex and hippocampus of SAMP8-APP/PS1 mice. Results from the cognitive behavioral tests revealed early deficits in learning and memory in SAMP8-APP/PS1 mice in the two way active avoidance and Morris water maze tests compared with C57-APP/PS1, SAMP8 wild-type and control mice. These results suggest that accelerated senescence exacerbates amyloid deposition and cognitive dysfunction in APP/PS1 mice and the senescence accelerated-APP/PS1 (SAMP8-APP/PS1) mouse model might be a valuable tool to study AD progression and to evaluate the effect of drugs on AD.
Collapse
Affiliation(s)
- Kenghoe Lok
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Hadass O, Tomlinson BN, Gooyit M, Chen S, Purdy JJ, Walker JM, Zhang C, Giritharan AB, Purnell W, Robinson CR, Shin D, Schroeder VA, Suckow MA, Simonyi A, Sun GY, Mobashery S, Cui J, Chang M, Gu Z. Selective inhibition of matrix metalloproteinase-9 attenuates secondary damage resulting from severe traumatic brain injury. PLoS One 2013; 8:e76904. [PMID: 24194849 PMCID: PMC3806745 DOI: 10.1371/journal.pone.0076904] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 08/29/2013] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and long-term disability. Following the initial insult, severe TBI progresses to a secondary injury phase associated with biochemical and cellular changes. The secondary injury is thought to be responsible for the development of many of the neurological deficits observed after TBI and also provides a window of opportunity for therapeutic intervention. Matrix metalloproteinase-9 (MMP-9 or gelatinase B) expression is elevated in neurological diseases and its activation is an important factor in detrimental outcomes including excitotoxicity, mitochondrial dysfunction and apoptosis, and increases in inflammatory responses and astrogliosis. In this study, we used an experimental mouse model of TBI to examine the role of MMP-9 and the therapeutic potential of SB-3CT, a mechanism-based gelatinase selective inhibitor, in ameliorating the secondary injury. We observed that activation of MMP-9 occurred within one day following TBI, and remained elevated for 7 days after the initial insult. SB-3CT effectively attenuated MMP-9 activity, reduced brain lesion volumes and prevented neuronal loss and dendritic degeneration. Pharmacokinetic studies revealed that SB-3CT and its active metabolite, p-OH SB-3CT, were rapidly absorbed and distributed to the brain. Moreover, SB-3CT treatment mitigated microglial activation and astrogliosis after TBI. Importantly, SB-3CT treatment improved long-term neurobehavioral outcomes, including sensorimotor function, and hippocampus-associated spatial learning and memory. These results demonstrate that MMP-9 is a key target for therapy to attenuate secondary injury cascades and that this class of mechanism-based gelatinase inhibitor–with such desirable pharmacokinetic properties–holds considerable promise as a potential pharmacological treatment of TBI.
Collapse
Affiliation(s)
- Orr Hadass
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri, United States of America ; Center for Translational Neuroscience, University of Missouri School of Medicine, Columbia, Missouri, United States of America ; MS in Pathology Program, University of Missouri Graduate School, Columbia, Missouri, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cognitive dysfunction precedes the onset of motor symptoms in the MitoPark mouse model of Parkinson's disease. PLoS One 2013; 8:e71341. [PMID: 23977020 PMCID: PMC3744561 DOI: 10.1371/journal.pone.0071341] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/29/2013] [Indexed: 11/19/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder primarily characterized by progressive loss of dopamine neurons, leading to loss of motor coordination. However, PD is associated with a high rate of non-motor neuropsychiatric comorbities that often develop before the onset of movement symptoms. The MitoPark transgenic mouse model is the first to recapitulate the cardinal clinical features, namely progressive neurodegeneration and death of neurons, loss of motor function and therapeutic response to L-DOPA. To investigate whether MitoPark mice exhibit early onset of cognitive impairment, a non-motor neuropsychiatric comorbidity, we measured performance on a spatial learning and memory task before (∼8 weeks) or after (∼20 weeks) the onset of locomotor decline in MitoPark mice or in littermate controls. Consistent with previous studies, we established that a progressive loss of spontaneous locomotor activity began at 12 weeks of age, which was followed by progressive loss of body weight beginning at 16–20 weeks. Spatial learning and memory was measured using the Barnes Maze. By 20 weeks of age, MitoPark mice displayed a substantial reduction in overall locomotor activity that impaired their ability to perform the task. However, in the 8-week-old mice, locomotor activity was no different between genotypes, yet MitoPark mice took longer, traveled further and committed more errors than same age control mice, while learning to successfully navigate the maze. The modest between-day learning deficit of MitoPark mice was characterized by impaired within-day learning during the first two days of testing. No difference was observed between genotypes during probe trials conducted one or twelve days after the final acquisition test. Additionally, 8-week-old MitoPark mice exhibited impaired novel object recognition when compared to control mice. Together, these data establish that mild cognitive impairment precedes the loss of motor function in a novel rodent model of PD, which may provide unique opportunities for therapeutic development.
Collapse
|
46
|
Nagy PM, Aubert I. B6eGFPChAT mice overexpressing the vesicular acetylcholine transporter exhibit spontaneous hypoactivity and enhanced exploration in novel environments. Brain Behav 2013; 3:367-83. [PMID: 24381809 PMCID: PMC3869679 DOI: 10.1002/brb3.139] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/18/2013] [Accepted: 03/22/2013] [Indexed: 12/15/2022] Open
Abstract
Cholinergic innervation is extensive throughout the central and peripheral nervous systems. Among its many roles, the neurotransmitter acetylcholine (ACh) contributes to the regulation of motor function, locomotion, and exploration. Cholinergic deficits and replacement strategies have been investigated in neurodegenerative disorders, particularly in cases of Alzheimer's disease (AD). Focus has been on blocking acetylcholinesterase (AChE) and enhancing ACh synthesis to improve cholinergic neurotransmission. As a first step in evaluating the physiological effects of enhanced cholinergic function through the upregulation of the vesicular acetylcholine transporter (VAChT), we used the hypercholinergic B6eGFPChAT congenic mouse model that has been shown to contain multiple VAChT gene copies. Analysis of biochemical and behavioral paradigms suggest that modest increases in VAChT expression can have a significant effect on spontaneous locomotion, reaction to novel stimuli, and the adaptation to novel environments. These observations support the potential of VAChT as a therapeutic target to enhance cholinergic tone, thereby decreasing spontaneous hyperactivity and increasing exploration in novel environments.
Collapse
Affiliation(s)
- Paul M Nagy
- Brain Sciences, Biological Sciences, Sunnybrook Research Institute2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Isabelle Aubert
- Brain Sciences, Biological Sciences, Sunnybrook Research Institute2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
47
|
Lithfous S, Dufour A, Després O. Spatial navigation in normal aging and the prodromal stage of Alzheimer's disease: insights from imaging and behavioral studies. Ageing Res Rev 2013; 12:201-13. [PMID: 22771718 DOI: 10.1016/j.arr.2012.04.007] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/20/2012] [Accepted: 04/30/2012] [Indexed: 11/28/2022]
Abstract
Normal aging and mild Alzheimer's disease (AD) are associated with declines in navigational skills, including allocentric and egocentric representations, cognitive mapping, landmark processing, and spatial memory. These changes, however, are associated with different patterns of structural and functional alterations in the neural network of navigation. In AD, these changes occur in the hippocampus, parahippocampal gyrus, parietal lobe, retrosplenial cortex, prefrontal cortex, and caudate nucleus, whereas in aging, modifications occur mainly in the prefrontal cortex and the hippocampus. The navigation abilities of patients with mild cognitive impairment (MCI) have been found to show different performance patterns, depending on their cognitive profiles. Since patients with MCI do not uniformly develop dementia of the Alzheimer type, it is important to identify reliable early cognitive markers of conversion to AD dementia. In this review, we propose that navigation deficits may help distinguish patients at higher risk of developing AD dementia from individuals with normal cognitive aging and those with other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ségolène Lithfous
- Laboratoire d'Imagerie & Neurosciences Cognitives, UMR CNRS - Université de Strasbourg, France.
| | | | | |
Collapse
|
48
|
Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, Gelpi E, Halle A, Korte M, Latz E, Golenbock DT. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature 2012; 493:674-8. [PMID: 23254930 PMCID: PMC3812809 DOI: 10.1038/nature11729] [Citation(s) in RCA: 1943] [Impact Index Per Article: 161.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 10/30/2012] [Indexed: 01/22/2023]
Abstract
Alzheimer´s Disease (AD) is the world’s most common dementing illness. Deposition of amyloid beta peptide (Aβ) drives cerebral neuroinflammation by activating microglia1,2. Indeed, Aβ activation of the NLRP3 inflammasome in microglia is fundamental for IL-1β maturation and subsequent inflammatory events3. However, it remains unknown whether NLRP3 activation contributes to AD in vivo. Here, we demonstrate strongly enhanced active caspase-1 expression in human MCI and AD brains suggesting a role for the inflammasome in this neurodegenerative disease. NLRP3−/− or caspase-1−/− mice carrying mutations associated with familiar AD were largely protected from loss of spatial memory and other AD-associated sequelae and demonstrated reduced brain caspase-1 and IL-1β activation as well as enhanced Aβ clearance. Furthermore, NLRP3 inflammasome deficiency skewed microglial cells to an M2 phenotype and resulted in the decreased deposition of Aβ in the APP/PS1 model of Alzheimer’s disease. These results reveal an important role for the NLRP3 / caspase-1 axis in AD pathogenesis, and suggest that NLRP3 inflammasome inhibition represents a novel therapeutic intervention for AD.
Collapse
Affiliation(s)
- Michael T Heneka
- Clinical Neuroscience Unit, Department of Neurology, University of Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Fowler SW, Walker JM, Klakotskaia D, Will MJ, Serfozo P, Simonyi A, Schachtman TR. Effects of a metabotropic glutamate receptor 5 positive allosteric modulator, CDPPB, on spatial learning task performance in rodents. Neurobiol Learn Mem 2012; 99:25-31. [PMID: 23137441 DOI: 10.1016/j.nlm.2012.10.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 10/10/2012] [Accepted: 10/23/2012] [Indexed: 11/28/2022]
Abstract
Metabotropic glutamate receptor 5 (mGlu5) has been implicated in a variety of learning and memory processes and is important for avoidance learning. The present studies used an mGlu5 receptor positive allosteric modulator, 3-cyano-N-(1,3 diphenyl-1H-hyrazol-5-yl)benzamide (CDPPB), to characterize the importance of mGlu5 receptors in aversively- and appetitively-motivated spatial learning tasks (tasks in which the instrumental contingency involves discriminative cues that differ in spatial location). C57Bl/6 male mice were initially trained in the Barnes maze in the absence of drug. Subsequently, CDPPB (30mg/kg, i.p.), administered 20min prior to each of 3 daily reversal learning training sessions in the Barnes maze, significantly enhanced performance compared to vehicle-treated controls and had a significant effect on search strategy. Mice treated with CDPPB also displayed significantly less perseverative behavior than control-treated animals. In a second experiment, male Sprague-Dawley rats were trained in an appetitively-motivated, delayed alternation version of a T-maze. 30mg/kg CDPPB (s.c.), delivered 20min prior to each of 5 daily training sessions, enhanced the delay rats were able to withstand between the sample and choice portions of each T-maze trial. The present results emphasize the role of mGlu5 receptors in spatial learning tasks and support previous studies which report mGlu5 positive allosteric modulators can enhance learning in some tasks and may have potential as nootropic drugs.
Collapse
Affiliation(s)
- S W Fowler
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, United States
| | | | | | | | | | | | | |
Collapse
|
50
|
Behavioral effects of Rho GTPase modulation in a model of Alzheimer's disease. Behav Brain Res 2012; 237:223-9. [PMID: 23026376 DOI: 10.1016/j.bbr.2012.09.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 09/18/2012] [Accepted: 09/23/2012] [Indexed: 12/19/2022]
Abstract
Small GTPases of the Rho family, including Rho, Rac and CDC42 subfamilies, play key role in neural connectivity and cognition. The pharmacological modulation of these regulatory proteins is associated with enhancement of learning and memory. We sought to determine whether the modulation of cerebral Rho GTPases may correct behavioral disturbances in a mouse model of Alzheimer's disease (AD). TgCRND8 mice show early-onset Abeta amyloid deposits associated with deficits in several cognitive tasks. We report that four-month old TgCRND8 mice display (a) increased locomotor activity in an open field, (b) mild deficits in the learning of a fixed platform position in a water maze task. More markedly, after displacement of the escape platform, TgCRND8 mice exhibit impairment in the learning of the novel position (reversal learning), as they perseverate searching in the familiar position. The administration of the Rho GTPase activator Cytotoxic Necrotizing Factor 1 (CNF1, 1.0 fmol kg(-1) intracerebroventricularly) reduces locomotor hyperactivity and corrects the deficits in reversal learning, thus re-establishing normal behavioral plasticity. We conclude that the pharmacological modulation of Rho GTPase signaling might be beneficial for the treatment of AD. Reversal learning in TgCRND8 mice may represent a convenient pre-clinical assay for the efficacy of therapeutic interventions in AD.
Collapse
|